1
|
So CY, Li Y, Chow KT. New insights on Galectin-9 expression in cancer prognosis: An updated systemic review and meta-analysis. PLoS One 2025; 20:e0320441. [PMID: 40138336 PMCID: PMC11940609 DOI: 10.1371/journal.pone.0320441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Accepted: 02/18/2025] [Indexed: 03/29/2025] Open
Abstract
Galectin-9 (Gal-9) has gained increasing attention in recent years in the field of cancer immunology. Its interactions with various immune cell types in the tumor microenvironment influence tumor progression, making it a novel target for immunotherapy. Despite its potential as a therapeutic target, the prognostic significance of Gal-9 in tumor cells remains unclear. Conflicting data exists on its expression levels and outcomes, prompting a comprehensive review and meta-analysis to elucidate its independent prognostic role across different cancer types. This study aims to examine the varying effects of Gal-9 expression across various cancer subtypes, providing insights into its potential as a prognostic marker and highlighting its significance in the realm of cancer treatment. To assess the prognostic significance of Gal-9 expression in cancer, we conducted a comprehensive database search across PubMed, Embase, and Web of Science, incorporating studies published until December 2024, regardless of language. Using pooled hazard ratios (HRs) with 95% confidence intervals (CIs), we evaluated the role of Gal-9 expression in predicting cancer outcomes across various cancer types. Our analysis encompassed 29 studies with a total of 4,720 patients to investigate the prognostic significance of Gal-9 expression across different cancer types. The results demonstrated that elevated Gal-9 expression was significantly associated with improved overall survival (OS) in solid tumors, with a pooled hazard ratio of 0.75 (95% CI: 0.63-0.90, p = 0.002). No statistically significant correlation was observed between Gal-9 expression and cancer recurrence (HR = 0.88, 95% CI: 0.65-1.19, p = 0.42). Conversely, in hematological cancers, high Gal-9 expression correlated with more rapid disease progression, as reflected by progression-free survival (PFS) or time to treatment (TTT) (HR = 2.29, 95% CI: 1.26-4.16, p = 0.007). The subgroup analyses further revealed that higher Gal-9 expression was associated with OS in gastrointestinal and urological cancers and was linked to disease-free survival (DFS) and recurrence-free survival (RFS) in hepatobiliary and urological cancers. Our research has uncovered that Gal-9 serves as a promising prognostic indicator for solid tumors, offering valuable insights into patient outcomes. High levels of Gal-9 expression within gastrointestinal, hepatobiliary, and urological cancers have been linked to better prognoses, while its presence in hematological cancers is associated with poorer outcomes. These contrasting findings emphasize the importance of interpreting biomarkers with careful consideration to the specific context. Moreover, our study sheds light on the diverse physiological roles of intracellular and secreted Gal-9, highlighting the intricate ways in which this protein influences cancer progression.
Collapse
Affiliation(s)
- Chun Yan So
- Department of Biomedical Sciences, City University of Hong Kong, Kowloon, Hong Kong SAR
| | - Yusong Li
- Department of Biomedical Sciences, City University of Hong Kong, Kowloon, Hong Kong SAR
| | - Kwan Ting Chow
- Department of Biomedical Sciences, City University of Hong Kong, Kowloon, Hong Kong SAR
| |
Collapse
|
2
|
Du W, Xia X, Gou Q, Xie Y, Gao L. Comprehensive review regarding the association of E2Fs with the prognosis and immune infiltrates in human head and neck squamous cell carcinoma. Asian J Surg 2024; 47:2106-2121. [PMID: 38320907 DOI: 10.1016/j.asjsur.2024.01.130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 12/14/2023] [Accepted: 01/19/2024] [Indexed: 02/08/2024] Open
Abstract
E2F transcription factors (E2Fs) are a group of genes that encode a family of transcription factors. They have been identified as being involved in the tumor progression of various cancer types. However, little is known about the expression level, genetic variation, molecular mechanism, and prognostic value and immune infiltration of different E2Fs in HNSCC.In this study, we utilized multiple databases to investigate the mRNA expression level, genetic alteration, and biological function of E2Fs in HNSCC patients. Then, the relationship between E2Fs expression and its association with the occurrence, progress, prognosis, and immune cell infiltration in patients with HNSCC was evaluated. We found that all eight E2Fs were higher expressed in HNSCC tissues than in normal tissues, and the expression levels of E2F1/2/3/4/5/6/8 were also associated with the stage and grade of HNSCC. The abnormal expression of E2F1/2/4/8 in HNSCC patients is related to the clinical outcome. The expression of E2Fs was statistically correlated with the immune cell infiltration in HNSCC and the infiltration of B cells and CD8+ T cells were positively associated with better OS in HNSCC patients. Furthermore, we verified the E2F2 at the tissue level in the validation experiment. Our study may provide novel insights into the choice of immunotherapy targets and potential prognostic biomarkers in HNSCC patients.
Collapse
Affiliation(s)
- Wei Du
- Department of Targetting Therapy & Immunology, Cancer Cencer, West China Hospital, Sichuan University, Chengdu, China
| | - Xueming Xia
- Division of Head & Neck Tumor Multimodaligy Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Qiheng Gou
- Division of Head & Neck Tumor Multimodaligy Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yuxin Xie
- Division of Head & Neck Tumor Multimodaligy Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Lanyang Gao
- Academician (Expert) Workstation of Sichuan Province, Metabolic Hepatobiliary and Pancreatic Diseases Key Laboratory of Luzhou City, The Affiliated Hospital of Southwest Medical University, Sichuan, China.
| |
Collapse
|
3
|
Basingab FS, Alzahrani RA, Alrofaidi AA, Barefah AS, Hammad RM, Alahdal HM, Alrahimi JS, Zaher KA, Algiraigri AH, El-Daly MM, Alkarim SA, Aldahlawi AM. Herpesvirus Entry Mediator as an Immune Checkpoint Target and a Potential Prognostic Biomarker in Myeloid and Lymphoid Leukemia. Biomolecules 2024; 14:523. [PMID: 38785930 PMCID: PMC11117912 DOI: 10.3390/biom14050523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 04/18/2024] [Accepted: 04/23/2024] [Indexed: 05/25/2024] Open
Abstract
Herpesvirus entry mediator (HVEM) is a molecular switch that can modulate immune responses against cancer. The significance of HVEM as an immune checkpoint target and a potential prognostic biomarker in malignancies is still controversial. This study aims to determine whether HVEM is an immune checkpoint target with inhibitory effects on anti-tumor CD4+ T cell responses in vitro and whether HVEM gene expression is dysregulated in patients with acute lymphocytic leukemia (ALL). HVEM gene expression in tumor cell lines and peripheral blood mononuclear cells (PBMCs) from ALL patients and healthy controls was measured using reverse transcription-quantitative polymerase chain reaction (RT-qPCR). Tumor cells were left untreated (control) or were treated with an HVEM blocker before co-culturing with CD4+ T cells in vitro in a carboxyfluorescein succinimidyl ester (CFSE)-dependent proliferation assay. HVEM expression was upregulated in the chronic myelogenous leukemia cell line (K562) (FC = 376.3, p = 0.086) compared with normal embryonic kidney cells (Hek293). CD4+ T cell proliferation was significantly increased in the HVEM blocker-treated K562 cells (p = 0.0033). Significant HVEM differences were detected in ALL PBMCs compared with the controls, and these were associated with newly diagnosed ALL (p = 0.0011) and relapsed/refractory (p = 0.0051) B cell ALL (p = 0.0039) patients. A significant differentiation between malignant ALL and the controls was observed in a receiver operating characteristic (ROC) curve analysis with AUC = 0.78 ± 0.092 (p = 0.014). These results indicate that HVEM is an inhibitory molecule that may serve as a target for immunotherapy and a potential ALL biomarker.
Collapse
Affiliation(s)
- Fatemah S. Basingab
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah 21859, Saudi Arabia
- Immunology Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21859, Saudi Arabia
| | - Reem A. Alzahrani
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah 21859, Saudi Arabia
- Immunology Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21859, Saudi Arabia
| | - Aisha A. Alrofaidi
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah 21859, Saudi Arabia
- Immunology Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21859, Saudi Arabia
| | - Ahmed S. Barefah
- Hematology Department, Faculty of Medicine, King Abdulaziz University Hospital, King Abdulaziz University, Jeddah 21859, Saudi Arabia
- Hematology Research Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21859, Saudi Arabia
| | - Rawan M. Hammad
- Hematology Department, Faculty of Medicine, King Abdulaziz University Hospital, King Abdulaziz University, Jeddah 21859, Saudi Arabia
| | - Hadil M. Alahdal
- Department of Biology, Faculty of Science, Princes Nourah bint Abdulrahman University, Riyadh 11671, Saudi Arabia
| | - Jehan S. Alrahimi
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah 21859, Saudi Arabia
- Immunology Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21859, Saudi Arabia
| | - Kawther A. Zaher
- Immunology Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21859, Saudi Arabia
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21859, Saudi Arabia
| | - Ali H. Algiraigri
- Hematology Department, Faculty of Medicine, King Abdulaziz University Hospital, King Abdulaziz University, Jeddah 21859, Saudi Arabia
- Hematology Research Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21859, Saudi Arabia
| | - Mai M. El-Daly
- Special Infectious Agents Unit-BSL3, King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21859, Saudi Arabia
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21859, Saudi Arabia
| | - Saleh A. Alkarim
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah 21859, Saudi Arabia
- Embryonic Stem Cells Research Unit and Embryonic and Cancer Stem Cells Research Group, King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21859, Saudi Arabia
| | - Alia M. Aldahlawi
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah 21859, Saudi Arabia
- Immunology Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21859, Saudi Arabia
| |
Collapse
|
4
|
Siddiqui SH, Kumari N, Mishra S, Radha P, Mohindra S, Singh RK, Krishnani N. PD-L1 Expression in Ampullary Adenocarcinoma. Int J Surg Pathol 2024; 32:263-272. [PMID: 37291997 DOI: 10.1177/10668969231177263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
INTRODUCTION Ampullary adenocarcinoma is a rare neoplasm often treated by the complex Whipple's procedure. Several histological factors predict poor prognosis including pancreatobiliary morphology, presence of lymphovascular, perineural invasion and local or distant metastasis. Systemic therapy with gemcitabine, 5-fluorouracil regimens are given with variable benefits. Immunotherapy checkpoint inhibitors have shown beneficial anti-tumor effects in several carcinomas, the most remarkable being in non-small cell lung cancer. Administration of these novel drugs is based on immunohistochemical expression (which may or may not be indicative of response to therapy) along with meticulous decision making by the multidisciplinary team. Immunohistochemistry (IHC) is an effective means of immune marker demonstration and has been used in various tumor types for predictive and prognostic purposes. METHODS PD-L1 IHC (clone E1L3N) was applied in 101 cases of ampullary adenocarcinoma. Tumor infiltrating lymphocytes were also evaluated. The immunoreactivity was assessed and categorized into following staining thresholds: <1%, <5%, <10% and ≥10% for tumor cells (membranous and/or cytoplasmic staining pattern), and 5% and 10% cut-offs for immune cells. RESULTS We found that at a 10% cut-off, 73.3% (74/101) patients were men (P = .006) older than 50 years of age (P < .001) presenting with a tumor measuring <3 cm (P = .001). It was significantly associated with intestinal differentiation (P = .004) and grade 1 tumors (P = .001). Twelve patients presented with recurrence as well (P = .03). CONCLUSION In the context of ampullary adenocarcinoma, this study highlights the positivity observed with the PD-L1 IHC clone E1L3N at different thresholds, with the particularly stronger associations being evident at a 10% cut-off.
Collapse
Affiliation(s)
- Saima Haleem Siddiqui
- Department of Pathology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, India
| | - Niraj Kumari
- Department of Pathology and Lab Medicine, All India Institute of Medical Sciences, Raebareli, Uttar Pradesh, India
| | - Shravan Mishra
- Department of Pathology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, India
| | - Paturu Radha
- Department of Pathology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, India
| | - Samir Mohindra
- Department of Gastroenterology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, India
| | - Rajneesh K Singh
- Department of Surgical Gastroenterology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, India
| | - Narendra Krishnani
- Department of Pathology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, India
| |
Collapse
|
5
|
Wang ZT, Deng ZM, Dai FF, Yuan MQ, Liu SY, Li BS, Cheng YX. Tumor immunity: A brief overview of tumor‑infiltrating immune cells and research advances into tumor‑infiltrating lymphocytes in gynecological malignancies (Review). Exp Ther Med 2024; 27:166. [PMID: 38476909 PMCID: PMC10928974 DOI: 10.3892/etm.2024.12453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 01/03/2023] [Indexed: 03/14/2024] Open
Abstract
Tumor immunity is a promising topic in the area of cancer therapy. The 'soil' function of the tumor microenvironment (TME) for tumor growth has attracted wide attention from scientists. Tumor-infiltrating immune cells in the TME, especially the tumor-infiltrating lymphocytes (TILs), serve a key role in cancer. Firstly, relevant literature was searched in the PubMed and Web of Science databases with the following key words: 'Tumor microenvironment'; 'TME'; 'tumor-infiltrating immunity cells'; 'gynecologic malignancies'; 'the adoptive cell therapy (ACT) of TILs'; and 'TIL-ACT' (https://pubmed.ncbi.nlm.nih.gov/). According to the title and abstract of the articles, relevant items were screened out in the preliminary screening. The most relevant selected items were of two types: All kinds of tumor-infiltrating immune cells; and advanced research on TILs in gynecological malignancies. The results showed that the subsets of TILs were various and complex, while each subpopulation influenced each other and their effects on tumor prognosis were diverse. Moreover, the related research and clinical trials on TILs were mostly concentrated in melanoma and breast cancer, but relatively few focused on gynecological tumors. In conclusion, the present review summarized the biological classification of TILs and the mechanisms of their involvement in the regulation of the immune microenvironment, and subsequently analyzed the development of tumor immunotherapy for TILs. Collectively, the present review provides ideas for the current treatment dilemma of gynecological tumor immune checkpoints, such as adverse reactions, safety, personal specificity and efficacy.
Collapse
Affiliation(s)
- Zi-Tao Wang
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Zhi-Min Deng
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Fang-Fang Dai
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Meng-Qin Yuan
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Shi-Yi Liu
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Bing-Shu Li
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Yan-Xiang Cheng
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| |
Collapse
|
6
|
Wojciechowicz K, Spodzieja M, Wardowska A. The BTLA-HVEM complex - The future of cancer immunotherapy. Eur J Med Chem 2024; 268:116231. [PMID: 38387336 DOI: 10.1016/j.ejmech.2024.116231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 02/09/2024] [Accepted: 02/10/2024] [Indexed: 02/24/2024]
Abstract
The BTLA-HVEM complex plays a pivotal role in cancer and cancer immunotherapy by regulating immune responses. Dysregulation of BTLA and HVEM expression contributes to immunosuppression and tumor progression across various cancer types. Targeting the interaction between BTLA and HVEM holds promise for enhancing anti-tumor immune responses. Disruption of this complex presents a valuable avenue for advancing cancer immunotherapy strategies. Aberrant expression of BTLA and HVEM adversely affects immune cell function, particularly T cells, exacerbating tumor evasion mechanisms. Understanding and modulating the BTLA-HVEM axis represents a crucial aspect of designing effective immunotherapeutic interventions against cancer. Here, we summarize the current knowledge regarding the structure and function of BTLA and HVEM, along with their interaction with each other and various immune partners. Moreover, the expression of soluble and transmembrane forms of BTLA and HVEM in different types of cancer and their impact on the prognosis of patients is also discussed. Additionally, inhibitors of the proteins binding that might be used to block BTLA-HVEM interaction are reviewed. All the presented data highlight the plausible clinical application of BTLA-HVEM targeted therapies in cancer and autoimmune disease management. However, further studies are required to confirm the practical use of this concept. Despite the increasing number of reports on the BTLA-HVEM complex, many aspects of its biology and function still need to be elucidated. This review can be regarded as an encouragement and a guide to follow the path of BTLA-HVEM research.
Collapse
Affiliation(s)
- Karolina Wojciechowicz
- Department of Physiopathology, Faculty of Medicine, Medical University of Gdansk, Poland.
| | - Marta Spodzieja
- Department of Biomedical Chemistry, Faculty of Chemistry, University of Gdansk, Poland
| | - Anna Wardowska
- Department of Physiopathology, Faculty of Medicine, Medical University of Gdansk, Poland.
| |
Collapse
|
7
|
Aldahlawi A, Basingab F, Alrahimi J, Zaher K, Pushparaj PN, Hassan MA, Al-Sakkaf K. Herpesvirus entry mediator as a potential biomarker in breast cancer compared with conventional cytotoxic T‑lymphocyte‑associated antigen 4. Biomed Rep 2023; 19:56. [PMID: 37560313 PMCID: PMC10407466 DOI: 10.3892/br.2023.1638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 03/17/2023] [Indexed: 08/11/2023] Open
Abstract
Breast cancer (BC) is the most common cancer in women worldwide, with 2.3 million cases recorded in 2020. Despite improvements in cancer treatment, patients with BC still succumb to the disease, due to regional and distant metastases when diagnosed at later stages. Several immune checkpoint inhibitors have been approved for BC treatment, based on their expression and role in maintaining immunosurveillance against tumors. The present study aimed to evaluate the expression of 12 immune checkpoints in patients with BC, and assess their role as diagnostic and therapeutic markers. Expression levels were measured using reverse transcription-quantitative polymerase chain reaction. Among the 12 immune markers, herpesvirus entry mediator (HVEM) was found to be significantly upregulated in patients with malignant BC compared to non-malignant controls, with a relative fold change (FC) of 1.46 and P=0.012. A similar finding was observed for cytotoxic T-lymphocyte-associated antigen 4 (CTLA4; FC=1.47 and P=0.035). In addition, receiver operating characteristic curve analysis revealed that HVEM expression allowed significant differentiation between groups, with an area under the curve of 0.74 (P=0.013). Upregulation in both HVEM and CTLA4 was revealed to be significantly associated with the human epidermal growth factor receptor-2 (HER2)-enriched phenotype (FC=3.53, P=0.009 and FC=5.98, P=0.002, respectively), while only HVEM was significantly associated with the triple-negative phenotype (FC=2.07, P=0.016). Furthermore, HVEM was significantly higher in patients with grade III tumors (FC=1.88, P=0.025) and negative vascular invasion (FC=1.67, P=0.046) compared with non-malignant controls. Serum protein levels were assessed by multiplex immunoassay, and a significant increase in HVEM was detected in patients with malignant BC compared with that in non-malignant controls (P=0.035). These data indicated that HVEM may serve as a potential biomarker and target for immunotherapy, especially for certain types of BC.
Collapse
Affiliation(s)
- Alia Aldahlawi
- Department of Biological Sciences, Faculty of Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Immunology Unit, King Fahad for Medical Research, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Fatemah Basingab
- Department of Biological Sciences, Faculty of Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Immunology Unit, King Fahad for Medical Research, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Jehan Alrahimi
- Department of Biological Sciences, Faculty of Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Immunology Unit, King Fahad for Medical Research, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Kawther Zaher
- Immunology Unit, King Fahad for Medical Research, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Peter Natesan Pushparaj
- Center of Excellence in Genomic Medicine Research, King Fahad for Medical Research, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Center for Transdisciplinary Research, Department of Pharmacology, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Chennai, India
| | - Mohammed A. Hassan
- Department of Medical Basic Sciences, College of Medicine and Health Sciences, Hadhramout University, Mukalla 50511, Republic of Yemen
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Kaltoom Al-Sakkaf
- Immunology Unit, King Fahad for Medical Research, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| |
Collapse
|
8
|
Bartolome J, Molto C, Benitez-Fuentes JD, Fernandez-Hinojal G, Manzano A, Perez-Segura P, Mittal A, Tamimi F, Amir E, Ocana A. Prognostic value of human leukocyte antigen G expression in solid tumors: a systematic review and meta-analysis. Front Immunol 2023; 14:1165813. [PMID: 37275862 PMCID: PMC10232772 DOI: 10.3389/fimmu.2023.1165813] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 05/02/2023] [Indexed: 06/07/2023] Open
Abstract
Introduction Identification of modulators of the immune response with inhibitory properties that could be susceptible for therapeutic intervention is a key goal in cancer research. An example is the human leukocyte antigen G (HLA-G), a nonclassical major histocompatibility complex (MHC) class I molecule, involved in cancer progression. Methods In this article we performed a systematic review and meta-analysis on the association between HLA-G expression and outcome in solid tumors. This study was performed in accordance with PRISMA guidelines and registered in PROSPERO. Results A total of 25 studies met the inclusion criteria. These studies comprised data from 4871 patients reporting overall survival (OS), and 961 patients, reporting disease free survival (DFS). HLA-G expression was associated with worse OS (HR 2.09, 95% CI = 1.67 to 2.63; P < .001), that was higher in gastric (HR = 3.40; 95% CI = 1.64 to 7.03), pancreatic (HR = 1.72; 95% CI = 0.79 to 3.74) and colorectal (HR = 1.55; 95% CI = 1.16 to 2.07) cancer. No significant differences were observed between the most commonly utilized antibody (4H84) and other methods of detection. HLA-G expression was associated with DFS which approached but did not meet statistical significance. Discussion In summary, we describe the first meta-analysis associating HLA-G expression and worse survival in a variety of solid tumors. Systematic Review Registration https://www.crd.york.ac.uk/PROSPERO/, identifier CRD42022311973.
Collapse
Affiliation(s)
- Jorge Bartolome
- Experimental Therapeutics Unit, Department of Medical Oncology, Hospital Clinico San Carlos and Health Research Institute of the Hospital Clinico San Carlos (IdISSC), Madrid, Spain
| | - Consolacion Molto
- Division of Medical Oncology and Hematology, Department of Medicine, Princess Margaret Cancer Centre and University of Toronto, Toronto, ON, Canada
| | | | | | - Aranzazu Manzano
- Experimental Therapeutics Unit, Department of Medical Oncology, Hospital Clinico San Carlos and Health Research Institute of the Hospital Clinico San Carlos (IdISSC), Madrid, Spain
| | - Pedro Perez-Segura
- Experimental Therapeutics Unit, Department of Medical Oncology, Hospital Clinico San Carlos and Health Research Institute of the Hospital Clinico San Carlos (IdISSC), Madrid, Spain
| | - Abhenil Mittal
- Division of Medical Oncology and Hematology, Department of Medicine, Princess Margaret Cancer Centre and University of Toronto, Toronto, ON, Canada
| | - Faris Tamimi
- Division of Medical Oncology and Hematology, Department of Medicine, Princess Margaret Cancer Centre and University of Toronto, Toronto, ON, Canada
| | - Eitan Amir
- Division of Medical Oncology and Hematology, Department of Medicine, Princess Margaret Cancer Centre and University of Toronto, Toronto, ON, Canada
| | - Alberto Ocana
- Experimental Therapeutics Unit, Department of Medical Oncology, Hospital Clinico San Carlos and Health Research Institute of the Hospital Clinico San Carlos (IdISSC), Madrid, Spain
| |
Collapse
|
9
|
Iqbal H, Petrosyan A, Yoon J, Roytman M. A Complex Diagnosis of Ampullary Adenocarcinoma Presenting As Decompensated Cirrhosis. Cureus 2023; 15:e37566. [PMID: 37193458 PMCID: PMC10183155 DOI: 10.7759/cureus.37566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/14/2023] [Indexed: 05/18/2023] Open
Abstract
Neoplasms arising from the ampulla of Vater are exceedingly rare, and there is a paucity of literature regarding their diagnosis and management. Ampullary cancer typically presents with jaundice and signs of biliary obstruction. We present a case of ampullary adenocarcinoma with concomitant choledocholithiasis that proved complex and diagnostically challenging.
Collapse
Affiliation(s)
- Humzah Iqbal
- Internal Medicine, University of California, San Francisco, Fresno, USA
| | - Arpine Petrosyan
- Internal Medicine, University of California, San Francisco, Fresno, USA
| | - Jennifer Yoon
- Gastroenterology and Hepatology, University of California, San Francisco, Fresno, USA
| | - Marina Roytman
- Gastroenterology and Hepatology, University of California, San Francisco, Fresno, USA
| |
Collapse
|
10
|
Morishita A, Oura K, Tadokoro T, Shi T, Fujita K, Tani J, Atsukawa M, Masaki T. Galectin-9 in Gastroenterological Cancer. Int J Mol Sci 2023; 24:ijms24076174. [PMID: 37047155 PMCID: PMC10094448 DOI: 10.3390/ijms24076174] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 03/07/2023] [Accepted: 03/20/2023] [Indexed: 03/29/2023] Open
Abstract
Immunochemotherapy has become popular in recent years. The detailed mechanisms of cancer immunity are being elucidated, and new developments are expected in the future. Apoptosis allows tissues to maintain their form, quantity, and function by eliminating excess or abnormal cells. When apoptosis is inhibited, the balance between cell division and death is disrupted and tissue homeostasis is impaired. This leads to dysfunction and the accumulation of genetically abnormal cells, which can contribute to carcinogenesis. Lectins are neither enzymes nor antibodies but proteins that bind sugar chains. Among soluble endogenous lectins, galectins interact with cell surface sugar chains outside the cell to regulate signal transduction and cell growth. On the other hand, intracellular lectins are present at the plasma membrane and regulate signal transduction by regulating receptor–ligand interactions. Galectin-9 expressed on the surface of thymocytes induces apoptosis of T lymphocytes and plays an essential role in immune self-tolerance by negative selection in the thymus. Furthermore, the administration of extracellular galectin-9 induces apoptosis of human cancer and immunodeficient cells. However, the detailed pharmacokinetics of galectin-9 in vivo have not been elucidated. In addition, the cell surface receptors involved in galectin-9-induced apoptosis of cancer cells have not been identified, and the intracellular pathways involved in apoptosis have not been fully investigated. We have previously reported that galectin-9 induces apoptosis in various gastrointestinal cancers and suppresses tumor growth. However, the mechanism of galectin-9 and apoptosis induction in gastrointestinal cancers and the detailed mechanisms involved in tumor growth inhibition remain unknown. In this article, we review the effects of galectin-9 on gastrointestinal cancers and its mechanisms.
Collapse
|
11
|
Zhang Z, Xiong Q, Xu Y, Cai X, Zhang L, Zhu Q. The PD-L1 Expression and Tumor-Infiltrating Immune Cells Predict an Unfavorable Prognosis in Pancreatic Ductal Adenocarcinoma and Adenosquamous Carcinoma. J Clin Med 2023; 12:jcm12041398. [PMID: 36835933 PMCID: PMC9965576 DOI: 10.3390/jcm12041398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 02/03/2023] [Accepted: 02/06/2023] [Indexed: 02/12/2023] Open
Abstract
The tumor microenvironment (TME) plays a vital role in the development, progression, and metastasis of pancreatic cancer (PC). The composition of the TME and its potential prognostic value remains to be fully understood, especially in adenosquamous carcinoma of pancreas (ASCP) patients. Immunohistochemistry was used to explore the clinical significance of CD3, CD4, CD8, FoxP3, and PD-L1 expression within the TME and to identify correlations with the prognosis of PC in a series of 29 patients with ASCP and 54 patients with pancreatic ductal adenocarcinoma (PDAC). Data from the Gene Expression Omnibus (GEO) and the Cancer Genome Atlas (TCGA) were accessed to obtain the scRNA-seq data and transcriptome profiles. Seurat was used to process the scRNA-seq data, and CellChat was used to analyze cell-cell communication. CIBERSORT was used to approximate the constitution of tumor-infiltrating immune cell (TICs) profiles. Higher levels of PD-L1 were linked with a shorter overall survival in ASCP (p = 0.0007) and PDAC (p = 0.0594). A higher expression of CD3+ and CD8+ T-cell infiltration was significantly correlated with a better prognosis in PC. By influencing the composition of tumor-infiltrating immune cells (TICs), high levels of PD-L1 expression are linked with a shorter overall survival in ASCP and PDAC.
Collapse
|
12
|
The Systematic Analyses of RING Finger Gene Signature for Predicting the Prognosis of Patients with Hepatocellular Carcinoma. JOURNAL OF ONCOLOGY 2022; 2022:2466006. [PMID: 36199791 PMCID: PMC9529411 DOI: 10.1155/2022/2466006] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 09/08/2022] [Indexed: 12/24/2022]
Abstract
RING finger (RNF) proteins are frequently dysregulated in human malignancies and are tightly associated with tumorigenesis. However, the expression profiles of RNF genes in hepatocellular carcinoma (HCC) and their relations with prognosis remain undetermined. Here, we aimed at constructing a prognostic model according to RNF genes for forecasting the outcomes of HCC patients using the data from The Cancer Genome Atlas (TCGA) program. We collected HCC datasets to validate the values of our model in predicting prognosis of HCC patients from International Cancer Genome Consortium (ICGC) platform. Then, functional experiments were carried out to explore the roles of the representative RNF in HCC progression. A total of 107 differentially expressed RNFs were obtained between TCGA-HCC tumor and normal tissues. After comprehensive evaluation, a prognostic signature composed of 11 RNFs (RNF220, RNF25, TRIM25, BMI1, RNF216P1, RNF115, RNF2, TRAIP, RNF157, RNF145, and RNF19B) was constructed based on TCGA cohort. Then, the Kaplan-Meier (KM) curves and the receiver operating characteristic curve (ROC) were employed to evaluate predictive power of the prognostic model in testing cohort (TCGA) and validation cohort (ICGC). The KM and ROC curves illustrated the good predictive power in testing and validation cohort. The areas under the ROC curve are 0.77 and 0.76 in these two cohorts, respectively. Among the prognostic signature genes, BMI1 was selected as a representative for functional study. We found that BMI1 protein level was significantly upregulated in HCC tissues. Moreover, the inhibitor of BMI1, PTC-209, displayed an excellent anti-HCC effect in vitro. Enrichment analysis of BMI1 downstream targets showed that BMI1 might be involved in tumor immunotherapy. Together, our overall analyses revealed that the 11-RNFs prognostic signature might provide us latent chances for evaluating HCC prognosis and developing novel HCC therapy.
Collapse
|
13
|
Zhao Y, Liu Y, Jia Y, Wang X, He J, Zhen S, Wang J, Liu L. Fibroblast activation protein in the tumor microenvironment predicts outcomes of PD-1 blockade therapy in advanced non-small cell lung cancer. J Cancer Res Clin Oncol 2022:10.1007/s00432-022-04250-4. [PMID: 35951090 DOI: 10.1007/s00432-022-04250-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Accepted: 08/02/2022] [Indexed: 10/15/2022]
Abstract
PURPOSE The identification of robust predictive biomarkers of the response to programmed cell death-1 (PD-1) blockade remains a critical concern. Here, we investigated on fibroblast activation protein (FAP) as a microenvironment-derived biomarker of clinical outcomes of PD-1 blockade therapy, and the correlation between FAP expression and T cell infiltration in advanced non-small cell lung cancer (NSCLC). METHODS A total of 135 patients with advanced NSCLC who received PD-1 blockade therapy were retrospectively analyzed. The potential associations among FAP expression, CD3 + T cell and CD8 + T cell infiltration, and clinical outcomes of immunotherapy were validated by immunohistochemistry, bioinformatic analyses, and statistical measurements. RESULTS FAP was widely expressed in advanced NSCLC tissues. FAP was correlated with decreased density of CD8 + T cells (Spearman's rho - 0.32, p < 0.001) and immunosuppressive tumor microenvironment (TME) status. No correlations were detected between FAP and PD-L1 expression or with the density of CD3 + T cells. The patients with higher expression of FAP showed worse response rate (16.4% vs. 38.7%, p < 0.001) and worse progression-free survival (HR = 2.56, 95% CI 1.69-3.87, p < 0.001). In addition, FAP contributed to shortened overall survival in subgroups of the patients with squamous cell lung cancer (p = 0.020), PD-1 blockade monotherapy (p = 0.017), and first-line therapy (p = 0.028). CONCLUSION FAP is a potential predictive biomarker of resistance to PD-1 blockade. Further investigation is warranted to identify a strategy for targeting FAP to alleviate the immunosuppressive TME and broaden the clinical effectiveness of PD-1 blockade therapy.
Collapse
Affiliation(s)
- Yan Zhao
- Department of Tumor Immunotherapy, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050035, China.,Department of Oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, China
| | - Yueping Liu
- Department of Pathology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, China
| | - Yunlong Jia
- Department of Tumor Immunotherapy, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050035, China
| | - Xiaoxiao Wang
- Department of Pathology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, China
| | - Jiankun He
- Department of Pathology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, China
| | - Shuman Zhen
- Department of Tumor Immunotherapy, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050035, China
| | - Jiali Wang
- Department of Tumor Immunotherapy, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050035, China
| | - Lihua Liu
- Department of Tumor Immunotherapy, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050035, China. .,Cancer Research Institute of Hebei Province, Shijiazhuang, 050011, China. .,China International Cooperation Laboratory of Stem Cell Research, Hebei Medical University, Shijiazhuang, 050011, China.
| |
Collapse
|
14
|
Tsujimae M, Masuda A, Ikegawa T, Tanaka T, Inoue J, Toyama H, Sofue K, Uemura H, Kohashi S, Inomata N, Nagao K, Masuda S, Abe S, Gonda M, Yamakawa K, Ashina S, Yamada Y, Tanaka S, Nakano R, Sakai A, Kobayashi T, Shiomi H, Kanzawa M, Itoh T, Fukumoto T, Ueda Y, Kodama Y. Comprehensive Analysis of Molecular Biologic Characteristics of Pancreatic Ductal Adenocarcinoma Concomitant with Intraductal Papillary Mucinous Neoplasm. Ann Surg Oncol 2022; 29:4924-4934. [PMID: 35606470 DOI: 10.1245/s10434-022-11704-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 02/14/2022] [Indexed: 11/18/2022]
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) concomitant with intraductal papillary mucinous neoplasm (IPMN) is defined as PDAC occurring apart from IPMN. This study comprehensively investigated the molecular biologic characteristics of PDAC concomitant with IPMN in major genetic alterations, tumor microenvironment, and prognosis by contrast with those of conventional PDAC. METHODS The study retrospectively reviewed the data of 158 surgically resected PDAC patients. The driver gene alteration status (KRAS, TP53, CDKN2A, SMAD4, and GNAS) together with the immune and fibrotic status in tumor was evaluated. The prognosis of PDAC concomitant with IPMN and that of conventional PDAC also were compared. RESULTS No statistically significant difference was found between PDAC concomitant with IPMN and conventional PDAC in the alteration frequency analysis of the major driver genes and the immune and fibrotic status in the tumor microenvironment. Overall survival and disease-free survival between patients who had PDAC concomitant with IPMN and those who had conventional PDAC did not show statistically significant differences in propensity-matched subjects. Furthermore, the co-existence of IPMN was not a poor prognostic factor in the multivariable-adjusted Cox proportional hazards model (hazard ratio, 0.95; 95 % confidence interval, 0.51-1.78). CONCLUSIONS In this study, PDAC concomitant with IPMN had tumor characteristics similar to those of conventional PDAC in terms of the major driver gene alterations, tumor microenvironment, and prognosis.
Collapse
Affiliation(s)
- Masahiro Tsujimae
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, Hyogo, Japan
| | - Atsuhiro Masuda
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, Hyogo, Japan.
| | - Takuya Ikegawa
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, Hyogo, Japan
| | - Takeshi Tanaka
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, Hyogo, Japan
| | - Jun Inoue
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, Hyogo, Japan
| | - Hirochika Toyama
- Division of Hepato-Biliary-Pancreatic Surgery, Department of Surgery, Kobe University Graduate School of Medicine, Kobe, Hyogo, Japan
| | - Keitaro Sofue
- Department of Radiology, Kobe University Graduate School of Medicine, Kobe, Hyogo, Japan
| | - Hisahiro Uemura
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, Hyogo, Japan
| | - Shinya Kohashi
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, Hyogo, Japan
| | - Noriko Inomata
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, Hyogo, Japan
| | - Kae Nagao
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, Hyogo, Japan
| | - Shigeto Masuda
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, Hyogo, Japan
| | - Shohei Abe
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, Hyogo, Japan
| | - Masanori Gonda
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, Hyogo, Japan
| | - Kohei Yamakawa
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, Hyogo, Japan
| | - Shigeto Ashina
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, Hyogo, Japan
| | - Yasutaka Yamada
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, Hyogo, Japan
| | - Shunta Tanaka
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, Hyogo, Japan
| | - Ryota Nakano
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, Hyogo, Japan
| | - Arata Sakai
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, Hyogo, Japan
| | - Takashi Kobayashi
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, Hyogo, Japan
| | - Hideyuki Shiomi
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, Hyogo, Japan
| | - Maki Kanzawa
- Division of Diagnostic Pathology, Kobe University Graduate School of Medicine, Kobe, Hyogo, Japan
| | - Tomoo Itoh
- Division of Diagnostic Pathology, Kobe University Graduate School of Medicine, Kobe, Hyogo, Japan
| | - Takumi Fukumoto
- Division of Hepato-Biliary-Pancreatic Surgery, Department of Surgery, Kobe University Graduate School of Medicine, Kobe, Hyogo, Japan
| | - Yoshihide Ueda
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, Hyogo, Japan
| | - Yuzo Kodama
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, Hyogo, Japan
| |
Collapse
|
15
|
Azarafza M, Tehrani M, Valadan R, Maleki I, Mohammad Mehdi Ghaffari-Hamedani S, Ghanadan A, Alizadeh-Navaei R, Ajami A. Role of BTLA/HVEM network in development of gastric cancer. Hum Immunol 2022; 83:637-644. [DOI: 10.1016/j.humimm.2022.07.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Revised: 07/15/2022] [Accepted: 07/15/2022] [Indexed: 12/24/2022]
|
16
|
Duan Z, Zhang Y, Tang Y, Gao R, Bao J, Liang B. Adjuvant therapy for periampullary carcinoma and the significance of histopathological typing: A systematic review. Transl Oncol 2022; 20:101414. [PMID: 35397420 PMCID: PMC9006738 DOI: 10.1016/j.tranon.2022.101414] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 03/24/2022] [Accepted: 03/30/2022] [Indexed: 12/19/2022] Open
Abstract
The efficacy of adjuvant therapy for periampullary carcinoma is controversial. There is a trend of classification periampullary carcinoma into PB-type and IN-type, and the prognosis of different subtypes may be significantly different. The PB-type patients who accepted gemcitabine based chemotherapy and IN-type patients who accepted 5-FU based chemotherapy, maybe improved the prognosis. Chemoradiotherapy appears to be more effective in patients with advanced stages. There are few related studies on targeted therapy and immunotherapy, and further research is needed.
Objective This review investigates the role of adjuvant therapy (AT) and the importance of histopathological typing in periampullary carcinoma (PAC) treatment. Background PAC is a relatively rare gastrointestinal malignancy. The regimen and effect of AT in PAC are still controversial. However, there is a treatment based on histopathological types (pancreaticobiliary-type, PB-type or intestinal-type, IN-type), but there are no clear guidelines indicating that typing can be used to guide the selection of AT drugs. Methods A literature search of PubMed and Web of Science databases was conducted for studies published from January 2001 to August 2021 on the use of AT in PAC. Results A total of 75 studies were included in this review. According to existing studies, AT for PAC is mostly based on 5-FU or gemcitabine, but the effect is unknown. However, when PAC is classified into different histopathological types, AT with gemcitabine is beneficial for patients with the PB-type of PAC, while 5-FU-based AT is beneficial for patients with the IN-type of PAC. In addition, the benefits of AT are more pronounced in patients with a high-risk disease, such as patients with stage II/III, T3/T4 tumors, or positive lymph node involvement. There are few studies on targeted therapy and immunotherapy for PAC. Conclusions This review suggests that AT has potential survival benefits, especially when based on the histopathologic type that helps the choice of drugs during AT in PAC patients.
Collapse
Affiliation(s)
- Zhiqing Duan
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yinuo Zhang
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yajie Tang
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Ruqing Gao
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jing Bao
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Bo Liang
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China.
| |
Collapse
|
17
|
Development and characterization of anti-galectin-9 antibodies that protect T cells from galectin-9-induced cell death. J Biol Chem 2022; 298:101821. [PMID: 35283189 PMCID: PMC9006662 DOI: 10.1016/j.jbc.2022.101821] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 03/03/2022] [Accepted: 03/08/2022] [Indexed: 12/13/2022] Open
Abstract
Antibodies that target immune checkpoint proteins such as programmed cell death protein 1, programmed death ligand 1, and cytotoxic T-lymphocyte–associated antigen 4 in human cancers have achieved impressive clinical success; however, a significant proportion of patients fail to respond to these treatments. Galectin-9 (Gal-9), a β-galactoside-binding protein, has been shown to induce T-cell death and facilitate immunosuppression in the tumor microenvironment by binding to immunomodulatory receptors such as T-cell immunoglobulin and mucin domain–containing molecule 3 and the innate immune receptor dectin-1, suggesting that it may have potential as a target for cancer immunotherapy. Here, we report the development of two novel Gal-9-neutralizing antibodies that specifically react with the N-carbohydrate-recognition domain of human Gal-9 with high affinity. We also show using cell-based functional assays that these antibodies efficiently protected human T cells from Gal-9-induced cell death. Notably, in a T-cell/tumor cell coculture assay of cytotoxicity, these antibodies significantly promoted T cell-mediated killing of tumor cells. Taken together, our findings demonstrate potent inhibition of human Gal-9 by neutralizing antibodies, which may open new avenues for cancer immunotherapy.
Collapse
|
18
|
Muller M, Haghnejad V, Schaefer M, Gauchotte G, Caron B, Peyrin-Biroulet L, Bronowicki JP, Neuzillet C, Lopez A. The Immune Landscape of Human Pancreatic Ductal Carcinoma: Key Players, Clinical Implications, and Challenges. Cancers (Basel) 2022; 14:cancers14040995. [PMID: 35205742 PMCID: PMC8870260 DOI: 10.3390/cancers14040995] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Revised: 02/14/2022] [Accepted: 02/15/2022] [Indexed: 02/04/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most aggressive and deadliest cancer worldwide with an overall survival rate, all stages combined, of still <10% at 5 years. The poor prognosis is attributed to challenges in early detection, a low opportunity for radical resection, limited response to chemotherapy, radiotherapy, and resistance to immune therapy. Moreover, pancreatic tumoral cells are surrounded by an abundant desmoplastic stroma, which is responsible for creating a mechanical barrier, preventing appropriate vascularization and leading to poor immune cell infiltration. Accumulated evidence suggests that PDAC is impaired with multiple “immune defects”, including a lack of high-quality effector cells (CD4, CD8 T cells, dendritic cells), barriers to effector cell infiltration due to that desmoplastic reaction, and a dominance of immune cells such as regulatory T cells, myeloid-derived suppressor cells, and M2 macrophages, resulting in an immunosuppressive tumor microenvironment (TME). Although recent studies have brought new insights into PDAC immune TME, its understanding remains not fully elucidated. Further studies are required for a better understanding of human PDAC immune TME, which might help to develop potent new therapeutic strategies by correcting these immune defects with the hope to unlock the resistance to (immune) therapy. In this review, we describe the main effector immune cells and immunosuppressive actors involved in human PDAC TME, as well as their implications as potential biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Marie Muller
- Department of Gastroenterology, Nancy University Hospital, University of Lorraine, 54500 Vandœuvre-lès-Nancy, France; (V.H.); (M.S.); (B.C.); (L.P.-B.); (J.-P.B.); (A.L.)
- Correspondence:
| | - Vincent Haghnejad
- Department of Gastroenterology, Nancy University Hospital, University of Lorraine, 54500 Vandœuvre-lès-Nancy, France; (V.H.); (M.S.); (B.C.); (L.P.-B.); (J.-P.B.); (A.L.)
| | - Marion Schaefer
- Department of Gastroenterology, Nancy University Hospital, University of Lorraine, 54500 Vandœuvre-lès-Nancy, France; (V.H.); (M.S.); (B.C.); (L.P.-B.); (J.-P.B.); (A.L.)
| | - Guillaume Gauchotte
- Department of Pathology, Nancy University Hospital, University of Lorraine, 54500 Vandœuvre-lès-Nancy, France;
- INSERM U1256, NGERE, Faculty of Medicine, University of Lorraine, 54500 Vandœuvre-lès-Nancy, France
| | - Bénédicte Caron
- Department of Gastroenterology, Nancy University Hospital, University of Lorraine, 54500 Vandœuvre-lès-Nancy, France; (V.H.); (M.S.); (B.C.); (L.P.-B.); (J.-P.B.); (A.L.)
| | - Laurent Peyrin-Biroulet
- Department of Gastroenterology, Nancy University Hospital, University of Lorraine, 54500 Vandœuvre-lès-Nancy, France; (V.H.); (M.S.); (B.C.); (L.P.-B.); (J.-P.B.); (A.L.)
- INSERM U1256, NGERE, Faculty of Medicine, University of Lorraine, 54500 Vandœuvre-lès-Nancy, France
| | - Jean-Pierre Bronowicki
- Department of Gastroenterology, Nancy University Hospital, University of Lorraine, 54500 Vandœuvre-lès-Nancy, France; (V.H.); (M.S.); (B.C.); (L.P.-B.); (J.-P.B.); (A.L.)
- INSERM U1256, NGERE, Faculty of Medicine, University of Lorraine, 54500 Vandœuvre-lès-Nancy, France
| | - Cindy Neuzillet
- Medical Oncology Department, Curie Institute, Versailles Saint-Quentin University (UVQ), Paris Saclay University, 92064 Saint-Cloud, France;
| | - Anthony Lopez
- Department of Gastroenterology, Nancy University Hospital, University of Lorraine, 54500 Vandœuvre-lès-Nancy, France; (V.H.); (M.S.); (B.C.); (L.P.-B.); (J.-P.B.); (A.L.)
| |
Collapse
|
19
|
Liang H, Li T, Fang X, Xing Z, Zhang S, Shi L, Li W, Guo L, Kuang C, Liu H, Yang Q. IDO1/TDO dual inhibitor RY103 targets Kyn-AhR pathway and exhibits preclinical efficacy on pancreatic cancer. Cancer Lett 2021; 522:32-43. [PMID: 34520819 DOI: 10.1016/j.canlet.2021.09.012] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Revised: 08/30/2021] [Accepted: 09/09/2021] [Indexed: 12/12/2022]
Abstract
Indoleamine 2,3-dioxygenase 1 (IDO1) catalyzing the conversion of tryptophan (Trp) to kynurenine (Kyn) in kynurenine pathway (KP) is involved in the immunosuppression in pancreatic cancer (PC), but the value of IDO1 as an independent prognostic marker for PC is uncertain. Moreover, the correlation between tryptophan 2,3-dioxygenase (TDO), an isozyme of IDO1, and PC is largely unknown. Using TCGA database, the correlation between IDO1 and/or TDO expression and PC patients' survival was analyzed. The expressions of IDO1 and TDO in PC cells and PC mice were examined. The effects of IDO1, TDO or dual inhibition on IDO1 and TDO effector pathway (Aryl hydrocarbon receptor, AhR) and on migration and invasion of PC cells were investigated. The block effect of IDO1/TDO dual inhibitor RY103 on KP was evaluated. The preclinical efficacy of RY103 and its immunomodulatory effect on KPIC orthotopic PC mice and Pan02 tumor-bearing mice were explored. Results showed that IDO1/TDO co-expression is an independent prognostic marker for PC. RY103 can significantly block KP and target Kyn-AhR pathway to blunt the migration and invasion of PC cells, exhibit preclinical efficacy and ameliorate IDO1/TDO-mediated immunosuppression in PC mice.
Collapse
Affiliation(s)
- Heng Liang
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Songhu Road 2005, Shanghai, 200438, China.
| | - Tianqi Li
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Songhu Road 2005, Shanghai, 200438, China.
| | - Xin Fang
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Songhu Road 2005, Shanghai, 200438, China.
| | - Zikang Xing
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Songhu Road 2005, Shanghai, 200438, China.
| | - Shengnan Zhang
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Songhu Road 2005, Shanghai, 200438, China.
| | - Lei Shi
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Songhu Road 2005, Shanghai, 200438, China.
| | - Weirui Li
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Songhu Road 2005, Shanghai, 200438, China.
| | - Leilei Guo
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Songhu Road 2005, Shanghai, 200438, China.
| | - Chunxiang Kuang
- Shanghai Key Lab of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Tongji University, Siping Road 1239, Shanghai, 200092, China.
| | - Hongrui Liu
- Department of Pharmacology, School of Pharmacy, Fudan University, Zhangheng Road 826, Shanghai, 201203, China.
| | - Qing Yang
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Songhu Road 2005, Shanghai, 200438, China.
| |
Collapse
|
20
|
Shayesteh S, Guillemin GJ, Rashidian A, Faghir-Ghanesefat H, Mani AR, Tavangar SM, Dehpour AR. 1-Methyl tryptophan, an indoleamine 2,3-dioxygenase inhibitor, attenuates cardiac and hepatic dysfunction in rats with biliary cirrhosis. Eur J Pharmacol 2021; 908:174309. [PMID: 34252442 DOI: 10.1016/j.ejphar.2021.174309] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 07/02/2021] [Accepted: 07/05/2021] [Indexed: 12/30/2022]
Abstract
Kynurenine Pathway (KP) is the dominant metabolic route of tryptophan which is catalyzed by indoleamine-2,3-dioxygenase (IDO). This pathway is upregulated in liver disease where the level of KP metabolites correlates with the severity of disease. Cirrhosis is associated with cardiac dysfunction, which manifests itself during severe physiological challenges such as liver transplantation. Cardiac dysfunction in cirrhosis is linked to systemic inflammation and impaired cardiac beta-adrenergic signaling pathways. The KP pathway is involved in modulation of cardiac signaling and is upregulated by systemic inflammation. Therefore, this study aimed to evaluate the effect of IDO inhibition on development of cardiac dysfunction in an experimental model of cirrhosis. Cirrhosis was induced by bile duct ligation (BDL). Experimental groups were given either 1-methyl tryptophan (1-MT, 1, 3, 9 mg/kg), or saline. 28 days after BDL, cardiac chronotropic response to epinephrine was assessed ex vivo. HPLC was employed to measure hepatic and cardiac levels of tryptophan, kynurenine and kynurenic acid. Cirrhosis in rats was associated with impaired cardiac chronotropic responsiveness to adrenergic stimulation. 1-MT dose-dependently improved cirrhosis-induced chronotropic dysfunction as well as elevated serum levels of CRP and IL-6 in BDL rats. Hepatic and cardiac kynurenine/tryptophan ratio were elevated in cirrhotic rats and were reduced following 1-MT administration. Chronic administration of 1-MT could also reduce hepatic inflammation, fibrosis and ductular proliferation. 1-MT attenuates cardiac dysfunction in rats with biliary cirrhosis. This protective effect is not limited to the cardiac function as liver histopathologic changes were also improved following chronic 1-MT administration.
Collapse
Affiliation(s)
- Sevda Shayesteh
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran; Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Gilles J Guillemin
- Neuroinflammation Group, Department of Biomedical Sciences, Centre for Motor Neuron Disease Research, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW, Australia
| | - Amir Rashidian
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Hedyeh Faghir-Ghanesefat
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran; Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Ali R Mani
- Division of Medicine, University College London, London, UK
| | - Seyed Mohammad Tavangar
- Department of Pathology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Ahmad R Dehpour
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran; Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
21
|
A Critical Assessment of the Association between HLA-G Expression by Carcinomas and Clinical Outcome. Int J Mol Sci 2021; 22:ijms22158265. [PMID: 34361031 PMCID: PMC8347921 DOI: 10.3390/ijms22158265] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 07/23/2021] [Accepted: 07/28/2021] [Indexed: 01/04/2023] Open
Abstract
Human leukocyte antigen-G (HLA-G) conveys immunological tolerance at the maternal-foetal interface. HLA-G expression by tumour cells may also play such a role, resulting in tumour immune evasion, making HLA-G a potential target for immunotherapies. The aim of this review was to determine to what extent it is justified that HLA-G expression is considered as a target for immune checkpoint inhibiting therapy by critically assessing the association between HLA-G expression by carcinomas and clinical outcome of patients. The used HLA-G-detecting mAb, HLA-G quantification methods and statistically significant HLA-G-associated clinicopathological parameters are discussed. Tumour HLA-G expression correlated with poor clinical outcome in breast, esophageal, gastric and hepatocellular carcinoma patients. Tumour HLA-G expression was not associated with clinical outcome in ovarian and oral carcinoma patients. Cervical, colorectal, lung, and pancreatic carcinoma patients presented discrepant and therefore inconclusive results regarding the association between tumour HLA-G expression and clinical outcome. These disparities might partly be the result of differences in the methodological approach to quantify HLA-G expression between studies. Therefore, implementation of universal methodological procedures is strongly advised. Overall, HLA-G expression did not univocally result in poor clinical outcome of carcinoma patients. This implies that tumour HLA-G expression is not necessarily part of an inhibited tumour-immune response and tumour progression. Consequently, it remains elusive whether HLA-G expression by carcinomas functions as an immune checkpoint molecule affecting a tumour-immune response. It may also reflect derailed control of gene expression in tumours, with no real functional consequences.
Collapse
|
22
|
Popp FC, Capino I, Bartels J, Damanakis AI, Li J, Datta RR, Löser H, Zhao Y, Quaas A, Lohneis P, Bruns CJ. Expression of Immune Checkpoint Regulators IDO, VISTA, LAG3, and TIM3 in Resected Pancreatic Ductal Adenocarcinoma. Cancers (Basel) 2021; 13:2689. [PMID: 34072549 PMCID: PMC8198722 DOI: 10.3390/cancers13112689] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Accepted: 05/20/2021] [Indexed: 01/22/2023] Open
Abstract
Pancreatic cancer features elaborate mechanisms of immune evasion. The potential of new immune molecules was explored to restore the antitumor immune response. If these immune molecules are associated with poor survival, specific drugs could take effect. Here, we analyze the expression of VISTA, LAG3, IDO, and TIM3 on tumor-infiltrating lymphocytes (TILs) and its impact on patient survival. We analyzed 153 pancreatic cancer patients from the prospectively managed database of the multicentered PANCALYZE study. Immunohistochemistry on a tissue microarray assessed VISTA, LAG3, IDO, and TIM3 expression of TILs from the patients undergoing primary resection. Complementarily, we analyzed publicly available transcriptomic data (n = 903). Successful completion of chemotherapy, and lymph node status were independent predictors of survival in the multivariate analysis of the clinicopathologic parameters. Fifteen tumors were exclusively VISTA-positive, thirteen tumors expressed VISTA together with TIM3, and ten tumors expressed VISTA together with IDO. Patients featuring tumors with high numbers of IDO-positive TILs had better patient survival (p = 0.037). VISTA, LAG3, and TIM3 expression did not correlate with survival. The analysis of publicly available data did not show survival differences. Tumors rarely co-express more than two immune molecules at the same time, and VISTA is most frequently co-expressed. Although IDO generally inhibits T-cell proliferation, a high expression of IDO was associated with improved survival. We expect immune checkpoint inhibitors against VISTA, LAG3, and TIM3 to be inefficient in a clinical application.
Collapse
Affiliation(s)
- Felix C. Popp
- Department of General, Visceral, Cancer and Transplantation Surgery, University of Cologne, 50937 Cologne, Germany; (F.C.P.); (I.C.); (J.B.); (A.I.D.); (J.L.); (R.R.D.); (Y.Z.)
| | - Ingracia Capino
- Department of General, Visceral, Cancer and Transplantation Surgery, University of Cologne, 50937 Cologne, Germany; (F.C.P.); (I.C.); (J.B.); (A.I.D.); (J.L.); (R.R.D.); (Y.Z.)
| | - Joana Bartels
- Department of General, Visceral, Cancer and Transplantation Surgery, University of Cologne, 50937 Cologne, Germany; (F.C.P.); (I.C.); (J.B.); (A.I.D.); (J.L.); (R.R.D.); (Y.Z.)
| | - Alexander I. Damanakis
- Department of General, Visceral, Cancer and Transplantation Surgery, University of Cologne, 50937 Cologne, Germany; (F.C.P.); (I.C.); (J.B.); (A.I.D.); (J.L.); (R.R.D.); (Y.Z.)
| | - Jiahui Li
- Department of General, Visceral, Cancer and Transplantation Surgery, University of Cologne, 50937 Cologne, Germany; (F.C.P.); (I.C.); (J.B.); (A.I.D.); (J.L.); (R.R.D.); (Y.Z.)
| | - Rabi R. Datta
- Department of General, Visceral, Cancer and Transplantation Surgery, University of Cologne, 50937 Cologne, Germany; (F.C.P.); (I.C.); (J.B.); (A.I.D.); (J.L.); (R.R.D.); (Y.Z.)
| | - Heike Löser
- Institute of Pathology, University of Cologne, 50937 Cologne, Germany; (H.L.); (A.Q.); (P.L.)
| | - Yue Zhao
- Department of General, Visceral, Cancer and Transplantation Surgery, University of Cologne, 50937 Cologne, Germany; (F.C.P.); (I.C.); (J.B.); (A.I.D.); (J.L.); (R.R.D.); (Y.Z.)
| | - Alexander Quaas
- Institute of Pathology, University of Cologne, 50937 Cologne, Germany; (H.L.); (A.Q.); (P.L.)
| | - Philipp Lohneis
- Institute of Pathology, University of Cologne, 50937 Cologne, Germany; (H.L.); (A.Q.); (P.L.)
| | - Christiane J. Bruns
- Department of General, Visceral, Cancer and Transplantation Surgery, University of Cologne, 50937 Cologne, Germany; (F.C.P.); (I.C.); (J.B.); (A.I.D.); (J.L.); (R.R.D.); (Y.Z.)
| | | |
Collapse
|
23
|
Geerdes EE, Sideras K, Aziz MH, van Eijck CH, Bruno MJ, Sprengers D, Boor PPC, Kwekkeboom J. Cancer Cell B7-H3 Expression Is More Prevalent in the Pancreato-Biliary Subtype of Ampullary Cancer Than in Pancreatic Cancer. Front Oncol 2021; 11:615691. [PMID: 33996541 PMCID: PMC8117087 DOI: 10.3389/fonc.2021.615691] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Accepted: 03/30/2021] [Indexed: 11/29/2022] Open
Abstract
B7-H3 is an immunomodulatory member of the B7-superfamily with limited expression in normal tissues, but overexpression in several types of cancer. Therefore it is currently being explored as a potential target for cancer immunotherapy. The biological relevance of B7-H3 expression in pancreatic cancer is unclear, while there are no data on B7-H3 expression in ampullary cancer. We aimed to compare intra-tumoral B7-H3 expression between these two closely related cancer types and analyze its association with post-surgical disease course. B7-H3 expression levels were determined by immunohistochemistry in tissue microarrays of resected tumors of 137 pancreatic cancer patients and 83 patients with ampullary cancer of the pancreato-biliary subtype. B7-H3 was more frequently expressed in cancer cells of ampullary cancer patients compared to pancreatic cancer patients (51% versus 21%; p< 0.001). In ampullary cancer patients, but not in pancreatic cancer patients, B7-H3 cancer cell expression was associated with longer disease-free survival and patient survival. However, the prognostic value of B7-H3 was lost upon adjustment for CA19-9 levels. The frequencies of B7-H3 expression in tumor stroma did not differ between the two types of cancer (66% versus 63%). In both cancer types, stromal B7-H3 expression was not associated with post-surgical disease course. Compared to pancreatic cancer, B7-H3 is more frequently expressed in cancer cells of patients with the pancreato-biliary subtype of ampullary cancer. These data suggest that B7-H3 may represent an interesting potential target for immunotherapy in ampullary cancer rather than in pancreatic cancer.
Collapse
Affiliation(s)
- Emma E Geerdes
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, Netherlands
| | - Kostandinos Sideras
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, Netherlands.,Department of Surgery, Erasmus MC-University Medical Center, Rotterdam, Netherlands
| | - M Hosein Aziz
- Department of Surgery, Erasmus MC-University Medical Center, Rotterdam, Netherlands
| | - Casper H van Eijck
- Department of Surgery, Erasmus MC-University Medical Center, Rotterdam, Netherlands
| | - Marco J Bruno
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, Netherlands
| | - Dave Sprengers
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, Netherlands
| | - Patrick P C Boor
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, Netherlands
| | - Jaap Kwekkeboom
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, Netherlands
| |
Collapse
|
24
|
Ma B, Meng H, Tian Y, Wang Y, Song T, Zhang T, Wu Q, Cui Y, Li H, Zhang W, Li Q. High expression of HVEM is associated with improved prognosis in intrahepatic cholangiocarcinoma. Oncol Lett 2020; 21:69. [PMID: 33365080 PMCID: PMC7716701 DOI: 10.3892/ol.2020.12330] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Accepted: 10/29/2020] [Indexed: 01/03/2023] Open
Abstract
Herpesvirus entry mediator (HVEM) displays dual signals in T-cell activation according to the ligands and intracytoplasmic effectors it interacts with. High HVEM expression may play an immunosuppressive role in several malignancies. The present study investigated the clinical impact of HVEM on intrahepatic cholangiocarcinoma (ICC), including its prognostic value, and association with clinicopathological features and immune status. The clinical data of 102 consecutive patients with ICC who underwent surgical treatment from January 2012 to December 2017 were collected. The expression of HVEM and different types of tumor-infiltrating lymphocytes (TILs) were investigated in ICC tissue samples by immunohistochemical staining. HVEM expression was detected in the tumor tissues of 92 (90.2%) patients with ICC. Patients with high HVEM expression were more likely to have increased peripheral blood lymphocyte (PBL) concentrations (P=0.031), decreased CEA (P=0.036), low TNM stage (P=0.043) and high frequencies of small-duct histological type (P=0.021) and BAP1 retained expression (P=0.010). Survival analysis showed that high HVEM expression was a favorable independent predictor of overall postoperative survival (P=0.034, hazard ratio=0.486, 95% confidence interval=0.249–0.945). In addition, no significant association of HVEM expression with CD4+ (P=0.512), CD8+ (P=0.750) or CD45RO+ (P=0.078) TILs was identified in the ICC tissues. These results indicate that HVEM may serve as a favorable prognostic marker for ICC. Furthermore, co-stimulatory signals from HVEM may play a dominant role in the progression of ICCs, which can be explained by an increase in the number of PBLs rather than a change in the number of TILs. However, the function of the HVEM network in ICC progression is complex and requires further study.
Collapse
Affiliation(s)
- Bingqi Ma
- Department of Hepatobiliary Surgery, Tianjin Medical University Cancer Institute and Hospital; National Clinical Research Center for Cancer; Key Laboratory of Cancer Prevention and Therapy, Tianjin; Tianjin's Clinical Research Center for Cancer, Tianjin 300060, P.R. China.,Affiliated Hospital of Weifang Medical University, Weifang, Shandong 261031, P.R. China
| | - Huijuan Meng
- Affiliated Hospital of Weifang Medical University, Weifang, Shandong 261031, P.R. China
| | - Ye Tian
- Department of Senior Ward, Tianjin Medical University Cancer Institute and Hospital; National Clinical Research Center for Cancer; Key Laboratory of Cancer Prevention and Therapy, Tianjin; Tianjin's Clinical Research Center for Cancer, Tianjin 300060, P.R. China
| | - Yingying Wang
- Department of Hepatobiliary Surgery, Tianjin Medical University Cancer Institute and Hospital; National Clinical Research Center for Cancer; Key Laboratory of Cancer Prevention and Therapy, Tianjin; Tianjin's Clinical Research Center for Cancer, Tianjin 300060, P.R. China
| | - Tianqiang Song
- Department of Hepatobiliary Surgery, Tianjin Medical University Cancer Institute and Hospital; National Clinical Research Center for Cancer; Key Laboratory of Cancer Prevention and Therapy, Tianjin; Tianjin's Clinical Research Center for Cancer, Tianjin 300060, P.R. China
| | - Ti Zhang
- Department of Hepatobiliary Surgery, Tianjin Medical University Cancer Institute and Hospital; National Clinical Research Center for Cancer; Key Laboratory of Cancer Prevention and Therapy, Tianjin; Tianjin's Clinical Research Center for Cancer, Tianjin 300060, P.R. China
| | - Qiang Wu
- Department of Hepatobiliary Surgery, Tianjin Medical University Cancer Institute and Hospital; National Clinical Research Center for Cancer; Key Laboratory of Cancer Prevention and Therapy, Tianjin; Tianjin's Clinical Research Center for Cancer, Tianjin 300060, P.R. China
| | - Yunlong Cui
- Department of Hepatobiliary Surgery, Tianjin Medical University Cancer Institute and Hospital; National Clinical Research Center for Cancer; Key Laboratory of Cancer Prevention and Therapy, Tianjin; Tianjin's Clinical Research Center for Cancer, Tianjin 300060, P.R. China
| | - Huikai Li
- Department of Hepatobiliary Surgery, Tianjin Medical University Cancer Institute and Hospital; National Clinical Research Center for Cancer; Key Laboratory of Cancer Prevention and Therapy, Tianjin; Tianjin's Clinical Research Center for Cancer, Tianjin 300060, P.R. China
| | - Wei Zhang
- Department of Hepatobiliary Surgery, Tianjin Medical University Cancer Institute and Hospital; National Clinical Research Center for Cancer; Key Laboratory of Cancer Prevention and Therapy, Tianjin; Tianjin's Clinical Research Center for Cancer, Tianjin 300060, P.R. China
| | - Qiang Li
- Department of Hepatobiliary Surgery, Tianjin Medical University Cancer Institute and Hospital; National Clinical Research Center for Cancer; Key Laboratory of Cancer Prevention and Therapy, Tianjin; Tianjin's Clinical Research Center for Cancer, Tianjin 300060, P.R. China
| |
Collapse
|
25
|
Zhu JH, Yan QL, Wang JW, Chen Y, Ye QH, Wang ZJ, Huang T. The Key Genes for Perineural Invasion in Pancreatic Ductal Adenocarcinoma Identified With Monte-Carlo Feature Selection Method. Front Genet 2020; 11:554502. [PMID: 33193628 PMCID: PMC7593847 DOI: 10.3389/fgene.2020.554502] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 08/17/2020] [Indexed: 12/20/2022] Open
Abstract
Background Pancreatic ductal adenocarcinoma (PDAC) is the most aggressive form of pancreatic cancer. Its 5-year survival rate is only 3–5%. Perineural invasion (PNI) is a process of cancer cells invading the surrounding nerves and perineural spaces. It is considered to be associated with the poor prognosis of PDAC. About 90% of pancreatic cancer patients have PNI. The high incidence of PNI in pancreatic cancer limits radical resection and promotes local recurrence, which negatively affects life quality and survival time of the patients with pancreatic cancer. Objectives To investigate the mechanism of PNI in pancreatic cancer, we analyzed the gene expression profiles of tumors and adjacent tissues from 50 PDAC patients which included 28 patients with perineural invasion and 22 patients without perineural invasion. Method Using Monte-Carlo feature selection and Incremental Feature Selection (IFS) method, we identified 26 key features within which 15 features were from tumor tissues and 11 features were from adjacent tissues. Results Our results suggested that not only the tumor tissue, but also the adjacent tissue, was informative for perineural invasion prediction. The SVM classifier based on these 26 key features can predict perineural invasion accurately, with a high accuracy of 0.94 evaluated with leave-one-out cross validation (LOOCV). Conclusion The in-depth biological analysis of key feature genes, such as TNFRSF14, XPO1, and ATF3, shed light on the understanding of perineural invasion in pancreatic ductal adenocarcinoma.
Collapse
Affiliation(s)
- Jin-Hui Zhu
- Department of General Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Qiu-Liang Yan
- Department of General Surgery, Jinhua People's Hospital, Jinhua, China
| | - Jian-Wei Wang
- Department of Surgical Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yan Chen
- Department of General Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Qing-Huang Ye
- Department of General Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhi-Jiang Wang
- Department of General Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Tao Huang
- Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
26
|
Meireson A, Devos M, Brochez L. IDO Expression in Cancer: Different Compartment, Different Functionality? Front Immunol 2020; 11:531491. [PMID: 33072086 PMCID: PMC7541907 DOI: 10.3389/fimmu.2020.531491] [Citation(s) in RCA: 120] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 08/25/2020] [Indexed: 12/11/2022] Open
Abstract
Indoleamine 2,3-dioxygenase 1 (IDO1) is a cytosolic haem-containing enzyme involved in the degradation of tryptophan to kynurenine. Although initially thought to be solely implicated in the modulation of innate immune responses during infection, subsequent discoveries demonstrated IDO1 as a mechanism of acquired immune tolerance. In cancer, IDO1 expression/activity has been observed in tumor cells as well as in the tumor-surrounding stroma, which is composed of endothelial cells, immune cells, fibroblasts, and mesenchymal cells. IDO1 expression/activity has also been reported in the peripheral blood. This manuscript reviews available data on IDO1 expression, mechanisms of its induction, and its function in cancer for each of these compartments. In-depth study of the biological function of IDO1 according to the expressing (tumor) cell can help to understand if and when IDO1 inhibition can play a role in cancer therapy.
Collapse
Affiliation(s)
- Annabel Meireson
- Department of Dermatology, Ghent University Hospital, Ghent, Belgium.,Cancer Research Institute Ghent, Ghent, Belgium
| | - Michael Devos
- Department of Dermatology, Ghent University Hospital, Ghent, Belgium
| | - Lieve Brochez
- Department of Dermatology, Ghent University Hospital, Ghent, Belgium.,Cancer Research Institute Ghent, Ghent, Belgium
| |
Collapse
|
27
|
Yin J, Li L, Wang C, Zhang Y. Increased Galectin-9 expression, a prognostic biomarker of aGVHD, regulates the immune response through the Galectin-9 induced MDSC pathway after allogeneic hematopoietic stem cell transplantation. Int Immunopharmacol 2020; 88:106929. [PMID: 32889240 DOI: 10.1016/j.intimp.2020.106929] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Revised: 08/17/2020] [Accepted: 08/19/2020] [Indexed: 02/06/2023]
Abstract
Galectin-9 (Gal-9) is a β-galactoside-binding soluble lectin family member that exerts its primary biological functions via specific glycoconjugate interactions. Gal-9 expression is closely related to tumor occurrence, development, metastasis and prognosis. In transplant immunology, a high level of Gal-9 expression has been shown to markedly reduce the severity of acute graft rejection and effectively prolong survival time in organ and bone marrow transplantation (BMT) models. The main mechanism of Gal-9-mediated immunoregulation involves the Tim-3/Gal-9 axis in T cells. However, myeloid-derived suppressor cell (MDSC) accumulation in transgenic mice with persistently high Gal-9 expression was observed in a model of lung inflammation, indicating that a potential immunosuppressive mechanism distinct from the Gal-9/Tim-3 axis might exist. In the present study, increased Gal-9 expression and MDSC frequencies before acute graft-versus-host disease (aGVHD) onset were observed in patients who developed aGVHD. Patients with higher Gal-9 expression (≥14.8417 ng/ml) exhibited reduced overall survival and increased cumulative incidences of GVHD at +100 day. We considered the elevated Gal-9 expression before aGVHD onset a secondary inflammatory response. This increase might be part of a negative feedback pathway corresponding to aGVHD pathogenesis. Additionally, a high Gal-9 concentration induced MDSC proliferation in vivo and in vitro. Gal-9-induced MDSCs (G9-MDSCs) suppressed T cell proliferation and activation. An infusion of G9-MDSCs into a graft contributed to the successful control of severe aGVHD and long-term survival in an allogeneic (allo)-BMT mouse model. Thus, we speculated that increased Gal-9 expression after allo-hematopoietic stem cell transplantation is a potential prognostic biomarker of aGVHD. The Gal-9-associated immunosuppressive effects on aGVHD development might occurr through G9-MDSCs and were independent of the Gal-9/Tim-3 axis.
Collapse
Affiliation(s)
- Jin Yin
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jie-Fang Avenue, Wuhan 430030, China
| | - Lin Li
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jie-Fang Avenue, Wuhan 430030, China
| | - Chunyan Wang
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jie-Fang Avenue, Wuhan 430030, China
| | - Yicheng Zhang
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jie-Fang Avenue, Wuhan 430030, China.
| |
Collapse
|
28
|
Xue Y, Balci S, Aydin Mericoz C, Taskin OC, Jiang H, Pehlivanoglu B, Muraki T, Memis B, Saka B, Kim GE, Bandopadhyay S, Knight J, El-Rayes BF, Sarmiento J, Reid MD, Erkan M, Basturk O, Adsay V. Frequency and clinicopathologic associations of DNA mismatch repair protein deficiency in ampullary carcinoma: Routine testing is indicated. Cancer 2020; 126:4788-4799. [PMID: 32857459 DOI: 10.1002/cncr.33135] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 06/09/2020] [Accepted: 07/06/2020] [Indexed: 12/11/2022]
Abstract
BACKGROUND The significance of DNA mismatch repair (MMR) deficiency in ampullary cancers (ACs) has not been established. METHODS In total, 127 ACs with invasive carcinomas measuring ≥3 mmthat had adequate tissue were analyzed immunohistochemically. RESULTS MMR loss was detected in 18% of ACs (higher than in colorectal cancers). Twelve tumors with MLH1-PMS2 loss were negative for BRAF V600E mutation, suggesting a Lynch syndrome association. MMR-deficient tumors (n = 23), comparedwith MMR-intact tumors (n = 104), showed a striking male predominance (male:female ratio, 4.7). Although the deficient tumors had slightly larger invasion size (2.7 vs 2.1 cm), they also had more expansile growth and less invasiveness, including less perineural invasion, and they ultimately had lower tumor (T) classification and less lymph node metastasis (30% vs 53%; P = .04). More important, patients who had MMR-deficient tumors had better clinical outcomes, with a 5-year overall survival rate of 68% versus 45% (P = .03), which was even more pronounced in those who had higher Tclassification (5-year overall survival, 69% vs 34%; P = .04). MMR deficiencyhad a statistically significant association with medullary phenotype, pushing-border invasion, and tumor-infiltrating immune cells, and it occurred more frequently in ampullary-duodenal type tumors. Programed cell death-ligand 1 (PD-L1) levels analyzed in the 22 MMR-deficient ACs revealed that all medullary carcinomas were positive. Nonmedullary MMR-deficient carcinomas expressed PD-L1 in 33% of tumors cells according to the criteria for a combined positive score ≥1, but all were negative according to the tumor proportion score≥1 method. CONCLUSIONS In ACs, MMR deficiency is even more frequent (18%) than in colon cancer and often has a Lynch-suggestive profile, thus routine testing is warranted. Male gender, pushing-border infiltration, ampullary-duodenal origin, medullary histology, and tumor-related inflammation have a significantly higher association with MMR deficiency. MMR-deficient tumors have less aggressive behavior. PD-L1 expression is common in medullary-phenotype ACs, thus immunotherapy should be considered at least for this group.
Collapse
Affiliation(s)
- Yue Xue
- Department of Pathology, Emory University, Atlanta, Georgia
| | - Serdar Balci
- Department of Pathology, Emory University, Atlanta, Georgia
| | - Cisel Aydin Mericoz
- Department of Pathology, Koç University School of Medicine, Istanbul, Turkey
| | - Orhun C Taskin
- Department of Pathology, Koç University School of Medicine, Istanbul, Turkey
| | - Hongmei Jiang
- Department of Statistics, Northwestern University, Evanston, Illinois
| | | | - Takashi Muraki
- Department of Pathology, Emory University, Atlanta, Georgia
| | - Bahar Memis
- Department of Pathology, Emory University, Atlanta, Georgia
| | - Burcu Saka
- Department of Pathology, Emory University, Atlanta, Georgia
| | - Grace E Kim
- Department of Pathology, University of California San Francisco, San Francisco, California
| | | | - Jessica Knight
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, Georgia
| | - Bassel F El-Rayes
- Department of Hematology and Medical Oncology, Emory University, Atlanta, Georgia
| | - Juan Sarmiento
- Department of Surgery, School of Medicine, Emory University, Atlanta, Georgia
| | | | - Mert Erkan
- Department of Surgery, Koç University School of Medicine, Istanbul, Turkey
| | - Olca Basturk
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Volkan Adsay
- Department of Pathology, Koç University School of Medicine, Istanbul, Turkey.,Koç University Research Center for Translational Medicine (KUTTAM), Istanbul, Turkey
| |
Collapse
|
29
|
Hiraoka N, Ino Y, Hori S, Yamazaki‐Itoh R, Naito C, Shimasaki M, Esaki M, Nara S, Kishi Y, Shimada K, Nakamura N, Torigoe T, Heike Y. Expression of classical human leukocyte antigen class I antigens, HLA-E and HLA-G, is adversely prognostic in pancreatic cancer patients. Cancer Sci 2020; 111:3057-3070. [PMID: 32495519 PMCID: PMC7419048 DOI: 10.1111/cas.14514] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 05/20/2020] [Accepted: 05/27/2020] [Indexed: 01/18/2023] Open
Abstract
The expression of classical human leukocyte antigen class I antigens (HLA-I) on the surfaces of cancer cells allows cytotoxic T cells to recognize and eliminate these cells. Reduction or loss of HLA-I is a mechanism of escape from antitumor immunity. The present study aimed to investigate the clinicopathological impacts of HLA-I and non-classical HLA-I antigens expressed on pancreatic ductal adenocarcinoma (PDAC) cells. We performed immunohistochemistry to detect expression of HLA-I antigens in PDAC using 243 PDAC cases and examined their clinicopathological influences. We also investigated the expression of immune-related genes to characterize PDAC tumor microenvironments. Lower expression of HLA-I, found in 33% of PDAC cases, was significantly associated with longer overall survival. Higher expression of both HLA-E and HLA-G was significantly associated with shorter survival. Multivariate analyses revealed that higher expression of these three HLA-I antigens was significantly correlated with shorter survival. Higher HLA-I expression on PDAC cells was significantly correlated with higher expression of IFNG, which also correlated with PD1, PD-L1 and PD-L2 expression. In vitro assay revealed that interferon gamma (IFNγ) stimulation increased surface expression of HLA-I in three PDAC cell lines. It also upregulated surface expression of HLA-E, HLA-G and immune checkpoint molecules, including PD-L1 and PD-L2. These results suggest that the higher expression of HLA-I, HLA-E and HLA-G on PDAC cells is an unfavorable prognosticator. It is possible that IFNγ promotes a tolerant microenvironment by inducing immune checkpoint molecules in PDAC tissues with higher HLA-I expression on PDAC cells.
Collapse
Affiliation(s)
- Nobuyoshi Hiraoka
- Division of Molecular PathologyNational Cancer Center Research InstituteTokyoJapan
- Department of Analytical PathologyNational Cancer Center Research InstituteTokyoJapan
- Division of Pathology and Clinical LaboratoriesNational Cancer Center HospitalTokyoJapan
| | - Yoshinori Ino
- Division of Molecular PathologyNational Cancer Center Research InstituteTokyoJapan
- Department of Analytical PathologyNational Cancer Center Research InstituteTokyoJapan
| | - Shutaro Hori
- Division of Molecular PathologyNational Cancer Center Research InstituteTokyoJapan
- Division of Pathology and Clinical LaboratoriesNational Cancer Center HospitalTokyoJapan
- Department of PathologyTokai University School of MedicineIseharaJapan
| | - Rie Yamazaki‐Itoh
- Division of Molecular PathologyNational Cancer Center Research InstituteTokyoJapan
| | - Chie Naito
- Division of Molecular PathologyNational Cancer Center Research InstituteTokyoJapan
| | - Mari Shimasaki
- Division of Molecular PathologyNational Cancer Center Research InstituteTokyoJapan
| | - Minoru Esaki
- Hepato‐Biliary and Pancreatic Surgery DivisionNational Cancer Center HospitalTokyoJapan
| | - Satoshi Nara
- Hepato‐Biliary and Pancreatic Surgery DivisionNational Cancer Center HospitalTokyoJapan
| | - Yoji Kishi
- Hepato‐Biliary and Pancreatic Surgery DivisionNational Cancer Center HospitalTokyoJapan
| | - Kazuaki Shimada
- Hepato‐Biliary and Pancreatic Surgery DivisionNational Cancer Center HospitalTokyoJapan
| | - Naoya Nakamura
- Department of PathologyTokai University School of MedicineIseharaJapan
| | | | - Yuji Heike
- Division of Biomedical SciencesSt. Luke’s International University Graduate School of Public HealthTokyoJapan
| |
Collapse
|
30
|
Amin S, Baine M, Meza J, Alam M, Lin C. The impact of immunotherapy on the survival of pancreatic adenocarcinoma patients who do not receive definitive surgery of the tumor. Clin Transl Radiat Oncol 2020; 24:34-40. [PMID: 32613090 PMCID: PMC7317682 DOI: 10.1016/j.ctro.2020.06.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 05/26/2020] [Accepted: 06/01/2020] [Indexed: 12/18/2022] Open
Abstract
Background and Purpose Immunotherapy has shown great efficacy in many cancers, but its role in pancreatic ductal adenocarcinoma (PDAC) remains unclear. The objective of this study was to investigate the impact of immunotherapy on the overall survival of PDAC patients who did not receive definitive surgery of the pancreatic primary tumor site using the National Cancer Database (NCDB). Materials and Methods Patients with pancreatic adenocarcinoma who did not receive surgery were identified from NCDB. Cox proportional hazard models were employed to assess the impact of immunotherapy on survival after adjusting for age at diagnosis, race, sex, place of living, income, education, treatment facility type, insurance status, year of diagnosis, and treatment types such as chemotherapy and radiation therapy. Results Of 263,886 patients who were analyzed, 911 (0.35%) received immunotherapy. Among patients who received chemotherapy (101,546), and chemoradiation (30,226) therapy, 555/101,546 (0.55%) received chemotherapy plus immunotherapy, and 299/3,022 (9.9%) received chemoradiation plus immunotherapy. In a multivariable analysis adjusted for the factors mentioned above, immunotherapy was associated with significantly improved OS (HR: 0.866 (0.800–0.937); P < 0.001) compared to no immunotherapy. Chemotherapy plus immunotherapy was significantly associated with improved OS (HR: 0.848 (0.766–0.938); P < 0.001) compared to chemotherapy without immunotherapy. Further, chemoradiation plus immunotherapy was associated with significantly improved OS (HR: 0.813 (0.707–0.936); P < 0.001) compared to chemoradiation alone. Conclusion In this study, the addition of immunotherapy to chemotherapy and chemoradiation therapy was associated with significantly improved OS in PDAC patients without definitive surgery. The study warrants future clinical trials of immunotherapy in PDAC.
Collapse
Affiliation(s)
- Saber Amin
- Department of Radiation Oncology, University of Nebraska Medical Center, USA
| | - Michael Baine
- Department of Radiation Oncology, University of Nebraska Medical Center, USA
| | - Jane Meza
- Department of Biostatistics, College of Public Health, University of Nebraska Medical Center, USA
| | - Morshed Alam
- Department of Biostatistics, College of Public Health, University of Nebraska Medical Center, USA
| | - Chi Lin
- Department of Radiation Oncology, University of Nebraska Medical Center, USA
| |
Collapse
|
31
|
Orhan A, Vogelsang RP, Andersen MB, Madsen MT, Hölmich ER, Raskov H, Gögenur I. The prognostic value of tumour-infiltrating lymphocytes in pancreatic cancer: a systematic review and meta-analysis. Eur J Cancer 2020; 132:71-84. [PMID: 32334338 DOI: 10.1016/j.ejca.2020.03.013] [Citation(s) in RCA: 102] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2019] [Revised: 03/08/2020] [Accepted: 03/19/2020] [Indexed: 12/15/2022]
Abstract
IMPORTANCE Tumour-infiltrating lymphocytes (TILs) have previously been found to influence patient prognosis in other gastrointestinal cancers, for instance in colorectal cancer. An immunosuppressive phenotype often characterizes pancreatic cancer with a low degree of immune cell infiltration. Cytotoxic CD8+ T cell infiltration in tumours is found to be the best predictive variable for response to immune checkpoint inhibitor therapy, emphasizing the importance of investigating TILs in pancreatic cancer, especially focussing on CD8+ T cells. OBJECTIVE Here, we systematically review the literature and perform meta-analyses to examine the prognostic value of TILs in human pancreatic ductal adenocarcinomas (PDAC). Secondarily, we review the literature regarding the histological localization of TILs and the impact on survival in PDAC. EVIDENCE REVIEW A literature search was conducted on PubMed, Embase, The Cochrane Library and Web of Science. Studies examining patients with PDAC and the impact of high vs. low infiltration of immune cells on long-term oncological survival measures were included. Time-to-event meta-analysis and frequency analysis were conducted using a random effects model. The risk of bias was assessed using the Newcastle-Ottowa Scale. Quality of the cumulative evidence was evaluated using the GRADE approach for prognostic studies. FINDINGS In total, 1971 articles were screened, of which 43 studies were included in the systematic review and 39 in the meta-analysis. High infiltration of CD8+ lymphocytes was significantly associated with improved overall survival (OS) [hazard ratio (HR) = 0.58, 95% confidence intervals (CIs): 0.50-0.68], disease-free survival (DFS) [HR = 0.64, 95% CI: 0.52-0.78], progression-free survival [HR = 0.66, 95% CI: 0.51-0.86] and cancer-specific survival [HR = 0.56, 95% CI: 0.32-0.99]. A high infiltration of CD3+ T cells was correlated with increased OS [HR = 0.58, 95% CI: 0.50-0.68] and DFS [HR = 0.74, 95% CI: 0.38-1.43]. Infiltration of CD4+ lymphocytes was associated with improved 12-months OS [risk ratio = 0.59, 95% CI: 0.35-0.99] and DFS [risk ratio = 0.68, 95% CI: 0.53-0.88]. High expression of FoxP3+ lymphocytes was associated with poor OS [HR = 1.48, 95% CI: 1.20-1.83]. The greatest impact on survival was observed in the CD8+ T cell and OS group, when infiltration was located to the tumour centre [HR = 0.53, 95% CI: 0.45-0.63]. However, subgroup analysis on the impact of the histological location of infiltration revealed no significant differences between the subgroups (tumour centre, invasive margin, stroma and all locations) in any of the examined cell types and outcomes. CONCLUSIONS AND RELEVANCE Subsets of TILs, especially CD3+, CD8+ and FoxP3+ T cells are strongly associated with long-term oncological outcomes in patients with PDAC. To our knowledge, this is the first systematic review and meta-analysis on the prognostic value of TILs in pancreatic cancer.
Collapse
Affiliation(s)
- Adile Orhan
- Center for Surgical Science, Zealand University Hospital, Koege, Denmark; Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark.
| | - Rasmus P Vogelsang
- Center for Surgical Science, Zealand University Hospital, Koege, Denmark
| | - Malene B Andersen
- Center for Surgical Science, Zealand University Hospital, Koege, Denmark
| | - Michael T Madsen
- Center for Surgical Science, Zealand University Hospital, Koege, Denmark
| | - Emma R Hölmich
- Center for Surgical Science, Zealand University Hospital, Koege, Denmark
| | - Hans Raskov
- Center for Surgical Science, Zealand University Hospital, Koege, Denmark
| | - Ismail Gögenur
- Center for Surgical Science, Zealand University Hospital, Koege, Denmark; Institute for Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
32
|
Hwang HK, Lee SH, Kim HI, Kim SH, Choi J, Kang CM, Lee WJ. Yonsei Criteria, a Potential Linkage to Intratumoral Foxp3⁺/CD8⁺ Ratio for the Prediction of Oncologic Outcomes in Resected Left-Sided Pancreatic Cancer. Yonsei Med J 2020; 61:291-300. [PMID: 32233171 PMCID: PMC7105403 DOI: 10.3349/ymj.2020.61.4.291] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 03/03/2020] [Accepted: 03/03/2020] [Indexed: 11/27/2022] Open
Abstract
PURPOSE This study sought to investigate associations among Yonsei criteria (tumor confined to the pancreas, intact fascia layer between the distal pancreas and the left adrenal gland and kidney, and tumor located more than 1-2 cm from the celiac axis) and tumor infiltrating lymphocytes in pancreatic cancer. MATERIALS AND METHODS Patients who underwent curative distal pancreatectomy due to left-sided pancreatic cancer from January 2000 to December 2011 were enrolled. Follow-up was completed September 30, 2015. RESULTS Fifty patients were enrolled. Having ≥ two metastatic lymph nodes (LNs, p=0.002), intraoperative transfusion (p=0.011), low levels of tumor infiltrating CD8⁺ T-cells (p=0.001), and a high Foxp3⁺/CD8⁺ ratio (p=0.009) were independent risk factors for disease-free survival. Not satisfying the Yonsei criteria (p=0.021), having ≥ two metastatic LNs (p=0.032), low levels of tumor infiltrating CD8⁺ T-cells (p=0.040) and a high Foxp3⁺/CD8⁺ ratio (p=0.032) were associated with unfavorable overall survival. High levels of CA19-9 and not satisfying the Yonsei criteria were significantly associated with a high Foxp3⁺/CD8⁺ ratio [Exp(β)=3.558; 95% confidence inverval: 1.000-12.658; p=0.050]. CONCLUSION Yonsei criteria may be clinically detectable biologic marker with which to predict immunologic status and survival in pancreatic cancer patients.
Collapse
Affiliation(s)
- Ho Kyoung Hwang
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, Yonsei University College of Medicine, Seoul, Korea
- Pancreatobiliary Cancer Center, Yonsei Cancer Center, Severance Hospital, Seoul, Korea
| | - Sung Hwan Lee
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam, Korea
| | - Hyoung Il Kim
- Department of Gastrointestinal Surgery, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Se Hoon Kim
- Department of Pathology, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Junjeong Choi
- Department of Pharmacy, Yonsei University College of Pharmacy, Seoul, Korea
| | - Chang Moo Kang
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, Yonsei University College of Medicine, Seoul, Korea
- Pancreatobiliary Cancer Center, Yonsei Cancer Center, Severance Hospital, Seoul, Korea.
| | - Woo Jung Lee
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, Yonsei University College of Medicine, Seoul, Korea
- Pancreatobiliary Cancer Center, Yonsei Cancer Center, Severance Hospital, Seoul, Korea
| |
Collapse
|
33
|
HHLA2 is expressed in pancreatic and ampullary cancers and increased expression is associated with better post-surgical prognosis. Br J Cancer 2020; 122:1211-1218. [PMID: 32071413 PMCID: PMC7156757 DOI: 10.1038/s41416-020-0755-4] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Accepted: 01/31/2020] [Indexed: 12/21/2022] Open
Abstract
Background HHLA2 is a recently discovered member of the B7-family of immune checkpoint molecules with limited expression in normal tissues but overexpression in several types of cancer. The aim was to determine the expression, prevalence and biological relevance of HHLA2 protein expression in two closely related human cancer types, namely pancreatic cancer and ampullary cancer. Methods HHLA2 expression levels were retrospectively determined by immunohistochemistry in tissue micro-arrays of surgically resected tumours of 122 pancreatic cancer patients and 72 patients with ampullary cancer of the pancreato-biliary subtype. Results HHLA2 was expressed at variable levels by tumour cells in 67% of pancreatic tumours and 93% of ampullary tumours. In the combined cohort high tumoural HHLA2 expression levels were significantly associated with delayed cancer recurrence and improved post-operative cancer-specific survival. The association of HHLA2 expression with cancer-specific survival and recurrence was statistically significant for the pancreatic cancer subgroup while a similar trend was found for the ampullary cancer subgroup. In multivariable analysis together with clinicopathologic characteristics, higher HHLA2 expression was an independent predictor of cancer-specific survival. Conclusion The wide expression of HHLA2 in tumour cells and its association with cancer recurrence and patient survival suggest that HHLA2 represents a relevant immune checkpoint molecule in pancreatic and ampullary cancers.
Collapse
|
34
|
Liu S, Zhang W, Liu K, Wang Y. CD160 expression on CD8 + T cells is associated with active effector responses but limited activation potential in pancreatic cancer. Cancer Immunol Immunother 2020; 69:789-797. [PMID: 32055919 DOI: 10.1007/s00262-020-02500-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Accepted: 01/23/2020] [Indexed: 12/22/2022]
Abstract
CD160 is an Ig-like glycoprotein expressed by the majority of circulating natural killer cells and γδ T cells. Whether CD160 could regulate CD8+ T-cell functions remains unknown. In this study, we investigated the effects of CD160 on CD8+ T cells in pancreatic cancer. First, we found that the frequency of PD-1+ cells was comparable between CD160+ and CD160-CD8+ T cells, with the former presenting significantly higher PD-1 expression level. In contrast, the frequency of TIM-3+ cells was higher among CD160+ cells but the expression level was comparable between CD160+ and CD160-CD8+ T cells. The IFN-γ and IL-2-expressing CD8+ T cells, directly ex vivo, were highly enriched in the CD160+ subset. However, when CD160+ and CD160-CD8+ T cells were stimulated, the proliferation levels of CD160+ and CD160- cells were initially comparable, but were significantly lower in CD160+CD8+ T cells than in CD160-CD8+ T cells later on. The IFN-γ and IL-2 transcription levels were initially higher in CD160+CD8+ T cells, but eventually reduced in CD160+CD8+ T cells compared to CD160-CD8+ T cells. Also, CD160+CD8+ T cells presented lower cytotoxic capacity than CD160-CD8+ T cells. Interestingly, we observed that tumor-infiltrating CD8+ T cells were significantly enriched with the CD160+ subset in pancreatic cancer patients. In addition, patients with higher frequencies of tumor CD160+CD8+ T cells presented lower survival. Overall, these data demonstrated that tumor-infiltrating CD8+ T cells were enriched with the CD160+ subset in pancreatic cancer, with active effector responses directly ex vivo but limited potential for further activation.
Collapse
Affiliation(s)
- Songyang Liu
- Department of Hepatobiliary and Pancreatic Surgery, The First Hospital of Jilin University, 71 Xinmin Avenue, Changchun, 130021, Jilin, China
| | - Wei Zhang
- Department of Hepatobiliary and Pancreatic Surgery, The First Hospital of Jilin University, 71 Xinmin Avenue, Changchun, 130021, Jilin, China
| | - Kai Liu
- Department of Hepatobiliary and Pancreatic Surgery, The First Hospital of Jilin University, 71 Xinmin Avenue, Changchun, 130021, Jilin, China
| | - Yingchao Wang
- Department of Hepatobiliary and Pancreatic Surgery, The First Hospital of Jilin University, 71 Xinmin Avenue, Changchun, 130021, Jilin, China.
| |
Collapse
|
35
|
Prognostic implications of immune classification in a multicentre cohort of patients with small intestinal adenocarcinoma. Pathology 2020; 52:228-235. [DOI: 10.1016/j.pathol.2019.09.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2019] [Revised: 09/08/2019] [Accepted: 09/15/2019] [Indexed: 01/03/2023]
|
36
|
Liao H, Zhang Z, Chen J, Liao M, Xu L, Wu Z, Yuan K, Song B, Zeng Y. Preoperative Radiomic Approach to Evaluate Tumor-Infiltrating CD8 + T Cells in Hepatocellular Carcinoma Patients Using Contrast-Enhanced Computed Tomography. Ann Surg Oncol 2019; 26:4537-4547. [PMID: 31520208 DOI: 10.1245/s10434-019-07815-9] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Indexed: 02/05/2023]
Abstract
BACKGROUND To help identify potential hepatocellular carcinoma (HCC) candidates for immunotherapies, we aimed to develop and validate a radiomics-based biomarker (Rad score) to predict the infiltration of tumor-infiltrating CD8+ T cells in HCC patients, and to evaluate the correlation of Rad score with tumor immune characteristics. METHODS Overall, 142 HCC patients (n = 100 and n = 42 in the training and validation sets, respectively) were subjected to radiomic feature extraction. Imaging features and immunochemistry data of patients in the training set were subjected to elastic-net regularized regression analysis to predict the level of CD8+ T cell infiltration. RESULTS A Rad score for CD8+ T-cell infiltration, which contained seven variables, was developed and was validated in the validation set (area under the curve [AUC]: training set 0.751, 95% confidence interval [CI] 0.656-0.846; validation set 0.705, 95% CI 0.547-0.863). The decision curve indicated the clinical usefulness of the Rad score. A higher Rad score correlated with superior overall and disease-free survival outcomes (p = 0.012 and 0.0088, respectively). Using the pathological slides, we found that the Rad score positively correlated with the percentage of tumor-infiltrating lymphocytes (TILs; Spearman rho = 0.51, p < 0.0001). Moreover, the Rad score could also discriminate inflamed tumors from immune-desert and immune-excluded tumors (Kruskal-Wallis, p < 0.0001), and higher Rad scores could be found in patients with positive programmed cell death ligand 1 expression in tumor/immune cells, as well as those with positive programmed cell death protein 1 expression. CONCLUSION The newly developed Rad score was a powerful predictor of CD8+ T-cell infiltration, which could be useful in identifying potential HCC patients who can benefit from immunotherapies when validated in large-scale prospective cohorts.
Collapse
Affiliation(s)
- Haotian Liao
- Department of Liver Surgery, Liver Transplantation Division, West China Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China
- Laboratory of Liver Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Zhen Zhang
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Jie Chen
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Mingheng Liao
- Department of Liver Surgery, Liver Transplantation Division, West China Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China
- Laboratory of Liver Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Lin Xu
- Department of Liver Surgery, Liver Transplantation Division, West China Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China
- Laboratory of Liver Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Zhenru Wu
- Laboratory of Pathology, Department of Pathology, West China Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Kefei Yuan
- Department of Liver Surgery, Liver Transplantation Division, West China Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China
- Laboratory of Liver Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Bin Song
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China.
| | - Yong Zeng
- Department of Liver Surgery, Liver Transplantation Division, West China Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China.
- Laboratory of Liver Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China.
| |
Collapse
|
37
|
Sun Q, Zhang Y, Liu M, Ye Z, Yu X, Xu X, Qin Y. Prognostic and diagnostic significance of galectins in pancreatic cancer: a systematic review and meta-analysis. Cancer Cell Int 2019; 19:309. [PMID: 31832021 PMCID: PMC6873495 DOI: 10.1186/s12935-019-1025-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Accepted: 11/11/2019] [Indexed: 02/06/2023] Open
Abstract
Background Galectins constitute a family of β-galactoside-binding proteins, which influence various hallmarks of pancreatic cancer, including cell proliferation, invasion and migration; immune escape; and angiogenesis. Although many studies have concentrated on the role of galectins in pancreatic cancer, the results remain controversial. Hence, we performed a comprehensive meta-analysis to clarify the precise diagnostic and prognostic value of galectins in pancreatic cancer. Methods PubMed/MEDLINE, EMBASE and Web of Science were used to search related published literature up to July 2019. Pooled hazard ratios (HRs), diagnostic accuracy variables and related 95% confidence intervals (CIs) were calculated using STATA 14.0 software. Results Eleven studies including 1227 participants met our inclusion criteria. High expression of galectin family was not correlated with overall survival (OS) in pancreatic cancer (HR, 1.19; 95% CI 0.67-2.11). According to subgroup analysis, high levels of galectin-1 were significantly correlated with worse OS in pancreatic cancer (HR, 4.77; 95% CI 2.47-9.21), while high levels of tandem-repeat galectins (galectin-4 or galectin-9) predicted both better OS (HR, 0.63; 95% CI 0.46-0.86) and disease-free survival (DFS) (HR, 0.63; 95% CI 0.48-0.83). The expression levels of galectin-3 did not directly correlate with prognosis (HR, 0.99; 95% CI 0.40-2.46). The pooled sensitivity, specificity, positive likelihood ratio, and negative likelihood ratios of galectin-3 were 0.64 (95% CI 0.41-0.82), 0.76 (95% CI 0.59-0.88), 2.70 (95% CI 1.21-6.1), and 0.47 (95% CI 0.23-0.98), respectively. The area under the curve (AUC) of galectin-3 was 0.77. Conclusion Taken together, our results suggest that high expression of galectin-1 and low levels of galectin-4 or galectin-9 are predictors of worse prognosis in pancreatic cancer patients. The expression of galectin-3 was not directly related to OS and other clinical characteristics. Although galectin-3 exhibited some diagnostic value in patients with pancreatic cancer in this meta-analysis, clinical application prospects remain to be validated. Further studies are warranted to confirm and strengthen these findings.
Collapse
Affiliation(s)
- Qiqing Sun
- 1Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032 China.,2Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032 China.,3Pancreatic Cancer Institute, Fudan University, Shanghai, 200032 China.,4Shanghai Pancreatic Cancer Institute, Shanghai, 200032 China
| | - Yiyin Zhang
- 1Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032 China.,2Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032 China.,3Pancreatic Cancer Institute, Fudan University, Shanghai, 200032 China.,4Shanghai Pancreatic Cancer Institute, Shanghai, 200032 China
| | - Mengqi Liu
- 1Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032 China.,2Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032 China.,3Pancreatic Cancer Institute, Fudan University, Shanghai, 200032 China.,4Shanghai Pancreatic Cancer Institute, Shanghai, 200032 China
| | - Zeng Ye
- 1Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032 China.,2Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032 China.,3Pancreatic Cancer Institute, Fudan University, Shanghai, 200032 China.,4Shanghai Pancreatic Cancer Institute, Shanghai, 200032 China
| | - Xianjun Yu
- 1Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032 China.,2Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032 China.,3Pancreatic Cancer Institute, Fudan University, Shanghai, 200032 China.,4Shanghai Pancreatic Cancer Institute, Shanghai, 200032 China
| | - Xiaowu Xu
- 1Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032 China.,2Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032 China.,3Pancreatic Cancer Institute, Fudan University, Shanghai, 200032 China.,4Shanghai Pancreatic Cancer Institute, Shanghai, 200032 China
| | - Yi Qin
- 1Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032 China.,2Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032 China.,3Pancreatic Cancer Institute, Fudan University, Shanghai, 200032 China.,4Shanghai Pancreatic Cancer Institute, Shanghai, 200032 China
| |
Collapse
|
38
|
Bertol BC, Dias FC, da Silva DM, Zambelli Ramalho LN, Donadi EA. Human Antigen Leucocyte (HLA)-G and HLA-E are differentially expressed in pancreatic disorders. Hum Immunol 2019; 80:948-954. [PMID: 31561913 DOI: 10.1016/j.humimm.2019.09.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 08/28/2019] [Accepted: 09/03/2019] [Indexed: 01/07/2023]
Abstract
BACKGROUND Little information is available regarding the expression of the immunomodulatory Human Leukocyte Antigen (HLA)-G and -E molecules in pancreatic disorders. AIM To analyze HLA-G and -E expression in specimens of alcoholic chronic pancreatitis (ACP), idiopathic chronic pancreatitis (ICP), type 1 (T1D) and type 2 diabetes (T2D) and in histologically normal pancreas (HNP). METHODS HLA-G and -E expression (ACP = 30, ICP = 10, T1D = 10, T2D = 30 and HNP = 20) was evaluated by immunohistochemistry in three different areas (acini, islets and inflammatory infiltrate). RESULTS Acini and islets from HNP specimens exhibited higher HLA-G and -E expression compared to corresponding areas from all other patient groups. In inflammatory infiltrate, HLA-G and -E expression was observed only among the pancreatic disorders. We observed higher HLA-G and -E expression in acini from T2D compared to ACP, as well as higher HLA-G expression compared to ICP. CONCLUSION The decreased expression of HLA-G and -E in islets and acini together with the expression of these molecules in the inflammatory infiltrating cells were shared features among chronic inflammatory and autoimmune pancreatic disorders evaluated in this study, possibly reflecting tissue damage.
Collapse
Affiliation(s)
- Bruna Cristina Bertol
- Postgraduate Program of Basic and Applied Immunology, Ribeirão Preto Medical School, University of São Paulo, 3900 Bandeirantes Avenue, ZIP Code: 14049-900 Ribeirão Preto, Brazil.
| | - Fabrício César Dias
- Department of Medicine, Division of Clinical Immunology, Ribeirão Preto Medical School, University of São Paulo, 3900 Bandeirantes Avenue, ZIP Code: 14049-900 Ribeirão Preto, Brazil.
| | - Deisy Mara da Silva
- Department of Pathology, Ribeirão Preto Medical School, University of São Paulo, 3900 Bandeirantes Avenue, ZIP Code: 14049-900 Ribeirão Preto, Brazil.
| | - Leandra Náira Zambelli Ramalho
- Department of Pathology, Ribeirão Preto Medical School, University of São Paulo, 3900 Bandeirantes Avenue, ZIP Code: 14049-900 Ribeirão Preto, Brazil.
| | - Eduardo Antônio Donadi
- Postgraduate Program of Basic and Applied Immunology, Ribeirão Preto Medical School, University of São Paulo, 3900 Bandeirantes Avenue, ZIP Code: 14049-900 Ribeirão Preto, Brazil; Department of Medicine, Division of Clinical Immunology, Ribeirão Preto Medical School, University of São Paulo, 3900 Bandeirantes Avenue, ZIP Code: 14049-900 Ribeirão Preto, Brazil.
| |
Collapse
|
39
|
Ferreira CR, Zhao S, Sanches JA, Miyashiro D, Cury-Martins J, Azevedo RS, Zerbini MCN, Natkunam Y, Gratzinger D. Clinicopathologic and microenvironmental analysis of primary cutaneous CD30-positive lymphoproliferative disorders: a 26 year experience from an academic medical center in Brazil. Diagn Pathol 2019; 14:115. [PMID: 31640798 PMCID: PMC6805531 DOI: 10.1186/s13000-019-0900-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2019] [Accepted: 10/09/2019] [Indexed: 12/13/2022] Open
Abstract
Background Primary cutaneous CD30+ lymphoproliferative disorders (pc-CD30-LPD) are a group of clonal T cell lymphoproliferative disorders that despite very similar tumor histology follow different and characteristic clinical courses, suggesting a homeostatic role of the tumor microenvironment. Little is known about tumor microenvironment and there is almost no literature about PD-L1 expression in pc-CD30-LPD. Methods This retrospective study presents a fully clinicopathologically characterized series of pc-CD30-LPDs from an academic medical center in Brazil, including 8 lymphomatoid papulomatosis (LyP), 9 primary cutaneous anaplastic large cell lymphoma (pcALCL) and 4 borderline lesions. All the cases were scored for FOXP3+ regulatory T-cells (Treg) and CD8+ cytotoxic tumor infiltrating lymphocytes (TIL) densities, as well as PD-L1 expression in tumor cells and tissue associated macrophages. The CD8+/FOXP3+ ratio was also evaluated. Results Among the 21 cases of pc-CD30-LPD, PD-L1 expression is frequent in both tumor cells and tissue associated macrophages in pc-CD30-LPD across categories, suggesting that the PD-L1 axis may be a common feature of pc-CD30-LPDs. While reactive T cell infiltrates vary widely from case to case, a common feature across pc-CD30-LPDs is higher density of CD8 than FOXP3 + T cells. The distribution of T cells within the lesions however differed between LyP and pcALCL: we found that LyP lesions tend to be permeated by CD8+ and FOXP3+ T cells, whereas pcALCL tend to be surrounded by a rim of CD8+ TIL and FOXP3+ Tregs with relatively lower density infiltrates in the center of the lesion. Conclusions LyP has a trend to have denser immune cells throughout the lesion, with higher FOXP3+ Treg and CD8+ TIL in the center than the edge comparing with pcALCL. PD-L1+ is frequent in tumor cells and tissue associated macrophages in pc-CD30-LPD. The differential distribution of CD8+ and FOXP3+ TILs in LyP as compared to pcALCL could provide a clue to the relapsing/remitting course of LyP as compared to the less frequent spontaneous regression of pcALCL.
Collapse
Affiliation(s)
- Cristiane Rúbia Ferreira
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, 94305, USA. .,Departments of Pathology, University of Sao Paulo, Sao Paulo, SP, 01246, Brazil. .,Anatomical Pathology Service, University Hospital of Sao Paulo University (HU-USP), Rua Professor Lineu Prestes, 2565, Sao Paulo, SP, 05508-000, Brazil.
| | - Shuchun Zhao
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - José Antonio Sanches
- Departments of Dermatology, University of Sao Paulo, Sao Paulo, SP, 01246, Brazil
| | - Denis Miyashiro
- Departments of Dermatology, University of Sao Paulo, Sao Paulo, SP, 01246, Brazil
| | - Jade Cury-Martins
- Departments of Dermatology, University of Sao Paulo, Sao Paulo, SP, 01246, Brazil
| | | | - Maria C N Zerbini
- Departments of Pathology, University of Sao Paulo, Sao Paulo, SP, 01246, Brazil
| | - Yasodha Natkunam
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Dita Gratzinger
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| |
Collapse
|
40
|
Malissen N, Macagno N, Granjeaud S, Granier C, Moutardier V, Gaudy-Marqueste C, Habel N, Mandavit M, Guillot B, Pasero C, Tartour E, Ballotti R, Grob JJ, Olive D. HVEM has a broader expression than PD-L1 and constitutes a negative prognostic marker and potential treatment target for melanoma. Oncoimmunology 2019; 8:e1665976. [PMID: 31741766 PMCID: PMC6844309 DOI: 10.1080/2162402x.2019.1665976] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 09/05/2019] [Accepted: 09/06/2019] [Indexed: 10/27/2022] Open
Abstract
HVEM (Herpes Virus Entry Mediator) engagement of BTLA (B and T Lymphocyte Attenuator) triggers inhibitory signals in T cells and could play a role in evading antitumor immunity. Here, HVEM expression levels in melanoma metastases were analyzed by immunohistochemistry, correlated with overall survival (OS) in 116 patients, and validated by TCGA transcriptomic data. Coincident expression of HVEM and its ligand BTLA was studied in tumor cells and tumor-infiltrating lymphocytes (TILs) by flow cytometry (n = 21) and immunofluorescence (n = 5). Candidate genes controlling HVEM expression in melanoma were defined by bioinformatics studies and validated by siRNA gene silencing. We found that in patients with AJCC stage III and IV melanoma, OS was poorer in those with high HVEM expression on melanoma cells, than in those with a low expression, by immunohistochemistry (p = .0160) or TCGA transcriptomics (p = .0282). We showed a coincident expression of HVEM at the surface of melanoma cells and of BTLA on TILs. HVEM was more widely expressed than PD-L1 in melanoma cells. From a mechanistic perspective, in contrast to PDL1, HVEM expression did not correlate with an IFNγ signature but with an aggressive gene signature. Interestingly, this signature contained MITF, a key player in melanoma biology, whose expression correlated strongly with HVEM. Finally, siRNA gene silencing validated MITF control of HVEM expression. In conclusion, HVEM expression seems to be a prognosis marker and targeting this axis by checkpoint-inhibitors may be of interest in metastatic melanoma.
Collapse
Affiliation(s)
- Nausicaa Malissen
- Tumor Immunology Team, IBISA Immunomonitoring platform, Cancer Research Center of Marseille, INSERM U1068, CNRS U7258, Aix-Marseille University, Institut Paoli-Calmettes, Marseille, France.,INSERM, CRCM, APHM, CHU Timone, Department of Dermatology and Skin Cancer, Aix Marseille University, Marseille, France
| | - Nicolas Macagno
- INSERM, MMG, APHM, CHU Timone, Department of Pathology, Aix Marseille University, Marseille, France
| | - Samuel Granjeaud
- Tumor Immunology Team, IBISA Immunomonitoring platform, Cancer Research Center of Marseille, INSERM U1068, CNRS U7258, Aix-Marseille University, Institut Paoli-Calmettes, Marseille, France
| | - Clémence Granier
- UMR_S970, HEGP, Centre de recherche cardio-vasculaire, Paris, France
| | - Vincent Moutardier
- APHM, CHU Nord, Department of Digestive surgery, Aix Marseille University, Marseille, France
| | - Caroline Gaudy-Marqueste
- Tumor Immunology Team, IBISA Immunomonitoring platform, Cancer Research Center of Marseille, INSERM U1068, CNRS U7258, Aix-Marseille University, Institut Paoli-Calmettes, Marseille, France.,INSERM, CRCM, APHM, CHU Timone, Department of Dermatology and Skin Cancer, Aix Marseille University, Marseille, France
| | - Nadia Habel
- INSERM U 1065, Team 1 Nice, Centre Méditerranéen de Médecine Moléculaire, Nice, France
| | - Marion Mandavit
- UMR_S970, HEGP, Centre de recherche cardio-vasculaire, Paris, France
| | - Bernard Guillot
- Department of Dermatology, CHU Montpellier, Montpellier, France
| | - Christine Pasero
- Tumor Immunology Team, IBISA Immunomonitoring platform, Cancer Research Center of Marseille, INSERM U1068, CNRS U7258, Aix-Marseille University, Institut Paoli-Calmettes, Marseille, France
| | - Eric Tartour
- UMR_S970, HEGP, Centre de recherche cardio-vasculaire, Paris, France
| | - Robert Ballotti
- INSERM U 1065, Team 1 Nice, Centre Méditerranéen de Médecine Moléculaire, Nice, France
| | - Jean-Jacques Grob
- Tumor Immunology Team, IBISA Immunomonitoring platform, Cancer Research Center of Marseille, INSERM U1068, CNRS U7258, Aix-Marseille University, Institut Paoli-Calmettes, Marseille, France.,INSERM, CRCM, APHM, CHU Timone, Department of Dermatology and Skin Cancer, Aix Marseille University, Marseille, France
| | - Daniel Olive
- Tumor Immunology Team, IBISA Immunomonitoring platform, Cancer Research Center of Marseille, INSERM U1068, CNRS U7258, Aix-Marseille University, Institut Paoli-Calmettes, Marseille, France
| |
Collapse
|
41
|
Yoshida S, Ito Z, Suka M, Bito T, Kan S, Akasu T, Saruta M, Okamoto M, Kitamura H, Fujioka S, Misawa T, Akiba T, Yanagisawa H, Sugiyama H, Koido S. Clinical Significance of Tumor-Infiltrating T Cells and Programed Death Ligand-1 in Patients with Pancreatic Cancer. Cancer Invest 2019; 37:463-477. [PMID: 31490702 DOI: 10.1080/07357907.2019.1661427] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The associations of the immunological status of the pancreatic ductal adenocarcinoma (PDA) microenvironment with prognosis were assessed. A high tumor-infiltrating lymphocyte (TIL) density was associated with a better prognosis. Importantly, even with a high density of TILs, the PDA cells with programed cell death-ligand 1 (PD-L1) expression showed a worse prognosis than the patients with negative PD-L1 expression. A significant association between a better prognosis and a tumor microenvironment with a high TIL density/negative PD-L1 expression was observed. Assessments of a combined immunological status in the tumor microenvironment may predict the prognosis of PDA patients following surgical resection.
Collapse
Affiliation(s)
- Sayumi Yoshida
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, The Jikei University School of Medicine, Kashiwa Hospital , Chiba , Japan
| | - Zensho Ito
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, The Jikei University School of Medicine, Kashiwa Hospital , Chiba , Japan
| | - Machi Suka
- Department of Public Health and Environmental Medicine, The Jikei University School of Medicine , Tokyo , Japan
| | - Tsuuse Bito
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, The Jikei University School of Medicine, Kashiwa Hospital , Chiba , Japan.,Institute of Clinical Medicine and Research, The Jikei University School of Medicine , Chiba , Japan
| | - Shin Kan
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, The Jikei University School of Medicine, Kashiwa Hospital , Chiba , Japan.,Institute of Clinical Medicine and Research, The Jikei University School of Medicine , Chiba , Japan
| | - Takafumi Akasu
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, The Jikei University School of Medicine, Kashiwa Hospital , Chiba , Japan
| | - Masayuki Saruta
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, The Jikei University School of Medicine , Tokyo , Japan
| | - Masato Okamoto
- Department of Oral Medicine and Stomatology, Tsurumi University School of Dental Medicine , Kanagawa , Japan
| | - Hiroaki Kitamura
- Department of Surgery, The Jikei University School of Medicine, Kashiwa Hospital , Chiba , Japan
| | - Shuichi Fujioka
- Department of Surgery, The Jikei University School of Medicine, Kashiwa Hospital , Chiba , Japan
| | - Takeyuki Misawa
- Department of Surgery, The Jikei University School of Medicine, Kashiwa Hospital , Chiba , Japan
| | - Tadashi Akiba
- Department of Surgery, The Jikei University School of Medicine, Kashiwa Hospital , Chiba , Japan
| | - Hiroyuki Yanagisawa
- Department of Public Health and Environmental Medicine, The Jikei University School of Medicine , Tokyo , Japan
| | - Haruo Sugiyama
- Department of Functional Diagnostic Science, Osaka University Graduate School of Medicine , Osaka , Japan
| | - Shigeo Koido
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, The Jikei University School of Medicine, Kashiwa Hospital , Chiba , Japan.,Institute of Clinical Medicine and Research, The Jikei University School of Medicine , Chiba , Japan
| |
Collapse
|
42
|
Zhang Y, Xu J, Hua J, Liu J, Liang C, Meng Q, Wei M, Zhang B, Yu X, Shi S. A PD-L2-based immune marker signature helps to predict survival in resected pancreatic ductal adenocarcinoma. J Immunother Cancer 2019; 7:233. [PMID: 31464648 PMCID: PMC6716876 DOI: 10.1186/s40425-019-0703-0] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2019] [Accepted: 07/31/2019] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Programmed cell death protein 1 (PD-1) is a key immune checkpoint that regulates peripheral tolerance and protects against autoimmunity. Programmed death ligand-2 (PD-L2) is a less studied ligand to PD-1 and has yet to be fully explored, especially in pancreatic ductal adenocarcinoma (PDAC). METHODS In this study, we performed immunohistochemistry to detect the PD-L2, CD3, CD8, transforming growth factor-β2 (TGF-β2) and FOXP3 levels in paraffin sections from 305 patients with resected PDAC as a training set. Expression levels of intratumoral and stromal immune markers were compared in relation to survival using Kaplan-Meier curves, random survival forest model and survival tree analysis. A multivariable Cox proportional-hazards model of associated markers was used to calculate the risk scores. RESULTS PD-L2 was expressed in 71.5% of PDAC samples and showed strong correlations with CD3+, CD8+ T cells and FOXP3+ regulatory T cell densities. High levels of intratumoral PD-L2 and FOXP3 were related to poor survival; only stromal FOXP3 overexpression was associated with worse prognosis. Four patterns generated from survival tree analysis demonstrated that PD-L2lowstromalFOXP3low patients had the longest survival, while PD-L2highintratumoralCD3low patients had the shortest survival (P < 0.001). The area under the curve was 0.631(95% confidence interval (CI): 0.447-0.826) for the immune marker-based signature and 0.549 (95% CI: 0.323-0.829; P < 0.001) for the clinical parameter-based signature, which was consistent with the results in the validation set including 150 patients (P < 0.001). A higher risk score indicated shorter survival and could serve as an independent prognostic factor. PD-L2 was also showed associated with TGF-β2 and other immune molecules based on bioinformatics analysis. CONCLUSIONS Our work highlighted PD-L2 as a promising immunotherapeutic target with prognostic value combined with complex tumor infiltrating cells in PDAC.
Collapse
Affiliation(s)
- Yiyin Zhang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology Shanghai Medical College, Fudan University, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
- Shanghai Pancreatic Cancer Institute, Shanghai, China
| | - Jin Xu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology Shanghai Medical College, Fudan University, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
- Shanghai Pancreatic Cancer Institute, Shanghai, China
| | - Jie Hua
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology Shanghai Medical College, Fudan University, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
- Shanghai Pancreatic Cancer Institute, Shanghai, China
| | - Jiang Liu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology Shanghai Medical College, Fudan University, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
- Shanghai Pancreatic Cancer Institute, Shanghai, China
| | - Chen Liang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology Shanghai Medical College, Fudan University, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
- Shanghai Pancreatic Cancer Institute, Shanghai, China
| | - Qingcai Meng
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology Shanghai Medical College, Fudan University, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
- Shanghai Pancreatic Cancer Institute, Shanghai, China
| | - Miaoyan Wei
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology Shanghai Medical College, Fudan University, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
- Shanghai Pancreatic Cancer Institute, Shanghai, China
| | - Bo Zhang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology Shanghai Medical College, Fudan University, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
- Shanghai Pancreatic Cancer Institute, Shanghai, China
| | - Xianjun Yu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.
- Department of Oncology Shanghai Medical College, Fudan University, Shanghai, China.
- Pancreatic Cancer Institute, Fudan University, Shanghai, China.
- Shanghai Pancreatic Cancer Institute, Shanghai, China.
| | - Si Shi
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.
- Department of Oncology Shanghai Medical College, Fudan University, Shanghai, China.
- Pancreatic Cancer Institute, Fudan University, Shanghai, China.
- Shanghai Pancreatic Cancer Institute, Shanghai, China.
| |
Collapse
|
43
|
IDO1 Expression Is Associated With Immune Tolerance and Poor Prognosis in Patients With Surgically Resected Esophageal Cancer. Ann Surg 2019; 269:1101-1108. [DOI: 10.1097/sla.0000000000002754] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
44
|
Danilova L, Ho WJ, Zhu Q, Vithayathil T, De Jesus-Acosta A, Azad NS, Laheru DA, Fertig EJ, Anders R, Jaffee EM, Yarchoan M. Programmed Cell Death Ligand-1 (PD-L1) and CD8 Expression Profiling Identify an Immunologic Subtype of Pancreatic Ductal Adenocarcinomas with Favorable Survival. Cancer Immunol Res 2019; 7:886-895. [PMID: 31043417 DOI: 10.1158/2326-6066.cir-18-0822] [Citation(s) in RCA: 181] [Impact Index Per Article: 30.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 02/04/2019] [Accepted: 04/23/2019] [Indexed: 12/11/2022]
Abstract
Immune-checkpoint therapy has failed to demonstrate meaningful clinical benefit in unselected cases of pancreatic adenocarcinoma (PDAC), but a subset of PDACs are known to upregulate pathways involved in acquired immune suppression. Further delineation of immunologic subtypes of PDAC is necessary to improve clinical trial designs and identify patients who might benefit from immune-checkpoint therapy. We used clinical survival and RNA expression data from The Cancer Genome Atlas (TCGA) to investigate the relationship between immune-modulating pathways and immune subset markers and their impact on survival in PDAC patients. Of the adaptive immune-resistance pathways, expression of PD-L1 and IDO1 was individually associated with poor survival. Although CD8 expression alone was not correlated with survival, the combination of PD-L1- and high CD8 expression identified a subtype with favorable survival. We further extended these observations using an independent PDAC cohort from our institution via IHC, again observing that the PD-L1-/CD8high subtype was associated with positive prognosis. Although PDAC is regarded as a poorly immunogenic cancer type, these findings infer that T-cell infiltration in the absence of adaptive immune-resistance pathways is a feature of long-term survival in PDAC and imply the importance of developing future immunotherapeutic strategies based on data-supported biomarkers to refine patient selection.
Collapse
Affiliation(s)
- Ludmila Danilova
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Won Jin Ho
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Qingfeng Zhu
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Teena Vithayathil
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Ana De Jesus-Acosta
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Nilofer S Azad
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Daniel A Laheru
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Elana J Fertig
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Robert Anders
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Elizabeth M Jaffee
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, Maryland.
| | - Mark Yarchoan
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, Maryland.
| |
Collapse
|
45
|
Huang L, Balavarca Y, van der Geest L, Lemmens V, Van Eycken L, De Schutter H, Johannesen TB, Zadnik V, Primic-Žakelj M, Mägi M, Grützmann R, Besselink MG, Schrotz-King P, Brenner H, Jansen L. Development and validation of a prognostic model to predict the prognosis of patients who underwent chemotherapy and resection of pancreatic adenocarcinoma: a large international population-based cohort study. BMC Med 2019; 17:66. [PMID: 30905320 PMCID: PMC6432746 DOI: 10.1186/s12916-019-1304-y] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Accepted: 03/11/2019] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Pancreatic cancer (PaC) remains extremely lethal worldwide even after resection. PaC resection rates are low, making prognostic studies in resected PaC difficult. This large international population-based study aimed at exploring factors associated with survival in patients with resected TNM stage I-II PaC receiving chemotherapy and at developing and internationally validating a survival-predicting model. METHODS Data of stage I-II PaC patients resected and receiving chemotherapy in 2003-2014 were obtained from the national cancer registries of Belgium, the Netherlands, Slovenia, and Norway, and the US Surveillance, Epidemiology, and End Results (SEER)-18 Program. Multivariable Cox proportional hazards models were constructed to investigate the associations of patient and tumor characteristics with overall survival, and analysis was performed in each country respectively without pooling. Prognostic factors remaining after backward selection in SEER-18 were used to build a nomogram, which was subjected to bootstrap internal validation and external validation using the European datasets. RESULTS A total of 11,837 resected PaC patients were analyzed, with median survival time of 18-23 months and 3-year survival rates of 21-31%. In the main analysis, patient age, tumor T stage, N stage, and differentiation were associated with survival across most countries, with country-specific association patterns and strengths. However, tumor location was mostly not significantly associated with survival. Resection margin, hospital type, tumor size, positive and harvested lymph node number, lymph node ratio, and comorbidity number were associated with survival in certain countries where the information was available. A median survival time- and 1-, 2-, 3-, and 5-year survival probability-predictive nomogram incorporating the backward-selected variables in the main analysis was established. It fits each European national cohort similarly well. Calibration curves showed very good agreement between nomogram-prediction and actual observation. The concordance index of the nomogram (0.60) was significantly higher than that of the T and N stage-based model (0.56) for predicting survival. CONCLUSIONS In these large international population-based cohorts, patients with resected PaC receiving chemotherapy have distinct characteristics independently associated with survival, with country-specific patterns and strengths. A robust benchmark population-based survival-predicting model is established and internationally validated. Like previous models predicting survival in resected PaC, our nomogram performs modestly.
Collapse
Affiliation(s)
- Lei Huang
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 581, 69120 Heidelberg, Germany
- Medical Faculty Heidelberg, Heidelberg University, Heidelberg, Germany
| | - Yesilda Balavarca
- Division of Preventive Oncology, German Cancer Research Center (DKFZ) and National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Lydia van der Geest
- Netherlands Cancer Registry (NCR), Netherlands Comprehensive Cancer Organization (IKNL), Utrecht, The Netherlands
| | - Valery Lemmens
- Netherlands Cancer Registry (NCR), Netherlands Comprehensive Cancer Organization (IKNL), Utrecht, The Netherlands
| | | | | | - Tom B. Johannesen
- Registry Department, The Cancer Registry of Norway (CRN), Oslo, Norway
| | - Vesna Zadnik
- Epidemiology and Cancer Registry, Institute of Oncology Ljubljana, Ljubljana, Slovenia
| | - Maja Primic-Žakelj
- Epidemiology and Cancer Registry, Institute of Oncology Ljubljana, Ljubljana, Slovenia
| | - Margit Mägi
- Estonian Cancer Registry, National Institute for Health Development, Tallinn, Estonia
| | - Robert Grützmann
- Department of Surgery, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Marc G. Besselink
- Dutch Pancreatic Cancer Group (DPCG), Department of Surgery, Cancer Center Amsterdam, Amsterdam University Medical Centers (UMC), Academic Medical Center (AMC), University of Amsterdam, Amsterdam, The Netherlands
| | - Petra Schrotz-King
- Division of Preventive Oncology, German Cancer Research Center (DKFZ) and National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Hermann Brenner
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 581, 69120 Heidelberg, Germany
- Division of Preventive Oncology, German Cancer Research Center (DKFZ) and National Center for Tumor Diseases (NCT), Heidelberg, Germany
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Lina Jansen
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 581, 69120 Heidelberg, Germany
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
46
|
Kim MH, Jang M, Kim H, Lee WJ, Kang CM, Choi HJ. Distinct immunological properties of the two histological subtypes of adenocarcinoma of the ampulla of Vater. Cancer Immunol Immunother 2019; 68:443-454. [PMID: 30604042 PMCID: PMC11028066 DOI: 10.1007/s00262-018-02293-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2018] [Accepted: 12/24/2018] [Indexed: 12/31/2022]
Abstract
Adenocarcinoma of the ampulla of Vater (AOV) is classified into intestinal type (IT) and pancreatobiliary type (PB); however, the immunological properties of these subtypes remain to be characterized. Here, we evaluated the clinical implications of PD-L1 expression and CD8+ T lymphocyte density in adenocarcinomas of the AOV and their potential association with Yes-associated protein (YAP). We analyzed 123 adenocarcinoma-of-the-AOV patients who underwent surgical resection, and tumors were classified into IT or PB type. Tumor or inflammatory cell PD-L1 expression, CD8+ T lymphocyte density in the cancer cell nest (intratumoral) or in the adjacent stroma, and YAP localization and intensity were analyzed using immunohistochemical staining. PB-type tumors showed higher tumoral PD-L1 expression than IT-type tumors, and tumoral PD-L1 expression was associated with a shorter disease-free survival (DFS) [hazard ratio (HR), 1.77; p = 0.045] and overall survival (OS) (HR 1.99; p = 0.030). Intratumoral CD8+ T lymphocyte density was higher in IT type than in PB type and was associated with a favorable DFS (HR 0.47; p = 0.022). The nuclear staining pattern of YAP in tumor cells, compared to non-nuclear staining patterns, was more frequently associated with PB type and increased tumoral PD-L1 expression. Nuclear YAP staining was a significant prognostic factor for OS (HR 2.21; p = 0.022). These results show that the two subtypes of adenocarcinoma of the AOV exhibit significant differences in tumoral PD-L1 expression and intratumoral CD8+ T lymphocyte density, which might contribute to their distinct clinical features.
Collapse
Affiliation(s)
- Min Hwan Kim
- Division of Medical Oncology, Department of Internal Medicine, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
- Pancreaticobiliary Cancer Clinic, Yonsei Cancer Center, Severance Hospital, Seoul, Republic of Korea
| | - Mi Jang
- Department of Pathology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Hoguen Kim
- Department of Pathology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Woo Jung Lee
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Chang Moo Kang
- Pancreaticobiliary Cancer Clinic, Yonsei Cancer Center, Severance Hospital, Seoul, Republic of Korea.
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea.
| | - Hye Jin Choi
- Division of Medical Oncology, Department of Internal Medicine, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea.
- Pancreaticobiliary Cancer Clinic, Yonsei Cancer Center, Severance Hospital, Seoul, Republic of Korea.
| |
Collapse
|
47
|
Abstract
Somatic mutations in cancer cells may influence tumor growth, survival, or immune interactions in their microenvironment. The tumor necrosis factor receptor family member HVEM (TNFRSF14) is frequently mutated in cancers and has been attributed a tumor suppressive role in some cancer contexts. HVEM functions both as a ligand for the lymphocyte checkpoint proteins BTLA and CD160, and as a receptor that activates NF-κB signaling pathways in response to BTLA and CD160 and the TNF ligands LIGHT and LTα. BTLA functions to inhibit lymphocyte activation, but has also been ascribed a role in stimulating cell survival. CD160 functions to co-stimulate lymphocyte function, but has also been shown to activate inhibitory signaling in CD4+ T cells. Thus, the role of HVEM within diverse cancers and in regulating the immune responses to these tumors is likely context specific. Additionally, development of therapeutics that target proteins within this network of interacting proteins will require a deeper understanding of how these proteins function in a cancer-specific manner. However, the prominent role of the HVEM network in anti-cancer immune responses indicates a promising area for drug development.
Collapse
|
48
|
Kanemura T, Miyata H, Makino T, Tanaka K, Sugimura K, Hamada-Uematsu M, Mizote Y, Uchida H, Miyazaki Y, Takahashi T, Kurokawa Y, Yamasaki M, Wada H, Nakajima K, Takiguchi S, Mori M, Doki Y, Tahara H. Immunoregulatory influence of abundant MFG-E8 expression by esophageal cancer treated with chemotherapy. Cancer Sci 2018; 109:3393-3402. [PMID: 30156356 PMCID: PMC6215892 DOI: 10.1111/cas.13785] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2018] [Revised: 07/26/2018] [Accepted: 08/14/2018] [Indexed: 12/16/2022] Open
Abstract
Milk fat globule‐epidermal growth factor factor 8 (MFG‐E8) is secreted from macrophages and is known to induce immunological tolerance mediated by regulatory T cells. However, the roles of the MFG‐E8 that is expressed by cancer cells have not yet been fully examined. Expression of MFG‐E8 was examined using immunohistochemistry in surgical samples from 134 patients with esophageal squamous cell carcinoma. The relationships between MFG‐E8 expression levels and clinicopathological factors, including tumor‐infiltrating lymphocytes, were evaluated. High MFG‐E8 expression was observed in 23.9% of the patients. The patients with tumors highly expressing MFG‐E8 had a significantly higher percentage of neoadjuvant chemotherapy (NAC) history (P < .0001) and shorter relapse‐free survival (P = 0.012) and overall survival (OS; P = .0047). On subgroup analysis, according to NAC history, patients with high MFG‐E8 expression had significantly shorter relapse‐free survival (P = .027) and OS (P = .0039) only when they had been treated with NAC. Furthermore, tumors with high MFG‐E8 expression had a significantly lower ratio of CD8+ T cells/regulatory T cells in tumor‐infiltrating lymphocytes (P = .042) only in the patients treated with NAC, and those with a lower ratio had a shorter OS (P = .026). High MFG‐E8 expression was also found to be an independent prognostic factor in multivariate analysis. The abundant MFG‐E8 expression in esophageal squamous cell carcinoma might have a negative influence on the long‐term survival of patients after chemotherapy by affecting T‐cell regulation in the tumor microenvironment.
Collapse
Affiliation(s)
- Takashi Kanemura
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Hiroshi Miyata
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Japan.,Department of Digestive Surgery, Osaka International Cancer Institute, Osaka, Japan
| | - Tomoki Makino
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Koji Tanaka
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Keijiro Sugimura
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Japan.,Department of Digestive Surgery, Osaka International Cancer Institute, Osaka, Japan
| | - Mika Hamada-Uematsu
- Department of Surgery and Bioengineering, Advanced Clinical Research Center, Institute of Medical Science, University of Tokyo, Tokyo, Japan.,Project Division of Cancer Biomolecular Therapy, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Yu Mizote
- Department of Surgery and Bioengineering, Advanced Clinical Research Center, Institute of Medical Science, University of Tokyo, Tokyo, Japan.,Project Division of Cancer Biomolecular Therapy, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Hiroaki Uchida
- Department of Surgery and Bioengineering, Advanced Clinical Research Center, Institute of Medical Science, University of Tokyo, Tokyo, Japan.,Project Division of Cancer Biomolecular Therapy, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Yasuhiro Miyazaki
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Tsuyoshi Takahashi
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Yukinori Kurokawa
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Makoto Yamasaki
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Hisashi Wada
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Kiyokazu Nakajima
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Shuji Takiguchi
- Department of Gastroenterological Surgery, Graduate School of Medicine, Nagoya City University, Nagoya, Japan
| | - Masaki Mori
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Yuichiro Doki
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Hideaki Tahara
- Department of Surgery and Bioengineering, Advanced Clinical Research Center, Institute of Medical Science, University of Tokyo, Tokyo, Japan.,Project Division of Cancer Biomolecular Therapy, Institute of Medical Science, University of Tokyo, Tokyo, Japan.,Department of Cancer Drug Discovery and Development, Research Center, Osaka International Cancer Institute, Osaka, Japan
| |
Collapse
|
49
|
Miyama Y, Morikawa T, Miyakawa J, Koyama Y, Kawai T, Kume H, Fukayama M. The prognostic value of PD-L1 expression in upper tract urothelial carcinoma varies according to platelet count. Cancer Med 2018; 7:4330-4338. [PMID: 30062756 PMCID: PMC6143937 DOI: 10.1002/cam4.1686] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Revised: 06/17/2018] [Accepted: 06/23/2018] [Indexed: 12/19/2022] Open
Abstract
Programmed cell death ligand-1 (PD-L1) is a ligand for programmed cell death-1 (PD-1) that negatively regulates T-cell activation and plays a crucial role in suppressing anti-tumor host immunity. Although PD-L1 is a promising immunotherapy target in various cancers, including urothelial carcinoma (UC), the prognostic significance of PD-L1 in UC is unclear. As platelets help protect tumor cells from immune elimination in the circulatory system, we hypothesized that tumor PD-L1 and circulating platelets might synergistically promote tumor metastasis, and that the prognostic significance of PD-L1 might vary according to platelet count. We immunohistochemically examined tumor PD-L1 expression in 271 patients with upper tract UC, which revealed PD-L1 positivity in 31 of 271 cases (11%). The associations of tumor PD-L1 expression with outcomes varied among patients with high or low platelet counts (Pinteraction < 0.004). Among patients with high platelet counts (N = 136), PD-L1 positivity (N = 15) was significantly associated with shorter metastasis-free survival (univariate hazard ratio [HR]: 6.23, 95% confidence interval [CI]: 2.95-13.1; multivariate HR: 2.68, 95% CI: 1.27-5.64) and shorter overall survival (univariate HR: 4.92, 95% CI: 2.14-11.3, multivariate HR: 2.78, 95% CI: 1.19-6.51). In contrast, among patients with low platelet counts (N = 135), PD-L1 positivity (N = 16) was not significantly associated with these outcomes. Our results suggest that tumor PD-L1 expression and platelet count might interact and help regulate tumor progression. Although a larger prospective study is needed to validate our findings, this relationship is important to consider, as immunotherapies targeting the PD-1/PD-L1 axis have gained significant attention as promising therapies for UC.
Collapse
Affiliation(s)
- Yu Miyama
- Department of Pathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Teppei Morikawa
- Department of Pathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.,Department of Diagnostic Pathology, NTT Medical Center Tokyo, Tokyo, Japan
| | - Jimpei Miyakawa
- Department of Urology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yuichi Koyama
- Department of Pathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Taketo Kawai
- Department of Urology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Haruki Kume
- Department of Urology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Masashi Fukayama
- Department of Pathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
50
|
Enninga EAL, Chatzopoulos K, Butterfield JT, Sutor SL, Leontovich AA, Nevala WK, Flotte TJ, Markovic SN. CD206-positive myeloid cells bind galectin-9 and promote a tumor-supportive microenvironment. J Pathol 2018; 245:468-477. [PMID: 29732570 DOI: 10.1002/path.5093] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Revised: 03/30/2018] [Accepted: 04/26/2018] [Indexed: 01/19/2023]
Abstract
In patients with metastatic melanoma, high blood levels of galectin-9 are correlated with worse overall survival and a bias towards a Th2 inflammatory state supportive of tumor growth. Although galectin-9 signaling through TIM3 on T cells has been described, less is known about the interaction of galectin-9 with macrophages. We aimed to determine whether galectin-9 is a binding partner of CD206 on macrophages and whether the result of this interaction is tumor-supportive. It was determined that incubation of CD68+ macrophages with galectin-9 or anti-CD206 blocked target binding and that both CD206 and galectin-9 were detected by immunoprecipitation of cell lysates. CD206 and galectin-9 had a binding affinity of 2.8 × 10-7 m. Galectin-9 causes CD206+ macrophages to make significantly more FGF2 and monocyte chemoattractant protein (MCP-1), but less macrophage-derived chemokine (MDC). Galectin-9 had no effect on classical monocyte subsets, but caused expansion of the non-classical populations. Lastly, there was a positive correlation between increasing numbers of CD206 macrophages and galectin-9 expression in tumors, and high levels of CD206 macrophages correlated negatively with melanoma survival. These results indicate that galectin-9 binds to CD206 on M2 macrophages, which appear to drive angiogenesis and the production of chemokines that support tumor growth and poor patient prognoses. Targeting this interaction systemically through circulating monocytes may therefore be a novel way to improve local anti-tumor effects by macrophages. Copyright © 2018 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
| | | | | | - Shari L Sutor
- Department of Oncology, Mayo Clinic, Rochester, Minnesota, USA
| | - Alexey A Leontovich
- Department of Bioinformatics and Statistics, Mayo Clinic, Rochester, Minnesota, USA
| | - Wendy K Nevala
- Department of Oncology, Mayo Clinic, Rochester, Minnesota, USA
| | - Thomas J Flotte
- Department of Anatomic Pathology, Mayo Clinic, Rochester, Minnesota, USA
| | | |
Collapse
|