1
|
Charalampopoulou A, Barcellini A, Magro G, Bellini A, Borgna SS, Fulgini G, Ivaldi GB, Mereghetti A, Orlandi E, Pullia MG, Savazzi S, Tabarelli De Fatis P, Volpi G, Facoetti A. Advancing Radiobiology: Investigating the Effects of Photon, Proton, and Carbon-Ion Irradiation on PANC-1 Cells in 2D and 3D Tumor Models. Curr Oncol 2025; 32:49. [PMID: 39851965 PMCID: PMC11763791 DOI: 10.3390/curroncol32010049] [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: 12/01/2024] [Revised: 01/09/2025] [Accepted: 01/17/2025] [Indexed: 01/26/2025] Open
Abstract
Introduction: Pancreatic cancer (PC) is one of the most aggressive and lethal malignancies, calling for enhanced research. Pancreatic ductal adenocarcinoma (PDAC) represents 70-80% of all cases and is known for its resistance to conventional therapies. Carbon-ion radiotherapy (CIRT) has emerged as a promising approach due to its ability to deliver highly localized doses and unique radiobiological properties compared to X-rays. In vitro radiobiology has relied on two-dimensional (2D) cell culture models so far; however, these are not sufficient to replicate the complexity of the in vivo tumor architecture. Three-dimensional (3D) models become a paradigm shift, surpassing the constraints of traditional models by accurately re-creating morphological, histological, and genetic characteristics as well as the interaction of tumour cells with the microenvironment. Materials and Methods: This study investigates the survival of pancreatic cancer cells in both 2D and spheroids, a 3D model, following photon, proton, and carbon-ion irradiation by means of clonogenic, MTT, spheroid growth, and vitality assays. Results: Our results demonstrate that carbon ions are more efficient in reducing cancer cell survival compared to photons and protons. In 2D cultures, carbon-ion irradiation reduced cell survival to approximately 15%, compared to 45% with photons and 30% with protons. In the 3D culture model, spheroid growth was similarly inhibited by carbon-ion irradiation; however, the overall survival rates were higher across all irradiation modalities compared to the 2D cultures. Carbon ions consistently showed the highest efficacy in reducing cell viability in both models. Conclusions: Our research highlights the pivotal role of 3D models in unraveling the complexities of pancreatic cancer radiobiology, offering new avenues for designing more effective and precise treatment protocols.
Collapse
Affiliation(s)
- Alexandra Charalampopoulou
- Radiobiology Unit, Research and Development Department, CNAO National Center for Oncological Hadrontherapy, 27100 Pavia, Italy; (A.B.); (S.S.B.); (G.F.); (G.V.); (A.F.)
- Hadron Academy PhD Course, School for Advanced Studies (IUSS), 27100 Pavia, Italy
| | - Amelia Barcellini
- Radiation Oncology Unit, Clinical Department, CNAO National Center for Oncological Hadrontherapy, 27100 Pavia, Italy; (A.B.); (E.O.)
- Department of Internal Medicine and Therapeutics, University of Pavia, 27100 Pavia, Italy
| | - Giuseppe Magro
- Medical Physics Unit, Clinical Department, CNAO National Center for Oncological Hadrontherapy, 27100 Pavia, Italy;
| | - Anna Bellini
- Radiobiology Unit, Research and Development Department, CNAO National Center for Oncological Hadrontherapy, 27100 Pavia, Italy; (A.B.); (S.S.B.); (G.F.); (G.V.); (A.F.)
| | - Sara Sevan Borgna
- Radiobiology Unit, Research and Development Department, CNAO National Center for Oncological Hadrontherapy, 27100 Pavia, Italy; (A.B.); (S.S.B.); (G.F.); (G.V.); (A.F.)
| | - Giorgia Fulgini
- Radiobiology Unit, Research and Development Department, CNAO National Center for Oncological Hadrontherapy, 27100 Pavia, Italy; (A.B.); (S.S.B.); (G.F.); (G.V.); (A.F.)
| | - Giovanni Battista Ivaldi
- Radiation Oncology Department, Clinical Scientific Institutes Maugeri IRCCS, 27100 Pavia, Italy;
| | - Alessio Mereghetti
- Research and Development Department, CNAO National Center for Oncological Hadrontherapy, 27100 Pavia, Italy; (A.M.); (M.G.P.); (S.S.)
| | - Ester Orlandi
- Radiation Oncology Unit, Clinical Department, CNAO National Center for Oncological Hadrontherapy, 27100 Pavia, Italy; (A.B.); (E.O.)
- Department of Clinical, Surgical, Diagnostic and Pediatric Sciences, University of Pavia, 27100 Pavia, Italy
| | - Marco Giuseppe Pullia
- Research and Development Department, CNAO National Center for Oncological Hadrontherapy, 27100 Pavia, Italy; (A.M.); (M.G.P.); (S.S.)
| | - Simone Savazzi
- Research and Development Department, CNAO National Center for Oncological Hadrontherapy, 27100 Pavia, Italy; (A.M.); (M.G.P.); (S.S.)
| | | | - Gaia Volpi
- Radiobiology Unit, Research and Development Department, CNAO National Center for Oncological Hadrontherapy, 27100 Pavia, Italy; (A.B.); (S.S.B.); (G.F.); (G.V.); (A.F.)
- Hadron Academy PhD Course, School for Advanced Studies (IUSS), 27100 Pavia, Italy
| | - Angelica Facoetti
- Radiobiology Unit, Research and Development Department, CNAO National Center for Oncological Hadrontherapy, 27100 Pavia, Italy; (A.B.); (S.S.B.); (G.F.); (G.V.); (A.F.)
| |
Collapse
|
2
|
Wang X, Guan J, Feng L, Li Q, Zhao L, Li Y, Ma R, Shi M, Han B, Hao G, Wang L, Li H, Wang X. A machine learning-based immune response signature to facilitate prognosis prediction in patients with endometrial cancer. Sci Rep 2024; 14:30801. [PMID: 39730507 DOI: 10.1038/s41598-024-81040-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 11/25/2024] [Indexed: 12/29/2024] Open
Abstract
Endometrial cancer is the most prevalent form of gynecologic malignancy, with a significant surge in incidence among youngsters. Although the advent of the immunotherapy era has profoundly improved patient outcomes, not all patients benefit from immunotherapy; some patients experience hyperprogression while on immunotherapy. Hence, there is a pressing need to further delineate the distinct immune response profiles in patients with endometrial cancer to enhance prognosis prediction and facilitate the prediction of immunotherapeutic responses. The ssGSEA method was used to evaluate the activities of the immune response pathways in patients with endometrial cancer. Unsupervised clustering was employed to identify the different immune response patterns. WGCNA was employed to identify the genes that highly correlated with the immune response patterns observed. Ninety-five machine learning combinations were utilized to identify the optimal prognosis model and the novel biomarker, SLC38A3. Experiments such as cell invasion, migration, scratch, and in vivo tumorigenicity were performed to determine the function of SLC28A3. Molecular docking techniques were employed to determine the targeted action of periodate-oxidized adenosine on SLC38A3. Patients exhibited both immune response-suppressing C1 phenotypes and immune response-activating C2 phenotypes, with significant differences in prognosis between these two phenotypes. WGCNA identified 418 genes that highly correlated with the immune response phenotypes, of which 69 genes were associated with prognosis. The immune response-related score (IRRS) established by multiple machine learning frameworks demonstrated stability in predicting patient prognosis and immune status. High expression of SLC38A3 contributes to cellular malignant traits, and periodate-oxidized adenosine bound stably to SLC38A3. IRRS accurately predicts disease prognosis and immune status in patients with endometrial cancer. SLC38A3 serves as a prognostic marker for these patients and can be stably targeted by periodate-oxidized adenosine.
Collapse
Affiliation(s)
- Xiaofeng Wang
- Department of Oncology, The Fourth Hospital of Shijiazhuang, Shijiazhuang, Hebei Province, China
| | - Jing Guan
- Department of Radiology, The Fourth Hospital of Shijiazhuang, Shijiazhuang, Hebei Province, China
| | - Li Feng
- The Fourth Hospital of Shijiazhuang, Shijiazhuang, Hebei Province, China
| | - Qingxue Li
- The Fourth Hospital of Shijiazhuang, Shijiazhuang, Hebei Province, China
| | - Liwei Zhao
- Department of Oncology, The Fourth Hospital of Shijiazhuang, Shijiazhuang, Hebei Province, China
| | - Yue Li
- Department of Oncology, The Fourth Hospital of Shijiazhuang, Shijiazhuang, Hebei Province, China
| | - Ruixiao Ma
- Department of Oncology, The Fourth Hospital of Shijiazhuang, Shijiazhuang, Hebei Province, China
| | - Mengnan Shi
- Department of Oncology, The Fourth Hospital of Shijiazhuang, Shijiazhuang, Hebei Province, China
| | - Biaogang Han
- Department of Oncology, The Fourth Hospital of Shijiazhuang, Shijiazhuang, Hebei Province, China
| | - Guorong Hao
- The Fourth Hospital of Shijiazhuang, Shijiazhuang, Hebei Province, China
| | - Lina Wang
- The Fourth Hospital of Shijiazhuang, Shijiazhuang, Hebei Province, China
| | - Hui Li
- The Fourth Hospital of Shijiazhuang, Shijiazhuang, Hebei Province, China.
| | - Xiuli Wang
- Department of Laboratory Medicine, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, China.
| |
Collapse
|
3
|
Davar D, Carneiro BA, Dy GK, Sheth S, Borad MJ, Harrington KJ, Patel SP, Galanis E, Samson A, Agrawal S, Chen Z, Fan C, Gong M, Burton J, Tu E, Durham N, Laubscher K, Arnaldez F, Zamarin D. Phase I study of a recombinant attenuated oncolytic virus, MEDI5395 (NDV-GM-CSF), administered systemically in combination with durvalumab in patients with advanced solid tumors. J Immunother Cancer 2024; 12:e009336. [PMID: 39551600 PMCID: PMC11574399 DOI: 10.1136/jitc-2024-009336] [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: 03/25/2024] [Accepted: 10/13/2024] [Indexed: 11/19/2024] Open
Abstract
BACKGROUND MEDI5395 is a recombinant attenuated Newcastle disease virus engineered to express a human granulocyte-macrophage colony-stimulating factor transgene. Preclinically, MEDI5395 demonstrated broad oncolytic activity, augmented by concomitant programmed cell death-1/programmed cell death ligand-1 (PD-L1) axis blockade. Durvalumab is an anti-PD-L1 immune checkpoint inhibitor approved for the treatment of various solid tumors. We describe the results of the first-in-human study combining intravenous MEDI5395 with durvalumab in patients with advanced solid tumors. METHODS This phase I, open-label, multicenter, dose-escalation, dose-expansion study recruited adult patients with advanced solid tumors, who had relapsed or were refractory or intolerant to ≥1 prior line of standard treatment. MEDI5395 was administered intravenously as six doses over 15-18 days. The dose-escalation phase assessed four-dose levels (108, 109, 1010, 1011 focus forming units (FFU)) of MEDI5395, with sequential or delayed durvalumab. Durvalumab 1500 mg was administered intravenously every 4 weeks up to 2 years. The dose-expansion phase was not initiated. The primary objectives were to evaluate safety and tolerability, dose-limiting toxicities (DLTs) and the dose and schedule of MEDI5395 plus durvalumab administration. Secondary objectives included the assessment of the efficacy, pharmacokinetics, pharmacodynamics, and immunogenicity of MEDI5395. RESULTS 39 patients were treated with MEDI5395; 36 patients also received durvalumab. All 39 patients experienced ≥1 treatment-emergent adverse event (TEAE), most commonly fatigue (61.5%), nausea (53.8%) and chills (51.3%). Grade 3-4 TEAEs occurred in 27 (69.2%) patients; these were deemed MEDI5395-related in 12 (30.8%) patients. Two patients experienced a DLT, and the maximum tolerated dose of MEDI5395 with sequential and delayed durvalumab at study termination was 1011 and 1010 FFU, respectively. Four patients (10.3%) achieved a partial response (PR). Patients with PR or stable disease tended to have higher baseline PD-L1 and CD8+ levels in their tumor tissue. A tendency to dose-dependent pharmacokinetics of the viral genome was observed in whole blood and a tendency to dose-dependent viral shedding was observed in saliva and urine. Neutralizing antibodies were observed in all patients but did not appear to impact efficacy negatively. CONCLUSION This study demonstrates the feasibility, safety and preliminary efficacy of MEDI5395 with durvalumab in patients with advanced solid tumors. TRIAL REGISTRATION NUMBER NCT03889275.
Collapse
Affiliation(s)
- Diwakar Davar
- UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
| | - Benedito A Carneiro
- Lifespan Cancer Institute, Legorreta Cancer Institute at Brown University, Providence, Rhode Island, USA
| | - Grace K Dy
- Department of Medicine, Roswell Park Cancer Institute, Buffalo, New York, USA
| | - Siddharth Sheth
- Division of Hematology/Oncology, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | | | - Kevin J Harrington
- Division of Radiotherapy and Imaging, The Institute of Cancer Research / The Royal Marsden NIHR Biomedical Research Centre, London, UK
| | - Sandip P Patel
- University of California San Diego, La Jolla, California, USA
| | | | - Adel Samson
- Leeds Institute of Medical Research at St. James’s, University of Leeds, Leeds, UK
| | - Sonia Agrawal
- Oncology Data Science, Research and Early Development, Oncology R&D, AstraZeneca R&D Gaithersburg, Gaithersburg, Maryland, USA
| | - Zhongying Chen
- Integrated Bioanalysis, Clinical Pharmacology and Safety Sciences (CPSS), BioPharmaceuticals R&D, AstraZeneca R&D Gaithersburg, Gaithersburg, Maryland, USA
| | - Chunling Fan
- Clinical Pharmacology and Quantitative Pharmacology, Clinical Pharmacology and Safety Sciences (CPSS), BioPharmaceuticals R&D, AstraZeneca R&D Gaithersburg, Gaithersburg, Maryland, USA
| | - Maozhen Gong
- AstraZeneca R&D Gaithersburg, Gaithersburg, Maryland, USA
| | - Jenny Burton
- Oncology R&D, AstraZeneca PLC, Cambridge, Cambridgeshire, UK
| | - Eric Tu
- Translational Medicine, Cell Therapy and Oncolytic Viruses, BioPharmaceuticals R&D, AstraZeneca R&D Gaithersburg, Gaithersburg, Maryland, USA
| | - Nicholas Durham
- Translational Medicine, Cell Therapy and Oncolytic Viruses, BioPharmaceuticals R&D, AstraZeneca R&D Gaithersburg, Gaithersburg, Maryland, USA
| | - Kevin Laubscher
- Oncology Data Science, Research and Early Development, Oncology R&D, AstraZeneca R&D Gaithersburg, Gaithersburg, Maryland, USA
| | - Fernanda Arnaldez
- Clinical Development, Oncology R&D, AstraZeneca R&D Gaithersburg, Gaithersburg, Maryland, USA
| | - Dmitriy Zamarin
- Early Drug Development, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| |
Collapse
|
4
|
Groeger S, Meyle J. The role of programmed death receptor (PD-)1/PD-ligand (L)1 in periodontitis and cancer. Periodontol 2000 2024; 96:150-169. [PMID: 38351432 PMCID: PMC11579837 DOI: 10.1111/prd.12548] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 11/02/2023] [Accepted: 12/14/2023] [Indexed: 11/22/2024]
Abstract
The programmed-death-ligand-1 (PD-L1) is an immune-modulating molecule that is constitutively expressed on various immune cells, different epithelial cells and a multitude of cancer cells. It is a costimulatory molecule that may impair T-cell mediated immune response. Ligation to the programmed-death-receptor (PD)-1, on activated T-cells and further triggering of the related signaling pathways can induce T-cells apoptosis or anergy. The upregulation of PD-L1 in various cancer types, including oral squamous cell carcinomas, was demonstrated and has been linked to immune escape of tumors and poor prognosis. A bidirectional relationship exists between the increased PD-L1 expression and periodontitis as well as the epithelial-mesenchymal transition (EMT), a process of interconversion of epithelial cells to mesenchymal cells that may induce immune escape of tumors. Interaction between exosomal PD-L1 and PD-1 on T-cells may cause immunosuppression by blocking the activation and proliferation of T-cells. The efficacy and importance of treatment with PD-1/PD-L1 checkpoint inhibitors and their prognostic influence on human cancers was demonstrated. Regarding PD-1/PD-L1 checkpoint inhibitors, resistances exist or may develop, basing on various factors. Further investigations of the underlying mechanisms will help to overcome the therapeutic limitations that result from resistances and to develop new strategies for the treatment of cancer.
Collapse
Affiliation(s)
- Sabine Groeger
- Department of Periodontology, Dental SchoolJustus‐Liebig‐University of GiessenGiessenGermany
- Department of Orthodontics, Dental SchoolJustus‐Liebig‐University of GiessenGiessenGermany
| | - Joerg Meyle
- Department of Periodontology, Dental SchoolJustus‐Liebig‐University of GiessenGiessenGermany
| |
Collapse
|
5
|
Solomon SR, Solh M, Morris LE, Holland HK, Bachier-Rodriguez L, Zhang X, Guzowski C, Jackson KC, Brown S, Bashey A. Phase 2 study of PD-1 blockade following autologous transplantation for patients with AML ineligible for allogeneic transplant. Blood Adv 2023; 7:5215-5224. [PMID: 37379271 PMCID: PMC10500475 DOI: 10.1182/bloodadvances.2023010477] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 05/25/2023] [Accepted: 06/11/2023] [Indexed: 06/30/2023] Open
Abstract
Allogeneic transplant remains the best postremission therapy for patients with nonfavorable risk acute myeloid leukemia (AML). However, some patients are ineligible because of psychosocial barriers, such as lack of appropriate caregiver support. We hypothesized that immune checkpoint inhibition after autologous transplant might represent effective postremission therapy in such patients. We conducted a phase 2 study of autologous transplantation followed by administration of pembrolizumab (8 cycles starting day +1). Twenty patients with nonfavorable AML in complete remission were treated (median age, 64 years; CR1, 80%); 55% were non-White and adverse-risk AML was present in 40%. Treatment was well tolerated, with only 1 nonrelapse death. Immune-related adverse events occurred in 9 patients. After a median follow-up of 80 months, 14 patients remain alive, with 10 patients in continuous remission. The estimated 2-year LFS was 48.4%, which met the primary end point of 2-year LFS >25%; the 2-year overall survival (OS), nonrelapse mortality, and cumulative incidences of relapse were 68%, 5%, and 46%, respectively. In comparison with a propensity score-matched cohort group of patients with AML receiving allogeneic transplant, the 3-year OS was similar (73% vs 76%). Patients in the study had inferior LFS (51% vs 75%) but superior postrelapse survival (45% vs 14%). In conclusion, programmed cell death protein-1 blockade after autologous transplant is a safe and effective alternative postremission strategy in patients with nonfavorable risk AML who are ineligible for allogeneic transplant, a context in which there is significant unmet need. This trial was registered at www.clinicaltrials.gov as #NCT02771197.
Collapse
Affiliation(s)
- Scott R. Solomon
- Blood and Marrow Transplant Program, Northside Hospital Cancer Institute, Atlanta, GA
| | - Melhem Solh
- Blood and Marrow Transplant Program, Northside Hospital Cancer Institute, Atlanta, GA
| | - Lawrence E. Morris
- Blood and Marrow Transplant Program, Northside Hospital Cancer Institute, Atlanta, GA
| | - H. Kent Holland
- Blood and Marrow Transplant Program, Northside Hospital Cancer Institute, Atlanta, GA
| | | | - Xu Zhang
- Center for Clinical and Transitional Sciences, University of Texas Health Science Center, Houston, TX
| | - Caitlin Guzowski
- Blood and Marrow Transplant Program, Northside Hospital Cancer Institute, Atlanta, GA
| | - Katelin C Jackson
- Blood and Marrow Transplant Program, Northside Hospital Cancer Institute, Atlanta, GA
| | - Stacey Brown
- Blood and Marrow Transplant Program, Northside Hospital Cancer Institute, Atlanta, GA
| | - Asad Bashey
- Blood and Marrow Transplant Program, Northside Hospital Cancer Institute, Atlanta, GA
| |
Collapse
|
6
|
PD-L1/PD-1 blockage enhanced the cytotoxicity of natural killer cell on the non-small cell lung cancer (NSCLC) by granzyme B secretion. Clin Transl Oncol 2023:10.1007/s12094-023-03120-w. [PMID: 36856921 DOI: 10.1007/s12094-023-03120-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 02/09/2023] [Indexed: 03/02/2023]
Abstract
OBJECTIVE To explore the role of PD-L1/PD-1 blockage in the cytotoxicity of natural killer cell in NSCLC. METHODS Two NSCLC cell lines, Calu-1 and H460, were tested for susceptibility to the cytolytic activity of freshly isolated healthy donor NK cells by a non-radioactive cellular cytotoxicity assay kit. Western blot analysis, FACS, ELISA and antibody blockage experiments were conducted to determine the mechanisms. NK cells isolated from NSCLC patients were also collected for functional assays. RESULTS Calu-1 and H460 cells were lysed by NK cells in a dose-dependent manner. H460 cells showed less susceptibility to NK cell-mediated lysis than Calu-1 cells at all ratios. The expression of PD-L1 on H460 cells was higher than that on Calu-1 cells, as determined by FACS and western blot analysis. The specific lysis of H460 cells by NK cells was enhanced when the PD-L1/PD-1 interaction was blocked by anti-PD-L1 antibody. This finding was also demonstrated in NK cells isolated from NSCLC patients. CONCLUSIONS The present study revealed that PD-L1/PD-1 blockage enhanced the cytotoxicity of natural killer cells in NSCLC via granzyme B secretion. This study will greatly facilitate the precise treatment of lung cancer through determination of PD-L1 expression in tumors.
Collapse
|
7
|
Righi I, Vaira V, Morlacchi LC, Croci GA, Rossetti V, Blasi F, Ferrero S, Nosotti M, Rosso L, Clerici M. PD-1 expression in transbronchial biopsies of lung transplant recipients is a possible early predictor of rejection. Front Immunol 2023; 13:1024021. [PMID: 36703976 PMCID: PMC9871480 DOI: 10.3389/fimmu.2022.1024021] [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: 08/20/2022] [Accepted: 12/16/2022] [Indexed: 01/11/2023] Open
Abstract
Introduction Chronic lung allograft dysfunction (CLAD) is the main cause of the reduced survival of lung transplanted (LTx) patients. The possible role of immune checkpoint molecules in establishing tolerance has been scarcely investigated in the setting of lung transplantation. Methods We conducted a retrospective, observational pilot study on a consecutive series of transbronchial cryobiopsies (TCB) obtained from 24 patients during LTx follow-up focusing on PD-1, one of the most investigated immune checkpoint molecules. Results Results showed that PD-1-expressing T lymphocytes were present in all TCB with a histological diagnosis of acute rejection (AR; 9/9), but not in most (11/15) of the TCB not resulting in a diagnosis of AR (p=0.0006). Notably, the presence of PD-1-expressing T lymphocytes in TCB resulted in a 10-times higher risk of developing chronic lung allograft dysfunction (CLAD), the main cause of the reduced survival of lung transplanted patients, thus being associated with a clearly worst clinical outcome. Discussion Results of this pilot study indicate a central role of PD-1 in the development of AR and its evolution towards CLAD and suggest that the evaluation of PD-1-expressing lymphocytes in TCB could offer a prognostic advantage in monitoring the onset of AR in patients who underwent lung transplantation.
Collapse
Affiliation(s)
- Ilaria Righi
- Thoracic Surgery and Lung Transplantation Unit, Department of Cardio- Thoracic - Vascular Disease, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Valentina Vaira
- Division of Pathology, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy,Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - Letizia Corinna Morlacchi
- Respiratory Unit and Adult Cystic Fibrosis Center, Internal Medicine Department, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Giorgio Alberto Croci
- Division of Pathology, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy,Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - Valeria Rossetti
- Respiratory Unit and Adult Cystic Fibrosis Center, Internal Medicine Department, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Francesco Blasi
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy,Respiratory Unit and Adult Cystic Fibrosis Center, Internal Medicine Department, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Stefano Ferrero
- Division of Pathology, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy,Department of Biomedical, Surgical and Dental Sciences, University of Milan, Milan, Italy
| | - Mario Nosotti
- Thoracic Surgery and Lung Transplantation Unit, Department of Cardio- Thoracic - Vascular Disease, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy,Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - Lorenzo Rosso
- Thoracic Surgery and Lung Transplantation Unit, Department of Cardio- Thoracic - Vascular Disease, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy,Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy,*Correspondence: Lorenzo Rosso,
| | - Mario Clerici
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy,Don C. Gnocchi Foundation, IRCCS, Milan, Italy
| |
Collapse
|
8
|
Pal K, Hussain T, Xie H, Li S, Yang P, Mansfield A, Lou Y, Chowdhury S, Mukhopadhyay D. Expression, correlation, and prognostic significance of different nicotinic acetylcholine receptors, programed death ligand 1, and dopamine receptor D2 in lung adenocarcinoma. Front Oncol 2022; 12:959500. [PMID: 36072788 PMCID: PMC9441878 DOI: 10.3389/fonc.2022.959500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 07/21/2022] [Indexed: 12/24/2022] Open
Abstract
Objective The objective of this study is to evaluate the expression of different nicotinic acetylcholine receptors (nAChRs), programmed death ligand-1 (PD-L1), and dopamine receptor D2 (DRD2) as prognostic factors in lung cancer and any correlation among them. Since all of the above genes are typically upregulated in response to smoking, we hypothesized that a correlation might exist between DRD2, PD-L1, and nAChR expression in NSCLC patients with a smoking history and a prediction model may be developed to assess the clinical outcome. Methods We retrospectively analyzed samples from 46 patients with primary lung adenocarcinoma who underwent surgical resection at Mayo Clinic Rochester from June 2000 to October 2008. The expression of PD-L1, DRD2, CHRNA5, CHRNA7, and CHRNA9 were analyzed by quantitative PCR and correlated amongst themselves and with age, stage and grade, smoking status, overall survival (OS), and relapse-free survival (RFS). Results Only PD-L1 showed a statistically significant increase in expression in patients older than 65. All the above genes showed higher expression in stage IIIB than IIIA, but none reached statistical significance. Interestingly, we did not observe significant differences among never, former, and current smokers, but patients with pack years greater than 30 showed significantly higher expression of CHRNA9. We observed a strong positive correlation between PD-L1/DRD2, PD-L1/CHRNA5, and CHRNA5/CHRNA7 and a weak positive correlation between DRD2/CHRNA5 and DRD2/CHRNA7. Older age was independently associated with poor OS, whereas lower CHRNA7 expression was independently associated with better OS. Conclusions We observed strong positive correlations among PD-L1, DRD2, and some of the nAChRs. We investigated their prognostic significance in lung cancer patients and found CHRNA7 to be an independent prognostic factor. Overall, the results obtained from this preliminary study warrant a large cohort-based analysis that may ultimately lead to potential patient-specific stratification biomarkers predicting cancer-treatment outcomes.
Collapse
Affiliation(s)
- Krishnendu Pal
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Jacksonville, FL, United States
| | - Tabish Hussain
- Genomics and Molecular Medicine Unit, CSIR-Institute of Genomics and Integrative Biology, Mathura Road, New Delhi, India
| | - Hao Xie
- Division of Medical Oncology, Mayo Clinic, Rochester, MN, United States
| | - Shenduo Li
- Division of Hematology and Medical Oncology, Mayo Clinic, Jacksonville, FL, United States
| | - Ping Yang
- Department of Quantitative Health Sciences, Mayo Clinic Scottsdale, AZ, United States
| | - Aaron Mansfield
- Division of Medical Oncology, Mayo Clinic, Rochester, MN, United States
| | - Yanyan Lou
- Division of Hematology and Medical Oncology, Mayo Clinic, Jacksonville, FL, United States
| | - Shantanu Chowdhury
- Integrative and Functional Biology Unit, CSIR- Institute of Genomics and Integrative Biology, New Delhi, India
- Academy of Scientific and Innovative Research, CSIR- Institute of Genomics and Integrative Biology, New Delhi, India
| | - Debabrata Mukhopadhyay
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Jacksonville, FL, United States
| |
Collapse
|
9
|
van der Woude H, Hally KE, Currie MJ, Gasser O, Henry CE. Importance of the endometrial immune environment in endometrial cancer and associated therapies. Front Oncol 2022; 12:975201. [PMID: 36072799 PMCID: PMC9441707 DOI: 10.3389/fonc.2022.975201] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 08/04/2022] [Indexed: 11/13/2022] Open
Abstract
Endometrial cancer is rising in prevalence. The standard treatment modality of hysterectomy is becoming increasingly inadequate due primarily to the direct link between endometrial cancer and high BMI which increases surgical risks. This is an immunogenic cancer, with unique molecular subtypes associated with differential immune infiltration. Despite the immunogenicity of endometrial cancer, there is limited pre-clinical and clinical evidence of the function of immune cells in both the normal and cancerous endometrium. Immune checkpoint inhibitors for endometrial cancer are the most well studied type of immune therapy but these are not currently used as standard-of-care and importantly, they represent only one method of immune manipulation. There is limited evidence regarding the use of other immunotherapies as surgical adjuvants or alternatives. Levonorgestrel-loaded intra-uterine systems can also be effective for early-stage disease, but with varying success. There is currently no known reason as to what predisposes some patients to respond while others do not. As hormones can directly influence immune cell function, it is worth investigating the immune compartment in this context. This review assesses the immunological components of the endometrium and describes how the immune microenvironment changes with hormones, obesity, and in progression to malignancy. It also describes the importance of investigating novel pathways for immunotherapy.
Collapse
Affiliation(s)
- Hannah van der Woude
- Department of Obstetrics, Gynaecology and Women’s Health, University of Otago, Wellington, New Zealand
| | | | - Margaret Jane Currie
- Department of Pathology and Biomedical Science, University of Otago, Christchurch, New Zealand
| | - Olivier Gasser
- Malaghan Institute of Medical Research, Wellington, New Zealand
| | - Claire Elizabeth Henry
- Department of Obstetrics, Gynaecology and Women’s Health, University of Otago, Wellington, New Zealand
- *Correspondence: Claire Elizabeth Henry,
| |
Collapse
|
10
|
Arnaud M, Demonchy J, Arrii E, Luperto M, Lion J, Fodil S, Pons S, Mooney N, Zafrani L. Endothelial Cells Activated by Extracellular Histones Promote Foxp3 + Suppressive Treg Cells In Vitro. Int J Mol Sci 2022; 23:ijms23094527. [PMID: 35562918 PMCID: PMC9103825 DOI: 10.3390/ijms23094527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 04/13/2022] [Accepted: 04/15/2022] [Indexed: 11/16/2022] Open
Abstract
Histones are widely recognized as pro-inflammatory mediators upon their release from the nucleus into the extracellular space. However, their impact on endothelial cell immunogenicity is unknown. Endothelial cells, Human Microvascular Endothelial cells 1 (HMEC1), have been exposed to recombinant histones in order to study their effect on the endothelial phenotype. We then studied the differentiation of CD4+-T lymphocytes subpopulations after three days of interaction with endothelial cells in vitro and observed that histone-treated endothelial cells differentiate a suppressive FoxP3+ T regulator subpopulation that expressed Human Leucocyte Antigen DR (HLA-DR) and Cytotoxic T-Lymphocyte-Associated protein 4 (CTLA4). Toll-Like Receptor 4 (TLR4) inhibition significantly decreased the expansion of these Treg cells. Moreover, blockade of Interleukin (IL)-6 and Intercellular Adhesion Molecule (ICAM)-1 in cocultures significantly decreased the expansion of Tregs, suggesting an IL-6 and ICAM-1 dependent pathway. Thus, beyond their inflammatory effects, extracellular histones may induce an increase of immunosuppressive Treg population via their action on endothelial cells. Further studies are needed to evaluate the impact on immunosuppression of an increase of peripheral suppressive Treg via endothelial cell activation by histones in vivo.
Collapse
Affiliation(s)
- Marine Arnaud
- Human Immunology, Pathophysiology and Immunotherapy, INSERM U 976, University Paris Cite, 75010 Paris, France; (M.A.); (J.D.); (E.A.); (M.L.); (J.L.); (S.F.); (S.P.); (N.M.)
| | - Jordane Demonchy
- Human Immunology, Pathophysiology and Immunotherapy, INSERM U 976, University Paris Cite, 75010 Paris, France; (M.A.); (J.D.); (E.A.); (M.L.); (J.L.); (S.F.); (S.P.); (N.M.)
| | - Eden Arrii
- Human Immunology, Pathophysiology and Immunotherapy, INSERM U 976, University Paris Cite, 75010 Paris, France; (M.A.); (J.D.); (E.A.); (M.L.); (J.L.); (S.F.); (S.P.); (N.M.)
| | - Marta Luperto
- Human Immunology, Pathophysiology and Immunotherapy, INSERM U 976, University Paris Cite, 75010 Paris, France; (M.A.); (J.D.); (E.A.); (M.L.); (J.L.); (S.F.); (S.P.); (N.M.)
| | - Julien Lion
- Human Immunology, Pathophysiology and Immunotherapy, INSERM U 976, University Paris Cite, 75010 Paris, France; (M.A.); (J.D.); (E.A.); (M.L.); (J.L.); (S.F.); (S.P.); (N.M.)
| | - Sofiane Fodil
- Human Immunology, Pathophysiology and Immunotherapy, INSERM U 976, University Paris Cite, 75010 Paris, France; (M.A.); (J.D.); (E.A.); (M.L.); (J.L.); (S.F.); (S.P.); (N.M.)
| | - Stéphanie Pons
- Human Immunology, Pathophysiology and Immunotherapy, INSERM U 976, University Paris Cite, 75010 Paris, France; (M.A.); (J.D.); (E.A.); (M.L.); (J.L.); (S.F.); (S.P.); (N.M.)
| | - Nuala Mooney
- Human Immunology, Pathophysiology and Immunotherapy, INSERM U 976, University Paris Cite, 75010 Paris, France; (M.A.); (J.D.); (E.A.); (M.L.); (J.L.); (S.F.); (S.P.); (N.M.)
| | - Lara Zafrani
- Human Immunology, Pathophysiology and Immunotherapy, INSERM U 976, University Paris Cite, 75010 Paris, France; (M.A.); (J.D.); (E.A.); (M.L.); (J.L.); (S.F.); (S.P.); (N.M.)
- Medical Intensive Care Unit, Assistance Publique des Hôpitaux de Paris, Saint Louis Hospital, 75010 Paris, France
- Correspondence:
| |
Collapse
|
11
|
Discovery and pharmacological characterization of cetrelimab (JNJ-63723283), an anti-programmed cell death protein-1 (PD-1) antibody, in human cancer models. Cancer Chemother Pharmacol 2022; 89:515-527. [PMID: 35298699 PMCID: PMC8956561 DOI: 10.1007/s00280-022-04415-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 02/07/2022] [Indexed: 12/25/2022]
Abstract
Purpose Preclinical characterization of cetrelimab (JNJ-63723283), a fully humanized immunoglobulin G4 kappa monoclonal antibody targeting programmed cell death protein-1 (PD-1), in human cancer models. Methods Cetrelimab was generated by phage panning against human and cynomolgus monkey (cyno) PD-1 extracellular domains (ECDs) and affinity maturation. Binding to primate and rodent PD-1 ECDs, transfected and endogenous cell-surface PD-1, and inhibition of ligand binding were measured. In vitro activity was evaluated using cytomegalovirus recall, mixed lymphocyte reaction, staphylococcal enterotoxin B stimulation, and Jurkat-PD-1 nuclear factor of activated T cell reporter assays. In vivo activity was assessed using human PD-1 knock-in mice implanted with MC38 tumors and a lung patient-derived xenograft (PDX) model (LG1306) using CD34 cord-blood-humanized NSG mice. Pharmacodynamics, toxicokinetics, and safety were assessed in cynos following single and/or repeat intravenous dosing. Results Cetrelimab showed high affinity binding to human (1.72 nM) and cyno (0.90 nM) PD-1 and blocked binding of programmed death-ligand 1 (PD-L1; inhibitory concentration [IC] 111.7 ng/mL) and PD-L2 (IC 138.6 ng/mL). Cetrelimab dose-dependently increased T cell-mediated cytokine production and stimulated cytokine expression. Cetrelimab 10 mg/kg reduced mean MC38 tumor volume in PD-1 knock-in mice at Day 21 (P < 0.0001) versus control. In a PDX lung model, 10 mg/kg cetrelimab (every 5 days for six cycles) increased frequency of peripheral T cells and reduced (P < 0.05) mean tumor volume versus control. Activity was consistent with that of established PD-1 inhibitors. Cetrelimab dosing was well tolerated in cynos and mean drug exposure increase was dose-dependent. Conclusion Cetrelimab potently inhibits PD-1 in vitro and in vivo, supporting its clinical evaluation.
Collapse
|
12
|
Bazhin AV, von Ahn K, Fritz J, Bunge H, Maier C, Isayev O, Neff F, Siveke JT, Karakhanova S. Pivotal antitumor role of the immune checkpoint molecule B7-H1 in pancreatic cancer. Oncoimmunology 2022; 11:2043037. [PMID: 35251770 PMCID: PMC8890402 DOI: 10.1080/2162402x.2022.2043037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Immune checkpoint molecule B7-H1 plays a decisive immune regulatory role in different pathologies including cancer, and manipulation of B7-H1 expression became an attractive approach in cancer immunotherapy. Pancreatic cancer (PDAC) is characterized by pronounced immunosuppressive environment and B7-H1 expression correlates with PDAC prognosis. However, the first attempts to diminish B7-H1 expression in patients were not so successful. This points the complicity of PDAC immunosuppressive network and requires further examinations. We investigated the effect of B7-H1 deficiency in PDAC. Our results clearly show that partial or complete B7-H1 inhibition in vivo let to reduced tumor volume and improved survival of PDAC-bearing mice. This oncological benefit is due to the abrogation of immunosuppression provided by MDSC, macrophages, DC and Treg, which resulted in simultaneous restoration of anti-tumor immune response, namely improved accumulation and functionality of effector-memory CD4 and CD8 T cells. Our results underline the potential of B7-H1 molecule to control immunosuppressive network in PDAC and provide new issues for further clinical investigations.
Collapse
Affiliation(s)
- Alexandr V. Bazhin
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Katharina von Ahn
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Jasmin Fritz
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Henriette Bunge
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Caroline Maier
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Orkhan Isayev
- Department of Cytology, Embryology and Histology, Azerbaijan Medical University, Baku, Azerbaijan
| | - Florian Neff
- Division of Solid Tumor Translational Oncology, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), partner site University Hospital Essen, Heidelberg, Germany
| | - Jens T. Siveke
- Division of Solid Tumor Translational Oncology, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), partner site University Hospital Essen, Heidelberg, Germany
- Bridge Institute of Experimental Tumor Therapy, West German Cancer Center, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Svetlana Karakhanova
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| |
Collapse
|
13
|
Defining Diffuse Large B-Cell Lymphoma Immunotypes by CD8+ T Cells and Natural Killer Cells. JOURNAL OF ONCOLOGY 2022; 2022:3168172. [PMID: 35237321 PMCID: PMC8885174 DOI: 10.1155/2022/3168172] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 01/12/2022] [Indexed: 01/03/2023]
Abstract
Background There is a poor prognosis for diffuse large B-cell lymphoma (DLBCL), one of the most common types of non-Hodgkin lymphoma (NHL). Through gene expression profiles, this study intends to reveal potential subtypes among patients with DLBCL by evaluating their prognostic impact on immune cells. Methods Immune subtypes were developed based on CD8+ T cells and natural killer cells calculated from gene expression profiles. The comparison of prognoses and enriched pathways was made between immune subtypes. Following this validation step, samples from the independent data set were analyzed to determine the correlation between immune subtype and prognosis and immune checkpoint blockade (ICB) response. To provide a model to predict the DLBCL immune subtypes, machine learning methods were used. The virtual screening and molecular docking were adopted to identify small molecules to target the immune subtype biomarkers. Results A training data set containing 432 DLBCL samples from five data sets and a testing dataset containing 420 DLBCL samples from GSE10846 were used to develop and validate immune subtypes. There were two novel immune subtypes identified in this study: an inflamed subtype (IS) and a noninflamed subtype (NIS). When compared with NIS, IS was associated with higher levels of immune cells and a better prognosis for immunotherapy. Based on the random forest algorithm, a robust machine learning model has been established by 12 hub genes, and the area under the curve (AUC) value is 0.948. Three small molecules were selected to target NIS biomarkers, including VGF, RAD54L, and FKBP8. Conclusion This study assessed immune cells as prognostic factors in DLBCL, constructed an immune subtype that could be used to identify patients who would benefit from ICB, and constructed a model to predict the immune subtype.
Collapse
|
14
|
Lee SA, Liu F, Yun DY, Riordan SM, Tay ACY, Liu L, Lee CS, Zhang L. Campylobacter concisus upregulates PD-L1 mRNA expression in IFN-γ sensitized intestinal epithelial cells and induces cell death in esophageal epithelial cells. J Oral Microbiol 2021; 13:1978732. [PMID: 34552702 PMCID: PMC8451702 DOI: 10.1080/20002297.2021.1978732] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 08/20/2021] [Accepted: 09/06/2021] [Indexed: 12/23/2022] Open
Abstract
Introduction: Campylobacter concisus is an oral bacterium that is associated with inflammatory bowel disease (IBD) and Barrett's esophagus (BE). Programmed cell death ligand-1 (PD-L1) is an immune checkpoint protein that is used by tumor cells for immune evasion and has increased expression in patients with IBD and BE. We examined whether C. concisus upregulates PD-L1 expression in intestinal and esophageal epithelial cells. Methods: Human intestinal epithelial HT-29 cells and esophageal epithelial FLO-1 cells with and without interferon (IFN)-γ sensitization were incubated with C. concisus strains. The level of PD-L1 mRNA was quantified using quantitative real-time PCR. Cytokines were measured using Enzyme-Linked Immunosorbent Assay (ELISA). Apoptosis of HT-29 and FLO-1 cells were measured using caspase 3/7 assay. Results: We found that intestinal epithelial cells with IFN-γ sensitization incubated with C. concisus significantly upregulated PD-L1 expression and significantly increased the production of interleukin (IL)-8. Whereas, PD-L1 expression was significantly inhibited in IFN-γ sensitized FLO-1 cells incubated with C. concisus strains. Furthermore, FLO-1 cells with and without IFN-γ sensitization incubated with C. concisus strains both had significantly higher levels of cell death. Conclusion: C. concisushas the potential to cause damage to both intestinal and esophageal epithelial cells, however, with different pathogenic effects.
Collapse
Affiliation(s)
- Seul A Lee
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, Australia
| | - Fang Liu
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, Australia
| | - Doo Young Yun
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, Australia
| | - Stephen M Riordan
- Gastrointestinal and Liver Unit,Prince of Wales Hospital, University of New South Wales, Sydney, Australia
| | - Alfred Chin Yen Tay
- Helicobacter Research Laboratory, Marshall Centre for Infectious Diseases Research and Training, School of Pathology and Laboratory Medicine, University of Western Australia, Perth, Australia
| | - Lu Liu
- School of Medical Sciences, University of New South Wales, Sydney, Australia
| | - Cheok Soon Lee
- School of Medicine, Western Sydney University, Sydney, Australia
- South Western Sydney Clinical School, University of New South Wales, Sydney, Australia
- Central Clinical School, University of Sydney, Sydney, Australia
- Department of Anatomical Pathology, Liverpool Hospital, Sydney, Australia
| | - Li Zhang
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, Australia
| |
Collapse
|
15
|
Righi I, Vaira V, Morlacchi LC, Croci GA, Rossetti V, Blasi F, Ferrero S, Nosotti M, Rosso L, Clerici M. Immune Checkpoints Expression in Chronic Lung Allograft Rejection. Front Immunol 2021; 12:714132. [PMID: 34489963 PMCID: PMC8418069 DOI: 10.3389/fimmu.2021.714132] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 07/26/2021] [Indexed: 11/13/2022] Open
Abstract
Chronic lung allograft dysfunction (CLAD) is the main cause of poor survival and low quality of life of lung transplanted patients. Several studies have addressed the role of dendritic cells, macrophages, T cells, donor specific as well as anti-HLA antibodies, and interleukins in CLAD, but the expression and function of immune checkpoint molecules has not yet been analyzed, especially in the two CLAD subtypes: BOS (bronchiolitis obliterans syndrome) and RAS (restrictive allograft syndrome). To shed light on this topic, we conducted an observational study on eight consecutive grafts explanted from patients who received lung re-transplantation for CLAD. The expression of a panel of immune molecules (PD1/CD279, PDL1/CD274, CTLA4/CD152, CD4, CD8, hFoxp3, TIGIT, TOX, B-Cell-Specific Activator Protein) was analyzed by immunohistochemistry in these grafts and in six control lungs. Results showed that RAS compared to BOS grafts were characterized by 1) the inversion of the CD4/CD8 ratio; 2) a higher percentage of T lymphocytes expressing the PD-1, PD-L1, and CTLA4 checkpoint molecules; and 3) a significant reduction of exhausted PD-1-expressing T lymphocytes (PD-1pos/TOXpos) and of exhausted Treg (PD-1pos/FOXP3pos) T lymphocytes. Results herein, although being based on a limited number of cases, suggest a role for checkpoint molecules in the development of graft rejection and offer a possible immunological explanation for the worst prognosis of RAS. Our data, which will need to be validated in ampler cohorts of patients, raise the possibility that the evaluation of immune checkpoints during follow-up offers a prognostic advantage in monitoring the onset of rejection, and suggest that the use of compounds that modulate the function of checkpoint molecules could be evaluated in the management of chronic rejection in LTx patients.
Collapse
Affiliation(s)
- Ilaria Righi
- Thoracic Surgery and Lung Transplantation Unit, Fondazione IRCCS Ca' Granda-Ospedale Maggiore Policlinico, Milan, Italy
| | - Valentina Vaira
- Division of Pathology, Fondazione IRCCS Ca' Granda-Ospedale Maggiore Policlinico, Milan, Italy.,Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - Letizia Corinna Morlacchi
- Respiratory Unit and Adult Cystic Fibrosis Center, Internal Medicine Department, Fondazione IRCCS Ca' Granda-Ospedale Maggiore Policlinico, Milan, Italy
| | - Giorgio Alberto Croci
- Division of Pathology, Fondazione IRCCS Ca' Granda-Ospedale Maggiore Policlinico, Milan, Italy.,Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - Valeria Rossetti
- Respiratory Unit and Adult Cystic Fibrosis Center, Internal Medicine Department, Fondazione IRCCS Ca' Granda-Ospedale Maggiore Policlinico, Milan, Italy
| | - Francesco Blasi
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy.,Respiratory Unit and Adult Cystic Fibrosis Center, Internal Medicine Department, Fondazione IRCCS Ca' Granda-Ospedale Maggiore Policlinico, Milan, Italy
| | - Stefano Ferrero
- Division of Pathology, Fondazione IRCCS Ca' Granda-Ospedale Maggiore Policlinico, Milan, Italy.,Department of Biomedical, Surgical and Dental Sciences, University of Milan, Milan, Italy
| | - Mario Nosotti
- Thoracic Surgery and Lung Transplantation Unit, Fondazione IRCCS Ca' Granda-Ospedale Maggiore Policlinico, Milan, Italy.,Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - Lorenzo Rosso
- Thoracic Surgery and Lung Transplantation Unit, Fondazione IRCCS Ca' Granda-Ospedale Maggiore Policlinico, Milan, Italy.,Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - Mario Clerici
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy.,Don C. Gnocchi Foundation, IRCCS, Milan, Italy
| |
Collapse
|
16
|
Wang C, Li X, Zhang L, Chen Y, Dong R, Zhang J, Zhao J, Guo X, Yang G, Li Y, Gu C, Xi Q, Zhang R. miR-194-5p down-regulates tumor cell PD-L1 expression and promotes anti-tumor immunity in pancreatic cancer. Int Immunopharmacol 2021; 97:107822. [PMID: 34098485 DOI: 10.1016/j.intimp.2021.107822] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Revised: 05/09/2021] [Accepted: 05/24/2021] [Indexed: 02/08/2023]
Abstract
Pancreatic cancer is a highly malignant cancer of the digestive tract. Studies have shown that in some types of cancer, a high level of microRNA-194-5p (miR-194-5p) is beneficial for controlling tumor progression, while in other cancers it plays a completely opposite role. However, how miR-194-5p affects anti-tumor immunity of pancreatic cancer remains unclear. In this study, we found that high expression of miR-194-5p in human pancreatic cancer patients is associated with a better survival rate, while increased expression of programmed cell death ligand 1 (PD-L1) in human pancreatic cancer patients is associated with a worse survival rate. In pancreatic cancer, the expression level of PD-L1 is negatively correlated with the expression level of miR-194-5p, and we identified that PD-L1 was target gene of miR-194-5p. In addition, we found that overexpression of miR-194-5p inhibited the migration, invasion and proliferation of pancreatic cancer cells in vitro. The orthotopic mouse model of pancreatic cancer shown that miR-194-5p suppressed the progression of pancreatic cancer, promoted the infiltration of CD8+ T cells in tumor immune microenvironments, and enhanced the IFN-γ production of CD8+ T cells. Consistently, the co-culture experiments showed that overexpression of miR-194-5p in tumor cell enhanced IFN-γ production by CD8+ T cells. In conclusion, miR-194-5p may serve as a novel immunotherapeutic target for pancreatic ductal adenocarcinoma (PDAC) by inhibiting the expression of PD-L1, and play important roles in inhibiting the progression of pancreatic cancer and boosting the anti-tumor effect of CD8+ T cells.
Collapse
Affiliation(s)
- Chengzhi Wang
- Department of Immunology, Key Laboratory of Immune Microenvironment and Diseases of Educational Ministry of China, School of Basic Sciences, Tianjin Medical University, Tianjin, China
| | - Xin Li
- Guangdong Province Key Laboratory for Biotechnology Drug Candidates, Institute of Basic Medical Sciences and Department of Biotechnology, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| | - Lijuan Zhang
- Department of Immunology, Key Laboratory of Immune Microenvironment and Diseases of Educational Ministry of China, School of Basic Sciences, Tianjin Medical University, Tianjin, China
| | - Ying Chen
- Guangdong Province Key Laboratory for Biotechnology Drug Candidates, Institute of Basic Medical Sciences and Department of Biotechnology, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| | - Ruijie Dong
- Department of Immunology, Key Laboratory of Immune Microenvironment and Diseases of Educational Ministry of China, School of Basic Sciences, Tianjin Medical University, Tianjin, China
| | - Jieyou Zhang
- Department of Immunology, Key Laboratory of Immune Microenvironment and Diseases of Educational Ministry of China, School of Basic Sciences, Tianjin Medical University, Tianjin, China
| | - Jingyi Zhao
- Department of Immunology, Key Laboratory of Immune Microenvironment and Diseases of Educational Ministry of China, School of Basic Sciences, Tianjin Medical University, Tianjin, China
| | - Xiangdong Guo
- Department of Immunology, Key Laboratory of Immune Microenvironment and Diseases of Educational Ministry of China, School of Basic Sciences, Tianjin Medical University, Tianjin, China
| | - Guangze Yang
- Department of Immunology, Key Laboratory of Immune Microenvironment and Diseases of Educational Ministry of China, School of Basic Sciences, Tianjin Medical University, Tianjin, China
| | - Yan Li
- Guangdong Province Key Laboratory for Biotechnology Drug Candidates, Institute of Basic Medical Sciences and Department of Biotechnology, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| | - Chao Gu
- Department of Immunology, Key Laboratory of Immune Microenvironment and Diseases of Educational Ministry of China, School of Basic Sciences, Tianjin Medical University, Tianjin, China
| | - Qing Xi
- The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China; School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou, China.
| | - Rongxin Zhang
- Guangdong Province Key Laboratory for Biotechnology Drug Candidates, Institute of Basic Medical Sciences and Department of Biotechnology, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China; Key Laboratory of Immune Microenvironment and Diseases of Educational Ministry of China, Tianjin Medical University, Tianjin, China.
| |
Collapse
|
17
|
Xing Y, Liu J, Sun S, Ming T, Wang Y, Luo J, Xiao G, Li X, Xie J, Cai X. New electrochemical method for programmed death-ligand 1 detection based on a paper-based microfluidic aptasensor. Bioelectrochemistry 2021; 140:107789. [PMID: 33677221 DOI: 10.1016/j.bioelechem.2021.107789] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 02/02/2021] [Accepted: 02/13/2021] [Indexed: 12/22/2022]
Abstract
As programmed death-ligand 1 (PD-L1) is considered a referenced therapeutic biomarker, a rapid and low-cost method to detect PD-L1 in body fluids is necessary. In this work, a paper-based microfluidic aptasensor for label-free electrochemical detection of PD-L1 in liquids was fabricated. The aptasensor integrates a reaction cell and a three-electrode system, and a differential pulse voltammetry electrochemical method was adopted. PD-L1 aptamer with a low equilibrium dissociation constant was used as a biorecognition molecule. To bind the aptamer and assist in the electrochemical measurement, nanocomposites were synthesized and used to modify the working electrode, which was composed of an amine-functionalized single-walled carbon nanotube, new methylene blue and gold nanoparticles. The basic performance of the aptasensor was tested in phosphate-buffered saline: the linear range was between 10 pg mL-1 and 2.5 ng mL-1, and the detection limit was 10 pg mL-1 (signal/noise = 3). Moreover, the aptasensor was used for the detection of serum samples and compared with an enzyme linked immunosorbent assay (ELISA). The results showed that the aptasensor provides a new low-cost, portable and highly sensitive detection method for PD-L1, as an alternative to ELISA.
Collapse
Affiliation(s)
- Yu Xing
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing 100190, PR China; University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Juntao Liu
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing 100190, PR China; University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Shuai Sun
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing 100190, PR China; University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Tao Ming
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing 100190, PR China; University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Yang Wang
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing 100190, PR China; University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Jinping Luo
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing 100190, PR China; University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Guihua Xiao
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing 100190, PR China; University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Xinrong Li
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing 100190, PR China; University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Jingyu Xie
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing 100190, PR China; University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Xinxia Cai
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing 100190, PR China; University of Chinese Academy of Sciences, Beijing 100049, PR China.
| |
Collapse
|
18
|
Xiang X, Wang J, Lu D, Xu X. Targeting tumor-associated macrophages to synergize tumor immunotherapy. Signal Transduct Target Ther 2021; 6:75. [PMID: 33619259 PMCID: PMC7900181 DOI: 10.1038/s41392-021-00484-9] [Citation(s) in RCA: 520] [Impact Index Per Article: 130.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 07/30/2020] [Accepted: 10/15/2020] [Indexed: 02/08/2023] Open
Abstract
The current treatment strategies in advanced malignancies remain limited. Notably, immunotherapies have raised hope for a successful control of these advanced diseases, but their therapeutic responses are suboptimal and vary considerably among individuals. Tumor-associated macrophages (TAMs) are a major component of the tumor microenvironment (TME) and are often correlated with poor prognosis and therapy resistance, including immunotherapies. Thus, a deeper understanding of the complex roles of TAMs in immunotherapy regulation could provide new insight into the TME. Furthermore, targeting of TAMs is an emerging field of interest due to the hope that these strategies will synergize with current immunotherapies. In this review, we summarize recent studies investigating the involvement of TAMs in immune checkpoint inhibition, tumor vaccines and adoptive cell transfer therapies, and discuss the therapeutic potential of targeting TAMs as an adjuvant therapy in tumor immunotherapies.
Collapse
Affiliation(s)
- Xiaonan Xiang
- Department of Hepatobiliary and Pancreatic Surgery, Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
- NHC Key Laboratory of Combined Multi-organ Transplantation, Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, CAMS, Hangzhou, China
| | - Jianguo Wang
- Department of Hepatobiliary and Pancreatic Surgery, Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
- NHC Key Laboratory of Combined Multi-organ Transplantation, Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, CAMS, Hangzhou, China
| | - Di Lu
- Department of Hepatobiliary and Pancreatic Surgery, Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
- NHC Key Laboratory of Combined Multi-organ Transplantation, Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, CAMS, Hangzhou, China
| | - Xiao Xu
- Department of Hepatobiliary and Pancreatic Surgery, Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- NHC Key Laboratory of Combined Multi-organ Transplantation, Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, CAMS, Hangzhou, China.
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
19
|
Orafaie A, Sadeghian H, Bahrami AR, Rafatpanah H, Matin MM. Design, synthesis and evaluation of PD-L1 peptide antagonists as new anticancer agents for immunotherapy. Bioorg Med Chem 2020; 30:115951. [PMID: 33360579 DOI: 10.1016/j.bmc.2020.115951] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 12/07/2020] [Accepted: 12/09/2020] [Indexed: 12/15/2022]
Abstract
Blocking the interaction of programmed cell death protein 1 (PD-1) and its ligand PD-L1 is known as a promising immunotherapy for treatment of a variety of tumors expressing PD-L1 on their cell surface. In the last decade, several antibodies against the PD-1/PD-L1 interaction have been approved, while there are few reports of small-molecule inhibitors against PD-1/PD-L1 axis. Due to many advantages of cancer treatment with small molecules over antibodies, we developed several peptidic PD-L1 antagonists using computational peptide design methods, and evaluated them both in vitro and in vivo. Importantly, among six peptides with best affinity to PD-L1, four peptides exhibited significant potency to block PD-1/PD-L1 axis at molecular level. Moreover, the PD-L1 expression in nine human colorectal cancer cell lines stimulated with interferon-γ was compared and LoVo cells with the highest expression were selected for further experiments. The peptides could also restore the function of activated Jurkat T cells, which had been suppressed by stimulated LoVo cells. A blockade assay in tumor-bearing mice experiments indicated that peptides HS5 and HS6 consisting of a d-amino acid in their structures, could also effectively reduce tumor growth in vivo, without induction of any observable liver or renal toxicity, tissue damages and loss of body weight. As new designed peptides showed no toxicity against murine colon cancer cells in vitro, the observed anti-tumor results in mice are most probably due to disrupting the PD-1/PD-L1 interaction. Thus, peptides described in this study can be considered as proper low molecular weight candidates for immunotherapy of cancer.
Collapse
Affiliation(s)
- Ala Orafaie
- Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Hamid Sadeghian
- Antimicrobial Resistance Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Laboratory Sciences, School of Paramedical Sciences, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Ahmad Reza Bahrami
- Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran; Industrial Biotechnology Research Group, Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Houshang Rafatpanah
- Immunology Research Center, Inflammation and Inflammatory Diseases Division, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Maryam M Matin
- Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran; Novel Diagnostics and Therapeutics Research Group, Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran.
| |
Collapse
|
20
|
Lee SA, Wang Y, Liu F, Riordan SM, Liu L, Zhang L. Escherichia coli K12 Upregulates Programmed Cell Death Ligand 1 (PD-L1) Expression in Gamma Interferon-Sensitized Intestinal Epithelial Cells via the NF-κB Pathway. Infect Immun 2020; 89:e00618-20. [PMID: 33046511 PMCID: PMC7927934 DOI: 10.1128/iai.00618-20] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 10/06/2020] [Indexed: 01/09/2023] Open
Abstract
Programmed cell death ligand-1 (PD-L1) is an immune checkpoint protein which is used by tumor cells for immune evasion. PD-L1 is upregulated in inflamed intestinal tissues. The intestinal tract is colonized by millions of bacteria, most of which are commensal bacterial species. We hypothesized that under inflammatory conditions, some commensal bacterial species contribute to increased PD-L1 expression in intestinal epithelium and examined this hypothesis. Human intestinal epithelial HT-29 cells with and without interferon (IFN)-γ sensitization were incubated with six strains of four enteric bacterial species. The mRNA and protein levels of PD-L1 in HT-29 cells were examined using quantitative real-time PCR and flow cytometry, respectively. The levels of interleukin (IL)-1β, IL-18, IL-6, IL-8, and tumor necrosis factor (TNF)-α secreted by HT-29 cells were measured using enzyme-linked immunosorbent assay. Apoptosis of HT-29 cells was measured using a caspase 3/7 assay. We found that Escherichia coli K12 significantly upregulated both PD-L1 mRNA and protein in IFN-γ-sensitized HT-29 cells. E. coli K12 induced the production of IL-8 in HT-29 cells, however, IL-8 did not affect HT-29 PD-L1 expression. Inhibition of the nuclear factor-kappa B pathway significantly reduced E. coli K12-induced PD-L1 expression in HT-29 cells. The other two E. coli strains and two enteric bacterial species did not significantly affect PD-L1 expression in HT-29 cells. Enterococcus faecalis significantly inhibited PD-L1 expression due to induction of cell death. Data from this study suggest that some gut bacterial species have the potential to affect immune function under inflammatory conditions via upregulating epithelial PD-L1 expression.
Collapse
Affiliation(s)
- Seul A Lee
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, Australia
| | - Yiming Wang
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, Australia
- Infection & Immunity Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
| | - Fang Liu
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, Australia
| | - Stephen M Riordan
- Gastrointestinal and Liver Unit, Prince of Wales Hospital, University of New South Wales, Sydney, Australia
| | - Lu Liu
- School of Medical Sciences, University of New South Wales, Sydney, Australia
| | - Li Zhang
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, Australia
| |
Collapse
|
21
|
Plasmodium sporozoites induce regulatory macrophages. PLoS Pathog 2020; 16:e1008799. [PMID: 32898164 PMCID: PMC7500643 DOI: 10.1371/journal.ppat.1008799] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 09/18/2020] [Accepted: 07/10/2020] [Indexed: 12/23/2022] Open
Abstract
Professional antigen-presenting cells (APCs), like macrophages (Mϕs) and dendritic cells (DCs), are central players in the induction of natural and vaccine-induced immunity to malaria, yet very little is known about the interaction of SPZ with human APCs. Intradermal delivery of whole-sporozoite vaccines reduces their effectivity, possibly due to dermal immunoregulatory effects. Therefore, understanding these interactions could prove pivotal to malaria vaccination. We investigated human APC responses to recombinant circumsporozoite protein (recCSP), SPZ and anti-CSP opsonized SPZ both in monocyte derived MoDCs and MoMϕs. Both MoDCs and MoMϕs readily took up recCSP but did not change phenotype or function upon doing so. SPZ are preferentially phagocytosed by MoMϕs instead of DCs and phagocytosis greatly increased after opsonization. Subsequently MoMϕs show increased surface marker expression of activation markers as well as tolerogenic markers such as Programmed Death-Ligand 1 (PD-L1). Additionally they show reduced motility, produce interleukin 10 and suppressed interferon gamma (IFNγ) production by antigen specific CD8+ T cells. Importantly, we investigated phenotypic responses to SPZ in primary dermal APCs isolated from human skin explants, which respond similarly to their monocyte-derived counterparts. These findings are a first step in enhancing our understanding of pre-erythrocytic natural immunity and the pitfalls of intradermal vaccination-induced immunity. Malaria continues to be the deadliest parasitic disease worldwide, and an effective vaccine yielding sterile immunity does not yet exist. Attenuated parasites can induce sterile protection in both human and rodent models for malaria, but these vaccines need to be administered directly into the bloodstream in order to convey protection; administration via the skin results in a much-reduced efficacy. We hypothesized this is caused by an early immune regulation initiated at the first site of contact with the immune system: the skin. However, the human skin stage of malaria has not been investigated to date. We used human antigen presenting cells as well as whole human skin explants to investigate (dermal) immune responses and found that Plasmodium sporozoites are able to suppress immune responses by inducing regulatory macrophages. Our study provides new insights in the mechanism of early immune regulation exploited by Plasmodium parasites and can help to explain why intradermal vaccination using whole attenuated sporozoites results in reduced protection.
Collapse
|
22
|
Chen IC, Sethy B, Liou JP. Recent Update of HDAC Inhibitors in Lymphoma. Front Cell Dev Biol 2020; 8:576391. [PMID: 33015069 PMCID: PMC7494784 DOI: 10.3389/fcell.2020.576391] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 08/18/2020] [Indexed: 12/12/2022] Open
Abstract
Modulating epigenetic modification has been recognized for over a decade as an effective therapeutic approach to cancer and many studies of histone deacetylase (HDAC), one of the best known epigenetic modulators, have been published. HDAC modulates cell proliferation and angiogenesis and plays an essential role in cell growth. Research shows that up-regulated HDACs are present in many cancer types and synthetic or natural HDAC inhibitors have been used to silence overregulated HDACs. Inhibiting HDACs may cause arrest of cell proliferation, angiogenesis reduction and cell apoptosis. Recent studies indicate that HDAC inhibitors can provide a therapeutic effect in various cancers, such as B-cell lymphoma, leukemia, multiple myeloma and some virus-associated cancers. Some evidence has demonstrated that HDAC inhibitors can increase the expression of immune-related molecules leading to accumulation of CD8 + T cells and causing unresponsive tumor cells to be recognized by the immune system, reducing tumor immunity. This may be a solution for the blockade of PD-1. Here, we review the emerging development of HDAC inhibitors in various cancer treatments and reduction of tumor immunity.
Collapse
Affiliation(s)
- I-Chung Chen
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Bidyadhar Sethy
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Jing-Ping Liou
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
23
|
Fishman JA, Hogan JI, Maus MV. Inflammatory and Infectious Syndromes Associated With Cancer Immunotherapies. Clin Infect Dis 2020; 69:909-920. [PMID: 30520987 DOI: 10.1093/cid/ciy1025] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Accepted: 11/30/2018] [Indexed: 12/11/2022] Open
Abstract
Immunotherapy using antibodies to immune checkpoint molecules or targeted chimeric antigen receptor-modified T cells (CAR-T cells) represent dramatic advances in cancer treatment. These therapies mediate immune-related adverse events that may mimic or amplify infectious presentations. Checkpoint inhibitor therapy may be associated with diverse irAEs including mild skin, endocrine, and autoimmune manifestations or severe inflammatory processes including colitis, pneumonitis, myocarditis, and shock. CAR-T-cell therapies may induce toxicities including cytokine-release syndrome with fevers and multiorgan dysfunction, CAR-T-cell-related encephalopathy syndrome with altered mental status and neurologic dysfunction, or hemophagocytic lymphohistiocytosis-macrophage-activation syndrome. Infectious risks may relate to prior cancer therapies or to treatments of inflammatory dysregulation, including corticosteroids and inhibitors of tumor necrosis factor-α and interleukin-6. Immune activation may unmask subclinical infections. Clinical approaches must attempt to identify infections in the face of immunotherapy-associated inflammatory processes. Empirical antimicrobial therapies should not be delayed based on the presumption of noninfectious syndromes.
Collapse
Affiliation(s)
- Jay A Fishman
- Transplant Infectious Disease and Compromised Host Program and Transplant Center, Massachusetts General Hospital, Boston.,Department of Medicine, Harvard Medical School, Boston
| | - John I Hogan
- Transplant Infectious Disease and Compromised Host Program and Transplant Center, Massachusetts General Hospital, Boston
| | - Marcela V Maus
- Department of Medicine, Harvard Medical School, Boston.,Cellular Immunotherapy, Massachusetts General Hospital Cancer Center, Boston
| |
Collapse
|
24
|
Bellenghi M, Puglisi R, Pontecorvi G, De Feo A, Carè A, Mattia G. Sex and Gender Disparities in Melanoma. Cancers (Basel) 2020; 12:E1819. [PMID: 32645881 PMCID: PMC7408637 DOI: 10.3390/cancers12071819] [Citation(s) in RCA: 86] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 06/22/2020] [Accepted: 07/03/2020] [Indexed: 12/19/2022] Open
Abstract
Worldwide, the total incidence of cutaneous melanoma is higher in men than in women, with some differences related to ethnicity and age and, above all, sex and gender. Differences exist in respect to the anatomic localization of melanoma, in that it is more frequent on the trunk in men and on the lower limbs in women. A debated issue is if-and to what extent-melanoma development can be attributed to gender-specific behaviors or to biologically intrinsic differences. In the search for factors responsible for the divergences, a pivotal role of sex hormones has been observed, although conflicting results indicate the involvement of other mechanisms. The presence on the X chromosome of numerous miRNAs and coding genes playing immunological roles represents another important factor, whose relevance can be even increased by the incomplete X chromosome random inactivation. Considering the known advantages of the female immune system, a different cancer immune surveillance efficacy was suggested to explain some sex disparities. Indeed, the complexity of this picture emerged when the recently developed immunotherapies unexpectedly showed better improvements in men than in women. Altogether, these data support the necessity of further studies, which consider enrolling a balanced number of men and women in clinical trials to better understand the differences and obtain actual gender-equitable healthcare.
Collapse
Affiliation(s)
- Maria Bellenghi
- Center for Gender-specific Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (M.B.); (R.P.); (G.P.); (G.M.)
| | - Rossella Puglisi
- Center for Gender-specific Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (M.B.); (R.P.); (G.P.); (G.M.)
| | - Giada Pontecorvi
- Center for Gender-specific Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (M.B.); (R.P.); (G.P.); (G.M.)
| | - Alessandra De Feo
- Laboratory of Experimental Oncology, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy;
| | - Alessandra Carè
- Center for Gender-specific Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (M.B.); (R.P.); (G.P.); (G.M.)
| | - Gianfranco Mattia
- Center for Gender-specific Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (M.B.); (R.P.); (G.P.); (G.M.)
| |
Collapse
|
25
|
Wang S, Wang G, Zhang L, Li F, Liu K, Wang Y, Shi Y, Cao K. Interleukin-17 promotes nitric oxide-dependent expression of PD-L1 in mesenchymal stem cells. Cell Biosci 2020; 10:73. [PMID: 32509271 PMCID: PMC7249370 DOI: 10.1186/s13578-020-00431-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2019] [Accepted: 05/13/2020] [Indexed: 12/12/2022] Open
Abstract
Background Interleukin-17A (IL-17) is an evolutionary conserved cytokine and best known for its role in boosting immune response. However, recent clinical researches showed that abundant IL-17 in tumor microenvironment was often associated with poor prognosis and reduced cytotoxic T cell infiltration. These contradictory phenomena suggest that IL-17 may have unique target cells in tumor microenvironment which switch its biological consequences from pro-inflammatory to anti-inflammatory. Mesenchymal stem/stromal cells (MSCs) are a major component of the tumor microenvironment. Upon cytokine stimulation, MSCs can express a plenary of inhibitory molecules, playing a critical role in tumor development and progression. Therefore, we aim to investigate the role of IL-17 in MSC-mediated immunosuppression. Results We found IFNγ and TNFα, two major cytokines in tumor microenvironment, could induce programmed death-ligand 1 (PD-L1) expression in MSCs. Interestingly, IL-17 has a synergistic effect with IFNγ and TNFα in elevating PD-L1 expression in MSCs. The presence of IL-17 empowered MSCs with strong immunosuppression abilities and enabled MSCs to promote tumor progression in a PD-L1 dependent manner. The upregulated PD-L1 expression in MSCs was due to the accumulation of nitric oxide (NO). On one hand, NO donor could mimic the effects of IL-17 on MSCs; on the other hand, IL-17 failed to enhance PD-L1 expression in inducible nitric oxide synthase (iNOS) deficient MSCs or with iNOS inhibitor presence. Conclusions Our study demonstrates that IL-17 can significantly increase the expression of PD-L1 by MSCs through iNOS induction. This IL-17-MSCs-PD-L1 axis shapes the immunosuppressive tumor microenvironment and facilitates tumor progression.
Collapse
Affiliation(s)
- Shijia Wang
- Key Laboratory of Stem Cell Biology, Shanghai Jiao Tong University School of Medicine (SJTUSM) & Shanghai Institutes for Biological Sciences (SIBS), Academy of Sciences (CAS), Shanghai, 200031 China
| | - Guan Wang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031 China
| | - Liying Zhang
- The First Affiliated Hospital of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Soochow University, Suzhou, 215123 China
| | - Fengying Li
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031 China
| | - Keli Liu
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031 China
| | - Ying Wang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031 China
| | - Yufang Shi
- Key Laboratory of Stem Cell Biology, Shanghai Jiao Tong University School of Medicine (SJTUSM) & Shanghai Institutes for Biological Sciences (SIBS), Academy of Sciences (CAS), Shanghai, 200031 China.,CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031 China.,The First Affiliated Hospital of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Soochow University, Suzhou, 215123 China
| | - Kai Cao
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031 China
| |
Collapse
|
26
|
Wang P, Li C, Ma X, Gai X. Clinical significance of the combined measurement of serum B7-H1 and interleukin-10 in colorectal cancer patients. Medicine (Baltimore) 2020; 99:e20044. [PMID: 32358386 PMCID: PMC7440053 DOI: 10.1097/md.0000000000020044] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Colorectal cancer (CRC) patients have been shown to express a cytokine signature that is detectable in serum and contributes to cancer pathogenesis. The objective of this study was to evaluate the potential clinical significance of preoperative circulating cytokine levels in CRC patients.The expression of serum B7-H1 and IL-10 was assessed by ELISA in 89 patients and 64 health volunteers. As a control marker, CEA serum levels were measured by electrochemical luminescence detection. The receiver operating characteristic (ROC) curve was used to analysis to demonstrate the potential diagnostic value of these biomarkers.The expression of serum B7-H1 was significantly increased in CRC patients (P = .001) and associated with the progression of TNM stage and a positive association with serum IL-10 levels was also evident. Furthermore, serum B7-H1 and IL-10 expression was not influenced by age, gender, tumor location, or mass, whereas a relationship existed with tumor metastasis and TNM stage. The serum levels of B7-H1 and IL-10 on the 7th postoperative day were significantly decreased compared with that of preoperative serum levels (P = .001, P = .003 respectively). The area under the ROC curves (AUC) for B7-H1 and IL-10 were 0.7063 and 0.5706, respectively. The optimal sensitivity and specificity of B7-H1 for discriminating between colon cancer patients and healthy controls were 85.21% and 56.43%, respectively, using a cut-off value of 3.46 ng/mL. However, the combined ROC analysis using B7-H1 and IL-10 revealed an AUC of 0.8791, with a sensitivity of 90.63% and a specificity of 75.18%.The outcomes of the present study demonstrate the clinical significance of serum B7-H1 and IL-10 concentrations. Combined detection of B7-H1 plus IL-10 showed significantly increased sensitivity and specificity for discriminating between colorectal cancer patients and healthy controls compared these markers detection individual. The measurement of B7-H1 or IL-10 in sera following surgery may provide an additional tool for assessing the curative effects of surgery in CRC patients.
Collapse
Affiliation(s)
- Peng Wang
- Department of Pathology
- Key Laboratory of Molecular Medicine, Beihua University, Jilin Province, China
| | - Chun Li
- Department of Pathology
- Key Laboratory of Molecular Medicine, Beihua University, Jilin Province, China
| | | | - Xiaodong Gai
- Department of Pathology
- Key Laboratory of Molecular Medicine, Beihua University, Jilin Province, China
| |
Collapse
|
27
|
|
28
|
Kalim M, Iqbal Khan MS, Zhan J. Programmed cell death ligand-1: A dynamic immune checkpoint in cancer therapy. Chem Biol Drug Des 2020; 95:552-566. [PMID: 32166894 DOI: 10.1111/cbdd.13677] [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: 10/03/2019] [Revised: 02/24/2020] [Accepted: 02/29/2020] [Indexed: 12/18/2022]
Abstract
Antibody-based immunotherapies play a pivotal role in cancer research with efficient achievements in tumor suppression. Tumor survival is assisted by modulation of immune checkpoints to create imbalances between immune cells and cancer cell's environment. The modulation results in T-cell signal inhibition ultimately inert its proliferation and activation against various tumor cells. PD-L1, a 40 kDa transmembrane protein of B7 family, binds with PD-1 on the membrane of T cells which results in inhibition of T-cell proliferation and activation. PD-L1/PD-1 pathway has generated novel target sites for antibodies that can block PD-L1/PD-1 interactions. The blockage results in T-cell proliferation and tumor cell suppression. The PD-L1 immune checkpoint strategies' development, expression and regulations, signal inhibitions, and developmental stages of PD-L1/PD-1 antibodies are briefly discussed here in this review. All this information will provide a base for new therapeutic development against PD-L1 and PD-1 immune checkpoint interactions and will make available promising treatment options.
Collapse
Affiliation(s)
- Muhammad Kalim
- Department of Biochemistry, Cancer Institute of the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Muhammad Saleem Iqbal Khan
- Department of Biochemistry, Cancer Institute of the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Jinbiao Zhan
- Department of Biochemistry, Cancer Institute of the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
29
|
Li G, Yang L, Li D, Zhang J, Du L, Xia L, Liu Y, Hu W. Effects of combined treatment with PD‑L1 Ig and CD40L mAb on immune tolerance in the CBA/J x DBA/2 mouse model. Mol Med Rep 2020; 21:1789-1798. [PMID: 32319625 PMCID: PMC7057827 DOI: 10.3892/mmr.2020.10977] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 01/01/2020] [Indexed: 12/31/2022] Open
Abstract
The embryo is a natural allograft and is the only exception to immune rejection, which reflects maternal immune tolerance towards the embryo. However, pregnancy loss is primarily caused by maternal immune rejection of the embryo. The aim of the present study was to explore the effects of combined treatment of programmed death-ligand 1 (PD-L1) immunoglobulin (Ig) and CD40-ligand (CD40L) monoclonal antibody (mAb) on immune tolerance in an abortion-prone mating model. Mice were divided into the normal, spontaneous abortion, PD-L1 Ig, CD40L mAb and the PD-L1 Ig + CD40L mAb groups. On day 14 of gestation, the embryo resorption abortion rates of all the groups was observed. The maternal hypo-responsiveness to paternal antigens was determined using a mixed lymphocyte response and the splenic CD4+CD25+ T-cell population, major histocompatibility complex (MHC)-II+, CD80+ and CD86+ cell populations in pregnant female CBA/J mice were analyzed using flow cytometry. The expression levels of intracellular cytokines in the splenic tissues of pregnant CBA/J female mice were analyzed using western blotting. The PD-L1 Ig + CD40L group displayed the lowest resorption rate compared with the other groups. A significant decrease in the proliferative response of maternal splenic immunocompetent cells against paternal antigens, and a significant increase in the proliferative response of maternal splenic CD4+CD25+ T cells was observed in the PD-L1 Ig + CD40L group compared with the spontaneous abortion group. The number of MHC-II+, CD80+ and CD86+ bone marrow-derived dendritic cells (DCs) generated by female mice, and the levels of tumor necrosis factor-α and interferon-γ in the spleens of female mice were significantly decreased in the PD-L1 Ig + CD40L mAb group compared with the spontaneous abortion group. By contrast, interleukin-4 levels were significantly increased in the PD-L1 Ig + CD40L mAb group compared with the spontaneous abortion group. The results suggested that the administration of PD-L1 Ig + CD40L mAb on day 4 of gestation, the period of peri-implantation, may induce paternal antigen-specific immunotolerance, leading to the embryo resorption rate of the abortion-prone model being similar to that of the normal pregnancy model. The results indicate that the combined treatment of PD-L1 Ig and anti-CD40L mAbs may serve as a potential therapeutic for pregnancy loss.
Collapse
Affiliation(s)
- Guanfei Li
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650101, P.R. China
| | - Lihua Yang
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650101, P.R. China
| | - Dan Li
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650101, P.R. China
| | - Jinhong Zhang
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650101, P.R. China
| | - Ling Du
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650101, P.R. China
| | - Libin Xia
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650101, P.R. China
| | - Yunhua Liu
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650101, P.R. China
| | - Wanqin Hu
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650101, P.R. China
| |
Collapse
|
30
|
Zhong C, Lang Q, Yu J, Wu S, Xu F, Tian Y. Phenotypical and potential functional characteristics of different immune cells expressing CD28H/B7-H5 and their relationship with cancer prognosis. Clin Exp Immunol 2020; 200:12-21. [PMID: 31901178 DOI: 10.1111/cei.13413] [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] [Accepted: 12/21/2019] [Indexed: 02/06/2023] Open
Abstract
CD28H and B7-H5 have been identified as receptor-ligand pairs in the B7/CD28 family, and have co-stimulatory activity in immune cells. Here, we have systematically reviewed the research reports concerning the CD28H/B7-H5 pathway. It was found that CD28H is mainly expressed in T cells and natural killer (NK) cells with naive and poorly differentiated properties, and repeated antigen stimulation leads to permanent loss of CD28H. In tumors, CD28H is mainly expressed in tissue-resident memory (TRM ) lymphocyte T cells, which is associated with improved tumor prognosis. B7-H5 is a ligand for CD28H and is widely expressed in tumor cells. B7-H5 expression is closely related to the prognosis of the tumor. Studies have shown that high expression of B7-H5 in tumor is related to a worse prognosis for lung cancer, osteosarcoma, oral squamous cell carcinoma (OSCC), breast carcinoma, human clear cell renal cell carcinoma (ccRCC), intrahepatic cholangiocarcinoma (ICC), bladder urothelial carcinoma (BUC) and colorectal cancer (CRC), but is associated with a better prognosis for pancreatic ductal adenocarcinoma (PDAC) and glioma. Controversial views exist in studies on gastric cancer prognosis.
Collapse
Affiliation(s)
- C Zhong
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Q Lang
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - J Yu
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - S Wu
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - F Xu
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Y Tian
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
31
|
Huang XM, Zhang NR, Lin XT, Zhu CY, Zou YF, Wu XJ, He XS, He XW, Wan YL, Lan P. Antitumor immunity of low-dose cyclophosphamide: changes in T cells and cytokines TGF-beta and IL-10 in mice with colon-cancer liver metastasis. Gastroenterol Rep (Oxf) 2020; 8:56-65. [PMID: 32104586 PMCID: PMC7034239 DOI: 10.1093/gastro/goz060] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 10/02/2019] [Accepted: 10/10/2019] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND The tumor immune microenvironment is one of the most important prognostic factors in liver metastasis from colorectal cancer. Low-dose cyclophosphamide (CTX) is widely believed to be involved in the modulation of the immune system. However, the underlying mechanism of low-dose CTX remains unknown. This study aimed to investigate the antitumor immunity of low-dose CTX in the treatment of colon-cancer liver metastasis. METHODS Thirty mice were randomly divided into five groups. After liver metastasis was established in colon-cancer models, mice in the treatment groups were injected with low-dose CTX (20 mg/kg) at different time points. Liver and spleen tissues were examined for T-cell markers via flow cytometry. Interleukin (IL)-10 and transforming growth factor (TGF)-β1 expression levels in liver tissues were analysed by immunohistochemistry. Serum interferon (IFN)-γ and IL-10 levels were detected by enzyme-linked immunosorbent assay. An additional 20 mice were randomly allocated into two groups and the survival times were recorded. RESULTS The expression levels of CD4+ T cells, CD8+ T cells, and IFN-γ were down-regulated, whereas those of IL-10 and TGF-β1 were up-regulated in liver metastasis from colon cancer in mice. Furthermore, the local and systemic microenvironments of the liver were altered, which led to reduced antitumor immune responses and subsequently liver metastasis. However, treatment with low-dose CTX reversed these effects. The survival times of mice treated with low-dose CTX were significantly longer than those of the other groups. CONCLUSIONS Low-dose CTX exerts its antitumor activity by changing the systemic and local immune microenvironments and enhancing immune regulation in mice. CTX could be used as a drug to prevent and treat liver metastasis from colon cancer.
Collapse
Affiliation(s)
- Xiao-Ming Huang
- Department of Hepatobiliary Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, P. R. China
- Guangdong Institute of Gastroenterology; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, P. R. China
| | - Nan-Rong Zhang
- Guangdong Institute of Gastroenterology; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, P. R. China
- Department of Anesthesiology, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, P. R. China
| | - Xu-Tao Lin
- Guangdong Institute of Gastroenterology; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, P. R. China
- Department of Gastrointestinal Endoscopy, Department of Colorectal Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, P. R. China
| | - Cai-Yan Zhu
- Guangdong Institute of Gastroenterology; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, P. R. China
- Department of Pharmacy, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, P. R. China
| | - Yi-Feng Zou
- Guangdong Institute of Gastroenterology; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, P. R. China
- Department of Colorectal Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, P. R. China
| | - Xiao-Jian Wu
- Guangdong Institute of Gastroenterology; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, P. R. China
- Department of Colorectal Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, P. R. China
| | - Xiao-Sheng He
- Guangdong Institute of Gastroenterology; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, P. R. China
- Department of Colorectal Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, P. R. China
| | - Xiao-Wen He
- Guangdong Institute of Gastroenterology; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, P. R. China
- Department of Colorectal Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, P. R. China
| | - Yun-Le Wan
- Department of Hepatobiliary Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, P. R. China
- Guangdong Institute of Gastroenterology; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, P. R. China
| | - Ping Lan
- Guangdong Institute of Gastroenterology; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, P. R. China
- Department of Colorectal Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, P. R. China
| |
Collapse
|
32
|
Abstract
Immune checkpoint inhibitors have quickly become a critical component to the management of advanced renal cell carcinoma. These therapies have been approved for patients who are treatment-naive and who have progressed on antiangiogenesis agents. Combinations of immune checkpoint inhibitors with antiangiogenesis agents show significant response rates and prolong survival. Adverse events associated with the use of checkpoint inhibition present unique challenges in the management of patients, and careful considerations are needed when checkpoint inhibitors are combined with antiangiogenesis agents. Nevertheless, the improvement in overall survival associated with these agents indicates that they will remain a vital component of kidney cancer treatment.
Collapse
Affiliation(s)
- Mamta Parikh
- Division of Hematology Oncology, Department of Internal Medicine, School of Medicine, University of California, Davis, Sacramento, CA.
| | - Poornima Bajwa
- Department of Internal Medicine, School of Medicine, University of California, Davis, Sacramento, CA
| |
Collapse
|
33
|
Tumor Microenvironment in Diffuse Large B-Cell Lymphoma: Role and Prognosis. Anal Cell Pathol (Amst) 2019; 2019:8586354. [PMID: 31934533 PMCID: PMC6942707 DOI: 10.1155/2019/8586354] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2019] [Accepted: 11/06/2019] [Indexed: 12/23/2022] Open
Abstract
Diffuse large B-cell lymphoma (DLBCL) represents 30-40% of all non-Hodgkin lymphomas (NHL) and is a disease with an aggressive behavior. Because about one-third of DLBCL patients will be refractory or resistant to standard therapy, several studies focused on identification of new individual prognostic and risk stratification biomarkers and new potential therapeutic targets. In contrast to other types of cancers like carcinomas, where tumor microenvironment was widely investigated, its role in DLBCL pathogenesis and patient survival is still poorly understood, although few studies had promising results. The composition of TME and its interaction with neoplastic cells may explain the role of several genes (beta2-microglobulin gene, CD58 gene), receptor-like programmed cell death-1 (PD-1) and its ligand (PD-L1), or other cell components (Treg) in tumor evasion of immune surveillance, resulting in tumor progression. Also, it was found that “gene expression profile” of the microenvironmental cells, the phenotype of tumor-associated macrophages (TAM), the expression of matricellular proteins like SPARC and fibronectin, the overexpression of several types of matrix metalloproteinases (MMPs) like MMP-2 and MMP-9, or the tissue inhibitors of matrix metalloproteinases (TIMPs) may lead to a favorable or adverse outcome. With this review, we try to highlight the influence of microenvironment components over lymphoid clone progression and their prognostic impact in DLBCL patients.
Collapse
|
34
|
Abid H, Khavandi M, Siddiqui N, Panjawatanan P, Patel A. Incidence and Risk of Thyroid Dysfunction in Advanced or Metastatic Non-small Cell Lung Cancer Patients Treated with Pembrolizumab: A Meta-analysis. Cureus 2019; 11:e5997. [PMID: 31807385 PMCID: PMC6876924 DOI: 10.7759/cureus.5997] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Accepted: 10/25/2019] [Indexed: 12/17/2022] Open
Abstract
Thyroid dysfunction is one of the major side effects associated with Pembrolizumab in the treatment of advanced or metastatic non-small cell lung cancer (NSCLC). We performed a systematic review and meta-analysis of randomized clinical trials to determine its overall incidence. A literature search was conducted using the electronic database engines PubMed and Google Scholar from inception to March 2019. Eligible studies were prospective randomized clinical trials with advanced or metastatic NSCLC. The pooled incidence, risk ratio (RR), and 95% confidence interval (CI) of thyroid dysfunction were calculated using the random-effect model. Given the possibility of a between-study variance, we used the random-effect model rather than the fixed-effect model. A total of four studies, including 1603 patients, were selected for analysis. Among patients receiving Pembrolizumab, the overall incidence of all-grade thyroid dysfunction was 19.8% (95% CI: 16.6-23.3%). Pembrolizumab was associated with a significantly increased risk of thyroid dysfunction of all grades, with a relative risk of 3.9 (95% CI: 2.08-7.42%, p= 0.084) in comparison with the controls. Therefore, there is a significant increase in developing thyroid dysfunction in advanced or metastatic NSCLC patients treated with Pembrolizumab.
Collapse
Affiliation(s)
- Haisam Abid
- Internal Medicine, Bassett Medical Center, Cooperstown, USA
| | | | - Nadir Siddiqui
- Internal Medicine, Bassett Medical Center, Cooperstown, USA
| | | | - Anush Patel
- Hematology / Oncology, Bassett Medical Center, Cooperstown, USA
| |
Collapse
|
35
|
Liu X, Shan C, Song Y, Du J. Prognostic Value of Programmed Cell Death Ligand-1 Expression in Nasopharyngeal Carcinoma: A Meta-Analysis of 1,315 Patients. Front Oncol 2019; 9:1111. [PMID: 31709181 PMCID: PMC6823255 DOI: 10.3389/fonc.2019.01111] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Accepted: 10/07/2019] [Indexed: 12/12/2022] Open
Abstract
Background: The prognostic value of programmed cell death ligand-1 (PD-L1) in patients with nasopharyngeal carcinoma (NPC) remains controversial. Therefore, we conducted this meta-analysis to understand the role of PD-L1 in NPC. Method: We searched PubMed, Embase, Web of Science, and Cochrane Library up to April 2019. We determined the pooled hazard ratio (HR) and 95% confidence intervals (CIs) to assess the relationship between PD-L1 and various survival outcomes. Begg's funnel plot was used to assess any publication bias. Results: Eleven studies involving 1,315 patients were included in this meta-analysis. For overall survival (OS), the HR was 1.48 and 95% CI was 1.00–2.18 (p = 0.049). For disease-free survival (DFS), the HR was 1.51 and 95% CI was 0.85–2.69 (p = 0.162). For distant metastasis-free survival (DMFS), the HR was 1.75 and 95% CI was 0.64–4.79 (p = 0.277). For local recurrence-free survival (LRFS), the HR was 0.67 and 95% CI was 0.06–8.16 (p = 0.756). The results of prognosis of PD-L1 and OS were more significant after sensitivity analysis. The pooled odds ratio indicated that PD-L1 expression was not associated with T stage, N stage, M stage, overall stage, sex, age, smoking, or alcohol intake. No publication bias was found. Conclusion: Our meta-analysis showed that PD-L1 overexpression in NPC was associated with a poor OS and may be useful as a novel prognostic factor for NPC.
Collapse
Affiliation(s)
- Xiaofeng Liu
- Department of Otolaryngology, Children's Hospital of Hebei Province, Shijiazhuang, China
| | - Chunguang Shan
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Yingluan Song
- Department of Otolaryngology, Children's Hospital of Hebei Province, Shijiazhuang, China
| | - Juan Du
- Department of Neurothoracic Surgery, Children's Hospital of Hebei Province, Shijiazhuang, China
| |
Collapse
|
36
|
Vital D, Ikenberg K, Moch H, Rössle M, Huber GF. The expression of PD-L1 in salivary gland carcinomas. Sci Rep 2019; 9:12724. [PMID: 31484986 PMCID: PMC6726636 DOI: 10.1038/s41598-019-49215-9] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Accepted: 08/21/2019] [Indexed: 02/07/2023] Open
Abstract
Objective was to analyze the role of PD-L1 and its relation to demographic, patho-clinical and outcome parameters in salivary gland carcinoma (SGC) patients. Patients treated for salivary gland carcinomas between 1994 and 2010 were included. A retrospective chart review for baseline characteristics, pathohistological, clinical and outcome data was performed. Immunohistochemistry for PD-L1 was performed using tissue microarrays. PD-L1 expression was assessed in tumor cells and tumor-infiltrating immune cells (TIIC) and statistical analysis with regard to baseline and outcome data was performed. Expression of PD-L1 (by means ≥1% of the cells with PD-L1 positivity) was present in the salivary gland carcinoma cells of 17%, in the TIIC of 20% and in both tumor cells and TIIC of 10% the patients. PD-L1 expression in tumor cells and both tumor cells and TIIC was related to tumor grading (p = 0.035 and p = 0.031, respectively). A trend towards higher grading was also seen for PD-L1 expression in TIICs (p = 0.058). Patients with salivary duct carcinomas and PD-L1 expressing TIICs showed a significantly worse DFS and OS (p = 0.022 and p = 0.003, respectively), those with both tumor cells and TIIC expressing PD-L1 a significantly worse DFS (p = 0.030). PD-L1 expression is present in 17% and 20% of salivary gland carcinoma cells and TIIC. Ten percent of the patient showed a PD-L1 positivity in both tumor cells and TIIC. This is related to high tumor grading and therefore might be a negative prognostic factor.
Collapse
Affiliation(s)
- Domenic Vital
- Department of Otorhinolaryngology, Head and Neck Surgery, Zurich University Hospital and University of Zurich, Frauenklinikstrasse 24, CH-8091, Zurich, Switzerland.
| | - Kristian Ikenberg
- Department of Pathology and Molecular Pathology, Zurich University Hospital and University of Zurich, Schmelzbergstrasse 12, CH-8091, Zurich, Switzerland
| | - Holger Moch
- Department of Pathology and Molecular Pathology, Zurich University Hospital and University of Zurich, Schmelzbergstrasse 12, CH-8091, Zurich, Switzerland
| | - Matthias Rössle
- Department of Pathology, Luzerner Kantonsspital, Spitalstrasse, CH-6000, Lucerne 16, Switzerland
| | - Gerhard F Huber
- Department of Otorhinolaryngology, Head and Neck Surgery, Kantonsspital St. Gallen, Rorschacher Strasse 95, CH-9007, St. Gallen, Switzerland
| |
Collapse
|
37
|
Rossi G, Pezzuto A, Sini C, Tuzi A, Citarella F, McCusker MG, Nigro O, Tanda E, Russo A. Concomitant medications during immune checkpoint blockage in cancer patients: Novel insights in this emerging clinical scenario. Crit Rev Oncol Hematol 2019; 142:26-34. [PMID: 31352168 DOI: 10.1016/j.critrevonc.2019.07.005] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 06/30/2019] [Accepted: 07/03/2019] [Indexed: 12/17/2022] Open
Abstract
The use of immune checkpoint inhibitors (ICIs) in cancer patients is rapidly growing. However, the potential impact of some widely used concomitant medications is still largely unclear. Emerging data suggest that gut microbiota may affect the efficacy of ICIs, leading to the hypothesis that concurrent antibiotics and proton pump inhibitors use could have a detrimental effect. In addition, steroid use might potentially impair the activity of immunotherapy, due its known immunosuppressive effects, and some safety concerns have been raised in patients receiving commonly used vaccination during ICIs. However, all randomized trials evaluating ICIs consistently excluded patients receiving high corticosteroid doses and data regarding other concomitant medications are lacking. Recently, several retrospective studies have tried to address this unmet medical need. Herein we discuss the latest evidence on the influence of these medications, critically analyzing the data reported so far and the possible implications in our clinical practice.
Collapse
Affiliation(s)
- Giovanni Rossi
- Lung Cancer Unit-Ospedale Policlinico San Martino-Genova, Italy
| | - Aldo Pezzuto
- Cardiovascular and Respiratory Science, S. Andrea Hospital- Sapienza University Rome, Italy
| | - Claudio Sini
- Oncologia Medica e CPDO ASSL di Olbia-ATS Sardegna, Italy
| | | | | | - Michael G McCusker
- University of Maryland Greenebaum Comprehensive Cancer Center, University of Maryland Medical Center, Baltimore, MD 21201, United States
| | - Olga Nigro
- ASST Sette Laghi, UO Oncologia, Varese, Italy
| | - Enrica Tanda
- Skin Cancer Unit-Ospedale Policlinico San Martino-Genova, Italy
| | - Alessandro Russo
- University of Maryland Greenebaum Comprehensive Cancer Center, University of Maryland Medical Center, Baltimore, MD 21201, United States; Medical Oncology Unit A.O. Papardo & Department of Human Pathology, University of Messina, Italy.
| |
Collapse
|
38
|
Lamano JB, Lamano JB, Li YD, DiDomenico JD, Choy W, Veliceasa D, Oyon DE, Fakurnejad S, Ampie L, Kesavabhotla K, Kaur R, Kaur G, Biyashev D, Unruh DJ, Horbinski CM, James CD, Parsa AT, Bloch O. Glioblastoma-Derived IL6 Induces Immunosuppressive Peripheral Myeloid Cell PD-L1 and Promotes Tumor Growth. Clin Cancer Res 2019; 25:3643-3657. [PMID: 30824583 PMCID: PMC6571046 DOI: 10.1158/1078-0432.ccr-18-2402] [Citation(s) in RCA: 136] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Revised: 01/02/2019] [Accepted: 02/25/2019] [Indexed: 12/21/2022]
Abstract
PURPOSE Upregulation of programmed death-ligand 1 (PD-L1) on circulating and tumor-infiltrating myeloid cells is a critical component of GBM-mediated immunosuppression that has been associated with diminished response to vaccine immunotherapy and poor survival. Although GBM-derived soluble factors have been implicated in myeloid PD-L1 expression, the identity of such factors has remained unknown. This study aimed to identify factors responsible for myeloid PD-L1 upregulation as potential targets for immune modulation. EXPERIMENTAL DESIGN Conditioned media from patient-derived GBM explant cell cultures was assessed for cytokine expression and utilized to stimulate naïve myeloid cells. Myeloid PD-L1 induction was quantified by flow cytometry. Candidate cytokines correlated with PD-L1 induction were evaluated in tumor sections and plasma for relationships with survival and myeloid PD-L1 expression. The role of identified cytokines on immunosuppression and survival was investigated in vivo utilizing immunocompetent C57BL/6 mice bearing syngeneic GL261 and CT-2A tumors. RESULTS GBM-derived IL6 was identified as a cytokine that is necessary and sufficient for myeloid PD-L1 induction in GBM through a STAT3-dependent mechanism. Inhibition of IL6 signaling in orthotopic murine glioma models was associated with reduced myeloid PD-L1 expression, diminished tumor growth, and increased survival. The therapeutic benefit of anti-IL6 therapy proved to be CD8+ T-cell dependent, and the antitumor activity was additive with that provided by programmed death-1 (PD-1)-targeted immunotherapy. CONCLUSIONS Our findings suggest that disruption of IL6 signaling in GBM reduces local and systemic myeloid-driven immunosuppression and enhances immune-mediated antitumor responses against GBM.
Collapse
Affiliation(s)
- Jonathan B Lamano
- Department of Neurological Surgery, Northwestern University, Chicago, Illinois
| | | | - Yuping D Li
- Department of Neurological Surgery, Northwestern University, Chicago, Illinois
| | | | - Winward Choy
- Department of Neurological Surgery, University of California San Francisco, San Francisco, California
| | - Dorina Veliceasa
- Department of Neurological Surgery, Northwestern University, Chicago, Illinois
| | - Daniel E Oyon
- Department of Neurological Surgery, Northwestern University, Chicago, Illinois
| | - Shayan Fakurnejad
- Stanford School of Medicine, Stanford University, Stanford, California
| | - Leonel Ampie
- Department of Neurosurgery, University of Virginia School of Medicine, University of Virginia, Charlottesville, Virginia
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, NIH, Bethesda, Maryland
| | - Kartik Kesavabhotla
- Department of Neurological Surgery, Northwestern University, Chicago, Illinois
| | - Rajwant Kaur
- Department of Neurological Surgery, Northwestern University, Chicago, Illinois
| | - Gurvinder Kaur
- Department of Neurological Surgery, Northwestern University, Chicago, Illinois
| | - Dauren Biyashev
- Department of Neurological Surgery, Northwestern University, Chicago, Illinois
| | - Dusten J Unruh
- Department of Neurological Surgery, Northwestern University, Chicago, Illinois
| | - Craig M Horbinski
- Department of Neurological Surgery, Northwestern University, Chicago, Illinois
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, Illinois
- Lou and Jean Malnati Brain Tumor Institute, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - C David James
- Department of Neurological Surgery, Northwestern University, Chicago, Illinois
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, Illinois
- Lou and Jean Malnati Brain Tumor Institute, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
- Department of Biochemistry and Molecular Genetics, Northwestern University, Chicago, Illinois
| | | | - Orin Bloch
- Department of Neurological Surgery, Northwestern University, Chicago, Illinois.
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, Illinois
- Lou and Jean Malnati Brain Tumor Institute, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| |
Collapse
|
39
|
Wang J, Sun M, Zhu X, Zhao H, Mao D, Zhang Z, Zhao X. Lentivirus-mediated RNA interference targeting programmed death receptor ligand 1 increases the immunologic anti-tumor effect of dendritic cell vaccination against pancreatic cancer in SCID-hu mice. Oncol Lett 2019; 18:1539-1547. [PMID: 31423221 PMCID: PMC6607057 DOI: 10.3892/ol.2019.10426] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Accepted: 04/30/2019] [Indexed: 12/31/2022] Open
Abstract
Programmed death receptor ligand 1 (PD-L1), which belongs to the B7 family, is overexpressed in a variety of human cancer types and serves a crucial role in immune escape by malignant cells. Programmed death receptor 1 (PD-1) is a specific PD-L1 receptor. PD-1/PD-L1 signaling inhibits the antitumor effects of dendritic cell (DC) immunization for tumor treatment. The aim of the present study was to determine whether inhibiting PD-L1 may increase the immunologic anti-tumor effect of dendritic cells against pancreatic cancer. In the present study, PD-L1 levels in non-cancerous and malignant tissue samples were compared, and the impact of PD-L1 downregulation on human pancreatic cancer PaTu8988 cells was determined by lentivirus-based RNA interference and DC immunotherapy. PD-L1 expression in pancreatic specimens was assessed by reverse transcription-quantitative polymerase chain reaction (RT-qPCR) and immunohistochemistry. PaTu8988 cells expressing reduced levels of PD-L1 were generated by lentivirus-based knockdown to assess the mechanism by which the inhibition of PD-L1 signaling in DC immunization affects therapeutic outcomes in pancreatic cancer-bearing SCID-hu mice. PD-L1 levels were markedly elevated in pancreatic adenocarcinoma samples compared with in non-cancerous tissue. PD-L1 silencing in pancreatic adenocarcinoma cells resulted in improved treatment outcomes of DC immunization in vitro and in vivo compared with traditional DC immunization. PD-L1 silencing enhances the antitumor response of cytotoxic T cells by increasing interferon γ production in vitro. In vivo, this method prevented tumor growth and lung metastasis, and prolonged survival in the SCID-hu model. In conclusion, the results of the present study suggested that suppressing PD-L1 in malignant cells during DC immunization may be a useful tool for immunotherapy in pancreatic adenocarcinoma.
Collapse
Affiliation(s)
- Jin Wang
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215000, P.R. China.,Jiangsu Key Laboratory of Gastrointestinal Tumor Immunology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215000, P.R. China.,Jiangsu Institute of Clinical Immunology, Soochow University, Suzhou, Jiangsu 215000, P.R. China.,Jiangsu Key Laboratory of Clinical Immunology, Soochow University, Suzhou, Jiangsu 215000, P.R. China
| | - Mingbing Sun
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215000, P.R. China
| | - Xinguo Zhu
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215000, P.R. China
| | - Hua Zhao
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215000, P.R. China
| | - Deli Mao
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215000, P.R. China
| | - Zhe Zhang
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215000, P.R. China
| | - Xin Zhao
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215000, P.R. China.,Jiangsu Key Laboratory of Gastrointestinal Tumor Immunology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215000, P.R. China.,Jiangsu Institute of Clinical Immunology, Soochow University, Suzhou, Jiangsu 215000, P.R. China.,Jiangsu Key Laboratory of Clinical Immunology, Soochow University, Suzhou, Jiangsu 215000, P.R. China
| |
Collapse
|
40
|
Bartee MY, Dryja PC, Bartee E. Chimeric tumor modeling reveals role of partial PDL1 expression in resistance to virally induced immunotherapy. J Immunother Cancer 2019; 7:11. [PMID: 30651147 PMCID: PMC6335801 DOI: 10.1186/s40425-018-0496-6] [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: 08/22/2018] [Accepted: 12/26/2018] [Indexed: 02/08/2023] Open
Abstract
Expression of PDL1 on the surface of tumor cells can blunt the efficacy of many cancer immunotherapies. For example, our lab has previously shown that tumors derived from malignant cells incapable of expressing PDL1 are highly susceptible to immunotherapy induced by oncolytic virus treatment while tumors derived from PDL1 capable cells are highly resistant. In patient biopsies, however, expression of PDL1 on malignant cells is often not uniform with some cells expressing PDL1 while others do not. Importantly, how this partial PDL1 positivity influences the outcomes of immunotherapy remains largely unknown. In the current work, we expand on our previous findings by generating partially PDL1 positive tumors in immune competent animals and asking what percentage of tumor cells must express PDL1 for a tumor to become functionally resistant to oncolytic treatment. Our results indicate that the responsiveness of partially PDL1+ tumors correlates linearly with the percentage of PDL1 capable cells present at the initiation of treatment. Additionally, we observe that tumors which relapse after treatment display a significant increase in the numbers of PDL1 capable cells present suggesting that specific editing of mixed tumors might play a role in disease relapse. These data indicate that varying levels of PDL1 expression can play a significant role in the outcomes of oncolytic immunotherapy and challenges the concept that tumors should be viewed as simply PDL1+ or PDL1−.
Collapse
Affiliation(s)
- Mee Y Bartee
- Department of Microbiology and Immunology, Medical University of South Carolina, Basic Science Building Rm 208C, 173 Ashley Ave, Charleston, SC, 29425, USA
| | - Parker C Dryja
- Department of Microbiology and Immunology, Medical University of South Carolina, Basic Science Building Rm 208C, 173 Ashley Ave, Charleston, SC, 29425, USA
| | - Eric Bartee
- Department of Microbiology and Immunology, Medical University of South Carolina, Basic Science Building Rm 208C, 173 Ashley Ave, Charleston, SC, 29425, USA.
| |
Collapse
|
41
|
Zhao JJ, Zhou ZQ, Wang P, Chen CL, Liu Y, Pan QZ, Zhu Q, Tang Y, Weng DS, Xia JC. Orchestration of immune checkpoints in tumor immune contexture and their prognostic significance in esophageal squamous cell carcinoma. Cancer Manag Res 2018; 10:6457-6468. [PMID: 30568505 PMCID: PMC6276823 DOI: 10.2147/cmar.s181949] [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] [Indexed: 01/05/2023] Open
Abstract
Introduction Esophageal squamous cell carcinoma (ESCC) develops in a background of chronic inflammation; therefore, it is a promising candidate for treatment by immunotherapy. Although tumor immunity is critically involved in tumor growth and metastasis in ESCC, important gaps exist in our understanding of its immune microenvironment. This study aimed to investigate the expression and prognostic significance of immune checkpoint proteins in ESCC and the associated T-cell densities. Materials and methods We investigated the infiltration of CD8+ T cells and the expressions of immune checkpoint proteins (PD-1, TIGIT, PD-L1, and PD-L2) in 154 primary ESCC patients by immunohistochemistry. The correlation of immune checkpoint proteins' expression and clinical outcomes was determined by Kaplan-Meier test and multivariate Cox regression analysis. Results PD-L1 and PD-L2 expression were detected in 45.5 and 59.7% of the ESCC samples, respectively. The high densities of PD-1+ and TIGIT+ tumor-infiltrating lymphocytes (TILs) were expressed in 47.4 and 49.4% of the ESCC patients, respectively. The number of PD-1+ TILs was significantly positively correlated with CD8+ TILs (P<0.001). Cases displaying high PD-L1 expression exhibited consistently high CD8+ T-cell infiltration (P=0.0157). Increased numbers of PD-1+ and TIGIT+ TILs alone or both, as well as PD-L1 and PD-L2 expression alone or both, were significantly and associated with a shorter overall survival among these patients. The combined analysis of the expression of PD-1, TIGIT, PD-L1, and PD-L2 found that a group of patients with PD-1+/TIGIT+ TILs and PD-L1- and/or PD-L2-positive tumor cells had the worst prognosis in primary ESCC. Conclusion These immune profiles of checkpoint proteins expression should guide the selection of ESCC patients to receive suitable immunotherapies.
Collapse
Affiliation(s)
- Jing-Jing Zhao
- Department of Experimental Research, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China, ; .,Department of Biotherapy, Sun Yat-Sen University Cancer Center, Guangzhou, China, ;
| | - Zi-Qi Zhou
- Department of Experimental Research, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China, ; .,Department of Biotherapy, Sun Yat-Sen University Cancer Center, Guangzhou, China, ;
| | - Peng Wang
- Department of Emergency Medicine, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China.,Department of Medical Research, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Chang-Long Chen
- Department of Experimental Research, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China, ; .,Department of Biotherapy, Sun Yat-Sen University Cancer Center, Guangzhou, China, ;
| | - Yuan Liu
- Department of Experimental Research, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China, ; .,Department of Biotherapy, Sun Yat-Sen University Cancer Center, Guangzhou, China, ;
| | - Qiu-Zhong Pan
- Department of Experimental Research, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China, ; .,Department of Biotherapy, Sun Yat-Sen University Cancer Center, Guangzhou, China, ;
| | - Qian Zhu
- Department of Experimental Research, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China, ; .,Department of Biotherapy, Sun Yat-Sen University Cancer Center, Guangzhou, China, ;
| | - Yan Tang
- Department of Experimental Research, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China, ; .,Department of Biotherapy, Sun Yat-Sen University Cancer Center, Guangzhou, China, ;
| | - De-Sheng Weng
- Department of Experimental Research, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China, ; .,Department of Biotherapy, Sun Yat-Sen University Cancer Center, Guangzhou, China, ;
| | - Jian-Chuan Xia
- Department of Experimental Research, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China, ; .,Department of Biotherapy, Sun Yat-Sen University Cancer Center, Guangzhou, China, ;
| |
Collapse
|
42
|
Ding L, Chen X, Xu X, Qian Y, Liang G, Yao F, Yao Z, Wu H, Zhang J, He Q, Yang B. PARP1 Suppresses the Transcription of PD-L1 by Poly(ADP-Ribosyl)ating STAT3. Cancer Immunol Res 2018; 7:136-149. [PMID: 30401677 DOI: 10.1158/2326-6066.cir-18-0071] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Revised: 08/10/2018] [Accepted: 11/01/2018] [Indexed: 11/16/2022]
Abstract
Studies have pointed to a role of PARP1 in regulating gene expression through poly(ADP-ribosyl)ating, sequence-specific, DNA-binding transcription factors. However, few examples exist that link this role of PARP1 to the immunogenicity of cancer cells. Here, we report that PARP1 poly(ADP-ribosyl)ates STAT3 and subsequently promotes STAT3 dephosphorylation, resulting in reduced transcriptional activity of STAT3 and expression of PD-L1. In this study, we showed that PARP1 silencing or pharmacologic inhibition enhanced the transcription of PD-L1 in cancer cells, which was accompanied by the upregulation of PD-L1 protein expression, both in the cytoplasm and on the cell surface. This induction of PD-L1 was attenuated in the absence of the transcription factor STAT3. Cell-based studies indicated that PARP1 interacted directly with STAT3 and caused STAT3 poly(ADP-ribosyl)ation. STAT3's activation of PD-L1 transcription was abolished by the overexpression of wild-type PARP1 but not mutant PARP1, which lacks catalytic activity. PARP1 downregulation or catalytic inhibition enhanced the phosphorylation of STAT3, which was reversed by the ectopic expression of wild-type PARP1 but not by mutated PARP1. An inverse correlation between PARP1 and PD-L1 was also observed in clinical ovarian cancer samples. Overall, our study revealed PARP1-mediated poly(ADP-ribosyl)ation of STAT3 as a key step in inhibiting the transcription of PD-L1, and this mechanism exists in a variety of cancer cells.
Collapse
Affiliation(s)
- Ling Ding
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Xi Chen
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Xiaqing Xu
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Yuli Qian
- Department of Reproductive Endocrinology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Guikai Liang
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Fengqi Yao
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Zhangting Yao
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Honghai Wu
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Jieqiong Zhang
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Qiaojun He
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China.
| | - Bo Yang
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China.
| |
Collapse
|
43
|
Winkel BMF, Dalenberg MR, de Korne CM, Feijt C, Langenberg MCC, Pelgrom L, Ganesh MS, Yazdanbakhsh M, Smits HH, de Jong EC, Everts B, van Leeuwen FWB, Hokke CH, Roestenberg M. Early Induction of Human Regulatory Dermal Antigen Presenting Cells by Skin-Penetrating Schistosoma Mansoni Cercariae. Front Immunol 2018; 9:2510. [PMID: 30429854 PMCID: PMC6220649 DOI: 10.3389/fimmu.2018.02510] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 10/10/2018] [Indexed: 12/14/2022] Open
Abstract
Following initial invasion of Schistosoma mansoni cercariae, schistosomula reside in the skin for several days during which they can interact with the dermal immune system. While murine experiments have indicated that exposure to radiation-attenuated (RA) cercariae can generate protective immunity which is initiated in the skin stage, contrasting non-attenuated cercariae, such data is missing for the human model. Since murine skin does not form a reliable marker for immune responses in human skin, we used human skin explants to study the interaction with non-attenuated and RA cercariae with dermal innate antigen presenting cells (APCs) and the subsequent immunological responses. We exposed human skin explants to cercariae and visualized their invasion in real time (initial 30 min) using novel imaging technologies. Subsequently, we studied dermal immune responses and found an enhanced production of regulatory cytokine interleukin (IL)-10, pro-inflammatory cytokine IL-6 and macrophage inflammatory protein (MIP)-1α within 3 days of exposure. Analysis of dermal dendritic cells (DDCs) for their phenotype revealed an increased expression of immune modulators programmed death ligand (PD-L) 1 and 2, and increased IL-10 production. Ex vivo primed DDCs suppress Th1 polarization of naïve T-cells and increase T-cell IL-10 production, indicating their regulatory potential. These immune responses were absent or decreased after exposure to RA parasites. Using transwells, we show that direct contact between APCs and cercariae is required to induce their regulatory phenotype. To the best of our knowledge this is the first study that attempts to provide insight in the human dermal S. mansoni cercariae invasion and subsequent immune responses comparing non-attenuated with RA parasites. We reveal that cercariae induce a predominantly regulatory immune response whereas RA cercariae fail to achieve this. This initial understanding of the dermal immune suppressive capacity of S. mansoni cercariae in humans provides a first step toward the development of an effective schistosome vaccine.
Collapse
Affiliation(s)
- Béatrice M F Winkel
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands.,Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Center, Leiden, Netherlands
| | - Mirjam R Dalenberg
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands
| | - Clarize M de Korne
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands.,Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Center, Leiden, Netherlands
| | - Carola Feijt
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands
| | | | - Leonard Pelgrom
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands
| | - Munisha S Ganesh
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands
| | - Maria Yazdanbakhsh
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands
| | | | - Esther C de Jong
- Department of Cell Biology and Histology, Academic Medical Center, Amsterdam, Netherlands
| | - Bart Everts
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands
| | - Fijs W B van Leeuwen
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands
| | - Cornelis H Hokke
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands
| | - Meta Roestenberg
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands.,Department of Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
44
|
Dong Z, Zhang L, Xu W, Zhang G. EGFR may participate in immune evasion through regulation of B7‑H5 expression in non‑small cell lung carcinoma. Mol Med Rep 2018; 18:3769-3779. [PMID: 30106102 PMCID: PMC6131583 DOI: 10.3892/mmr.2018.9361] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Accepted: 01/09/2018] [Indexed: 01/02/2023] Open
Abstract
Lung cancer is one of the most prevalent malignancies worldwide; it has been ranked the most lethal type of cancer. Non‑small cell lung carcinoma (NSCLC) comprises >80% of all types of lung cancer. Although certain achievements have been made in the treatment of NSCLC, including the targeted gene drug epidermal growth factor receptor‑tyrosine kinase inhibitor (EGFR‑TKI), the five‑year survival rate of patients has not significantly increased. A previous study demonstrated that B7‑H5, a novel co‑stimulatory molecule in the B7 molecule family, was negatively correlated with EGFR in pancreatic cancer. Thus, in the present study, we aimed to investigate whether EGFR participates in immune evasion, probably through regulation of B7‑H5 expression. NCI‑H1299 NSCLCL cells were divided into control, mock, small interfering‑EGFR and EGFR‑TKI groups. The cell viability and apoptosis rate were analysed by a Cell Counting Kit‑8 assay and flow cytometry. The transforming growth factor (TGF)‑β and interleukin (IL)‑10 content was measured using an ELISA. The expression levels of EGFR, B7‑H5, Survivin, apoptosis regulator Bax, apoptosis regulator Bcl‑2 (Bcl‑2), TGF‑β, vascular endothelial growth factor (VEGF), IL‑10 and cyclooxygenase (COX)‑2 were assessed via quantitative PCR and western blotting. The activation of the tyrosine‑protein kinase JAK2 (JAK2)/signal transducer and activator of transcription 3 (STAT3) signalling pathway was detected using western blotting. The results demonstrated a notable negative correlation between EGFR and B7‑H5 expression levels in cancer tissues and cell lines. Inhibition of EGFR expression via gene silencing and EGFR inhibition markedly decreased cell viability and increased the apoptosis of NCI‑H1299 cells, by upregulating survivin and Bcl‑2 expression. The protein expression levels of TGF‑β, VEGF, IL‑10 and COX‑2 were additionally decreased, with weak activation of the JAK2/STAT3 signalling pathway. EGFR may be involved in immune evasion, possibly through regulation of B7‑H5 expression in NSCLC.
Collapse
Affiliation(s)
- Zhaohui Dong
- Intensive Care Unit, The First People's Hospital of Huzhou, Huzhou, Zhejiang 313000, P.R. China
| | - Lanying Zhang
- Intensive Care Unit, The First People's Hospital of Huzhou, Huzhou, Zhejiang 313000, P.R. China
| | - Wei Xu
- Intensive Care Unit, The First People's Hospital of Huzhou, Huzhou, Zhejiang 313000, P.R. China
| | - Gensheng Zhang
- Intensive Care Unit, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, P.R. China
| |
Collapse
|
45
|
Ryu R, Ward KE. Atezolizumab for the First-Line Treatment of Non-small Cell Lung Cancer (NSCLC): Current Status and Future Prospects. Front Oncol 2018; 8:277. [PMID: 30087855 PMCID: PMC6066722 DOI: 10.3389/fonc.2018.00277] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Accepted: 07/03/2018] [Indexed: 12/31/2022] Open
Abstract
Purpose: Atezolizumab is a programmed death ligand 1 (PDL-1) blocking antibody that was approved for metastatic non-small cell lung cancer (NSCLC) in patients with disease progression. Various studies have been initiated to explore the effectiveness of atezolizumab among different patient cohorts and disease statuses, including as first-line therapy. The purpose of this paper is to identify and summarize the trials that use atezolizumab as a first-line agent in chemotherapy-naïve patients with NSCLC. Methods: A database search was performed on Pubmed, Embase, and Wiley Cochrane Library-Central Register of Controlled Trials to identify clinical trials using atezolizumab as first-line therapy in NSCLC. Additionally, ClinicalTrials.gov and the International Clinical Trials Registry Platform (ICTRP) were searched to identify relevant clinical trials. Conference abstracts from the American Society of Clinical Oncology, the European Society for Medical Oncology, and the American Association for Cancer Research were hand-searched. Any trial in which atezolizumab was used as first-line therapy in chemotherapy-naive patients with NSCLC was included. Results: Fifteen studies were ultimately included, all of which are current and ongoing. Of the 15 studies, 5 have reported results. When given in the first-line setting, atezolizumab had higher rates of objective response, progression-free survival, and overall survival, compared to the second and third-line settings. Among the 15 studies, atezolizumab is used as monotherapy (n = 5), in combination with chemotherapy (n = 6), in combination with targeted therapy such as bevacizumab (n = 1), as neoadjuvant/adjuvant therapy (n = 3), in combination with stereotactic body radiation therapy (n = 1), and in combination with or following chemoradiation (n = 1). Conclusion: Available evidence shows promising safety and efficacy with the use of atezolizumab as first-line therapy in NSCLC. Atezolizumab is currently being studied in a variety of treatment settings. If clinical benefits are shown, atezolizumab may deem to be a useful first-line agent in NSCLC.
Collapse
Affiliation(s)
- Rachel Ryu
- Department of Pharmacy Practice, College of Pharmacy, University of Rhode Island, Kingston, RI, United States
| | | |
Collapse
|
46
|
Shi B, Li Q, Ma X, Gao Q, Li L, Chu J. High expression of programmed cell death protein 1 on peripheral blood T-cell subsets is associated with poor prognosis in metastatic gastric cancer. Oncol Lett 2018; 16:4448-4454. [PMID: 30214579 PMCID: PMC6126159 DOI: 10.3892/ol.2018.9190] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2017] [Accepted: 05/24/2018] [Indexed: 12/16/2022] Open
Abstract
Immune checkpoints in solid tumors serve important roles in metastasis. The present study was designed to explore the expression of programmed cell death protein 1 (PD-1) on peripheral blood T-cell subsets and its role in the clinicopathological features and prognosis of patients with metastatic gastric cancer. The expression of PD-1 in peripheral blood T-cell subsets was detected in 100 metastatic gastric cancer patients prior to the first line chemotherapy by flow cytometric analysis. The potential associaton between the peripheral blood T-cell subsets PD-1 level and the clinicopathological features of patients with metastatic gastric cancer and the clinical outcomes was analyzed. The percent of high PD-1 expressed cluster of differentiation (CD)3+, CD3+CD4+ and CD3+CD8+ T-cells was 20.4, 13.0 and 9.4%, respectively in patients with metastatic gastric cancer. The overall survival (OS) and progression-free survival (PFS) rate of the 100 patients with metastatic gastric cancer was 12.2 and 3.9 months, respectively. Kaplan-Meier curve with long-rank analysis indicated that patients with higher PD-1+/CD3+, PD-1+/CD3+CD4+ and PD-1+/CD3+CD8+ levels had a worse prognosis (all P<0.05). Univariate and multivariate analysis revealed that high PD-1+/CD3+ [hazard ratio (HR), 2.145; P=0.015], high PD-1+/CD3+CD4+ (HR, 1.866; P=0.034) and high PD-1+/CD3+CD8+ (HR, 1.817; P=0.033) level in peripheral blood were independent risk factors for predicting the survival time of patients with metastatic gastric cancer. High PD-1+/CD3+, high PD-1+/CD3+CD4+ and high PD-1+/CD3+CD8+ expression conferred a lower overall survival rate in patients with metastatic gastric cancer. These results suggest that high PD-1 expression on peripheral blood T-cell subsets may potentially be novel prognostic biomarker for metastatic gastric cancer.
Collapse
Affiliation(s)
- Bian Shi
- Department of Integrated Traditional Chinese and Western Medical Oncology, Zhengzhou University Affiliated Cancer Hospital, Zhengzhou, Henan 450001, P.R. China
| | - Qiujian Li
- Department of Integrated Traditional Chinese and Western Medical Oncology, Zhengzhou University Affiliated Cancer Hospital, Zhengzhou, Henan 450001, P.R. China
| | - Xuhui Ma
- Department of Integrated Traditional Chinese and Western Medical Oncology, Zhengzhou University Affiliated Cancer Hospital, Zhengzhou, Henan 450001, P.R. China
| | - Qilong Gao
- Department of Integrated Traditional Chinese and Western Medical Oncology, Zhengzhou University Affiliated Cancer Hospital, Zhengzhou, Henan 450001, P.R. China
| | - Lu Li
- Department of Integrated Traditional Chinese and Western Medical Oncology, Zhengzhou University Affiliated Cancer Hospital, Zhengzhou, Henan 450001, P.R. China
| | - Junfeng Chu
- Department of Oncology, Zhengzhou University Affiliated Cancer Hospital, Zhengzhou, Henan 450001, P.R. China
| |
Collapse
|
47
|
Läubli H, Balmelli C, Kaufmann L, Stanczak M, Syedbasha M, Vogt D, Hertig A, Müller B, Gautschi O, Stenner F, Zippelius A, Egli A, Rothschild SI. Influenza vaccination of cancer patients during PD-1 blockade induces serological protection but may raise the risk for immune-related adverse events. J Immunother Cancer 2018; 6:40. [PMID: 29789020 PMCID: PMC5964701 DOI: 10.1186/s40425-018-0353-7] [Citation(s) in RCA: 99] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Accepted: 05/09/2018] [Indexed: 12/12/2022] Open
Abstract
Background Immune checkpoint inhibiting antibodies were introduced into routine clinical practice for cancer patients. Checkpoint blockade has led to durable remissions in some patients, but may also induce immune-related adverse events (irAEs). Lung cancer patients show an increased risk for complications, when infected with influenza viruses. Therefore, vaccination is recommended. However, the efficacy and safety of influenza vaccination during checkpoint blockade and its influence on irAEs is unclear. Similarly, the influence of vaccinations on T cell-mediated immune reactions in patients during PD-1 blockade remains poorly defined. Methods We vaccinated 23 lung cancer patients and 11 age-matched healthy controls using a trivalent inactivated influenza vaccine to investigate vaccine-induced immunity and safety during checkpoint blockade. Results We did not observe significant differences between patients and healthy controls in vaccine-induced antibody titers against all three viral antigens. Influenza vaccination resulted in protective titers in more than 60% of patients/participants. In cancer patients, the post-vaccine frequency of irAEs was 52.2% with a median time to occurrence of 3.2 months after vaccination. Six of 23 patients (26.1%) showed severe grade 3/4 irAEs. This frequency of irAEs might be higher than the rate previously published in the literature and the rate observed in a non-study population at our institution (all grades 25.5%, grade 3/4 9.8%). Conclusions Although this is a non-randomized trial with a limited number of patients, the increased rate of immunological toxicity is concerning. This finding should be studied in a larger patient population.
Collapse
Affiliation(s)
- Heinz Läubli
- Department of Internal Medicine, Division of Medical Oncology, University Hospital Basel, Basel, Switzerland.,Cancer Immunology, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Catharina Balmelli
- Department of Internal Medicine, Division of Medical Oncology, University Hospital Basel, Basel, Switzerland
| | - Lukas Kaufmann
- Applied Microbiology Research, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Michal Stanczak
- Cancer Immunology, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Mohammedyaseen Syedbasha
- Applied Microbiology Research, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Dominik Vogt
- Applied Microbiology Research, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Astrid Hertig
- Department of Internal Medicine, Division of Medical Oncology, University Hospital Basel, Basel, Switzerland
| | - Beat Müller
- Oncology, Cantonal Hospital Lucerne, Lucerne, Switzerland
| | | | - Frank Stenner
- Department of Internal Medicine, Division of Medical Oncology, University Hospital Basel, Basel, Switzerland.,Cancer Immunology, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Alfred Zippelius
- Department of Internal Medicine, Division of Medical Oncology, University Hospital Basel, Basel, Switzerland.,Cancer Immunology, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Adrian Egli
- Applied Microbiology Research, Department of Biomedicine, University of Basel, Basel, Switzerland.,Clinical Microbiology, University Hospital Basel, Basel, Switzerland
| | - Sacha I Rothschild
- Department of Internal Medicine, Division of Medical Oncology, University Hospital Basel, Basel, Switzerland. .,Cancer Immunology, Department of Biomedicine, University of Basel, Basel, Switzerland.
| |
Collapse
|
48
|
Low expression of CD40L in tumor-free lymph node of oral cavity cancer related with poor prognosis. Int J Clin Oncol 2018; 23:851-859. [DOI: 10.1007/s10147-018-1294-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2018] [Accepted: 04/23/2018] [Indexed: 01/04/2023]
|
49
|
Abstract
Immunotherapy has become standard of care in advanced non-small cell lung cancer (NSCLC) in a number of settings. Radiotherapy remains an important and potentially curative treatment for localized and locally advanced NSCLC not amenable to surgery. While the principal cytotoxic effect of ionizing radiation is via DNA damage, the effect on tumour microenvironment, promoting dendritic cell presentation of tumour-derived antigens to T cells stimulating the host adaptive immune system to mount an immune response against tumours cells, has become of particular interest when combining immunomodulating agents with radiation. The 'abscopal effect' of radiation where non-irradiated metastatic lesions may respond to radiation may be immune-mediated, via radiation primed anti-tumour T cells. Immune priming by radiation offers the potential for increasing the efficacy of immunotherapy and this is subject to on-going clinical trials underpinned by immunological bioassays. Increasing understanding of the interaction between tumour, radiation and immune cells at a molecular level provides a further opportunity for intervention to enhance the potential synergy between radiation and immunotherapy. Applying the potential efficacy of combination therapy to clinical practice requires caution particularly to ensure the safety of the two treatment modalities in early phase clinical trials, many of which are currently underway. We review the biological basis for combining radiation and immunotherapy and examine the existing pre-clinical and clinical evidence and the challenges posed by the new combination of treatments.
Collapse
Affiliation(s)
- Neeraj Bhalla
- Clatterbridge Cancer Centre NHS Foundation Trust, Bebington, Wirral, Merseyside, UK
| | - Rachel Brooker
- Clatterbridge Cancer Centre NHS Foundation Trust, Bebington, Wirral, Merseyside, UK
| | - Michael Brada
- Clatterbridge Cancer Centre NHS Foundation Trust, Bebington, Wirral, Merseyside, UK.,Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool, UK
| |
Collapse
|
50
|
Martin AM, Nirschl CJ, Polanczyk MJ, Bell WR, Nirschl TR, Harris-Bookman S, Phallen J, Hicks J, Martinez D, Ogurtsova A, Xu H, Sullivan LM, Meeker AK, Raabe EH, Cohen KJ, Eberhart CG, Burger PC, Santi M, Taube JM, Pardoll DM, Drake CG, Lim M. PD-L1 expression in medulloblastoma: an evaluation by subgroup. Oncotarget 2018; 9:19177-19191. [PMID: 29721192 PMCID: PMC5922386 DOI: 10.18632/oncotarget.24951] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Accepted: 03/13/2018] [Indexed: 01/01/2023] Open
Abstract
Background This study evaluated the expression of PD-L1 and markers of immune mediated resistance in human medulloblastoma (MB), the most common malignant pediatric brain tumor. Results Overall levels of PD-L1 in human MB were low; however, some cases demonstrated robust focal expression associated with increased immune infiltrates. The case with highest PD-L1 expression was a sonic hedgehog (SHH) MB. In cell lines, SHH MB, which are low-MYC expressing, demonstrated both constitutive and inducible expression of PD-L1 while those in Group 3/4 that expressed high levels of MYC had only inducible expression. In vitro, IFN-γ robustly stimulated the expression of PD-L1 in all cell lines while radiation induced variable expression. Forced high MYC expression did not significantly alter PD-L1. Methods Human MB tumor samples were evaluated for expression of PD-L1 and immune cell markers in relation to molecular subgroup assignment. PD-L1 expression was functionally analyzed under conditions of interferon gamma (IFN-γ), radiation, and MYC overexpression. Conclusions MB expresses low levels of PD-L1 facilitating immune escape. Importantly, TH1 cytokine stimulation appears to be the most potent inducer of PD-L1 expression in vitro suggesting that an inflamed tumor microenvironment is necessary for PD-1 pathway activation in this tumor.
Collapse
Affiliation(s)
- Allison M Martin
- Johns Hopkins School of Medicine, Sidney Kimmel Cancer Center, Division of Pediatric Oncology, Baltimore, MD, USA
| | - Christopher J Nirschl
- Johns Hopkins School of Medicine, Sidney Kimmel Cancer Center, Division of Cancer Immunology, Baltimore, MD, USA
| | - Magda J Polanczyk
- Johns Hopkins School of Medicine, Sidney Kimmel Cancer Center, Division of Pediatric Oncology, Baltimore, MD, USA
| | - W Robert Bell
- Department of Laboratory Medicine and Pathology, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Thomas R Nirschl
- Johns Hopkins School of Medicine, Department of Pathobiology, Baltimore, MD, USA
| | - Sarah Harris-Bookman
- Johns Hopkins School of Medicine, Department of Ophthalmology, Baltimore, MD, USA
| | - Jillian Phallen
- Johns Hopkins School of Medicine, Sidney Kimmel Cancer Center, Division of Cancer Biology, Baltimore, MD, USA
| | - Jessica Hicks
- Johns Hopkins School of Medicine, Department of Pathology, Division of Kidney and Urologic Pathology, Baltimore, MD, USA
| | - Daniel Martinez
- Children's Hospital of Philadelphia, Department of Pathology and Laboratory Medicine, Philadelphia, PA, USA
| | - Aleksandra Ogurtsova
- Johns Hopkins School of Medicine, Department of Dermatology, Division of Dermatologic Pathology and Oral Pathology, Baltimore, MD, USA
| | - Haiying Xu
- Johns Hopkins School of Medicine, Department of Dermatology, Division of Dermatologic Pathology and Oral Pathology, Baltimore, MD, USA
| | | | - Alan K Meeker
- Johns Hopkins School of Medicine, Department of Pathology, Division of Kidney and Urologic Pathology, Baltimore, MD, USA
| | - Eric H Raabe
- Johns Hopkins School of Medicine, Sidney Kimmel Cancer Center, Division of Pediatric Oncology, Baltimore, MD, USA.,Johns Hopkins School of Medicine, Department of Pathology, Division of Neuropathology, Baltimore, MD, USA
| | - Kenneth J Cohen
- Johns Hopkins School of Medicine, Sidney Kimmel Cancer Center, Division of Pediatric Oncology, Baltimore, MD, USA
| | - Charles G Eberhart
- Johns Hopkins School of Medicine, Department of Pathology, Division of Neuropathology, Baltimore, MD, USA
| | - Peter C Burger
- Johns Hopkins School of Medicine, Department of Pathology, Division of Neuropathology, Baltimore, MD, USA
| | - Mariarita Santi
- Children's Hospital of Philadelphia, Department of Pathology and Laboratory Medicine, Philadelphia, PA, USA
| | - Janis M Taube
- Johns Hopkins School of Medicine, Department of Dermatology, Division of Dermatologic Pathology and Oral Pathology, Baltimore, MD, USA
| | - Drew M Pardoll
- Johns Hopkins School of Medicine, Sidney Kimmel Cancer Center, Division of Cancer Immunology, Baltimore, MD, USA
| | - Charles G Drake
- Columbia University Medical Center, Division of Hematology/Oncology, New York, NY, USA
| | - Michael Lim
- Johns Hopkins School of Medicine, Department of Neurosurgery, Division of Neurosurgical Oncology, Baltimore, MD, USA
| |
Collapse
|