1
|
Escors D, Chocarro L, Echaide M, Rodriguez-Neira C, Vilaplana B, Kochan G. Programmed Death-1 Ligand 1 Domain Organization, Signaling Motifs, and Interactors in Cancer Immunotherapy. Cancers (Basel) 2025; 17:1635. [PMID: 40427133 PMCID: PMC12110588 DOI: 10.3390/cancers17101635] [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: 03/10/2025] [Revised: 04/29/2025] [Accepted: 05/09/2025] [Indexed: 05/29/2025] Open
Abstract
Immunotherapies targeting the programmed cell death-1 ligand 1 (PD-L1) and programmed cell death 1 (PD-1) pathway sparked a revolution in cancer treatment. These breakthrough therapies work by disrupting the interaction between PD-1-expressed on T cells-and its ligand PD-L1, commonly found on the surface of cancer cells. By using monoclonal antibodies to block this binding, the immune system is unleashed to fight cancer more effectively. However, PD-L1's role extends far beyond immune evasion. When situated on cancer cells, PD-L1 transmits inhibitory signals through PD-1, silencing the effector functions of T cells. However, PD-L1 also engages in reverse signaling, also called intrinsic signaling, delivering intracellular instructions that contribute to cancer cell survival, even in the absence of PD-1 binding. This signaling cascade shields cancer cells from apoptosis, drives proliferation, regulates DNA damage responses, and even functions as a co-transcriptional transactivator, amplifying cancer's ability to thrive. The intricate mechanisms behind PD-L1's intrinsic signaling are under intense investigation. In this review, we provide a historical perspective on the discoveries leading to PD-L1's structure, signaling motifs, and interacting partners, shedding light on its multifaceted roles and the promising therapeutic possibilities ahead.
Collapse
Affiliation(s)
- David Escors
- OncoImmunology Unit, Navarrabiomed-Fundacion Miguel Servet, Hospital Universitario de Navarra (HUN), Instituto de Investigación Sanitaria de Navarra (IdISNA), Universidad Publica de Navarra (UPNA), 31008 Pamplona, Spain (M.E.); (C.R.-N.); (B.V.)
| | | | | | | | | | - Grazyna Kochan
- OncoImmunology Unit, Navarrabiomed-Fundacion Miguel Servet, Hospital Universitario de Navarra (HUN), Instituto de Investigación Sanitaria de Navarra (IdISNA), Universidad Publica de Navarra (UPNA), 31008 Pamplona, Spain (M.E.); (C.R.-N.); (B.V.)
| |
Collapse
|
2
|
Feghaly C, Challita R, Hadir HB, Mobayed T, Bitar TA, Harbi M, Ghorayeb H, El-Hassan R, Bodgi L. Bladder Cancer Treatments in the Age of Personalized Medicine: A Comprehensive Review of Potential Radiosensitivity Biomarkers. Biomark Insights 2024; 19:11772719241297168. [PMID: 39512649 PMCID: PMC11542137 DOI: 10.1177/11772719241297168] [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: 06/26/2024] [Accepted: 10/17/2024] [Indexed: 11/15/2024] Open
Abstract
Bladder cancer is one of the most frequently diagnosed cancers in men. While cystectomy remains the primary treatment, advances in radiotherapy and chemotherapy have highlighted the value of bladder-preserving strategies, which can also enhance patients' quality of life. Despise these advances, around 20% of patients may still require salvage cystectomy due to tumor radioresistance. This underscores the need to develop radiosensitivity predictive assays. Radiotherapy acts by inducing DNA damage, primarily through DNA double-strand breaks, which can significantly affect treatment outcomes if left unrepaired. In addition to activating DNA repair pathways, the response to radiation also involves the tumor microenvironment, cell death pathways, immune responses and different types of cell death and proliferation receptors. In recent years, personalized medicine, which tailors treatments to individual patients, has gained increasing attention in cancer care. The development of chemo- and radiosensitivity predictive assays has become a key focus of cancer research. Despite the potential impact of such assays on bladder cancer treatment, there is still no reliable test that can help clinicians and informs patients in choosing the best treatment. This review aims to highlight studies that attempted to characterize bladder cancer radiosensitivity and to discuss the potential biomarkers that could be used to develop bladder cancer radiosensitivity predictive assays.
Collapse
Affiliation(s)
- Charbel Feghaly
- Department of Radiation Oncology, American University of Beirut Medical Center, Beirut, Lebanon
| | - Rafka Challita
- Department of Anatomy, Cell Biology and Physiological Sciences, American University of Beirut, Beirut, Lebanon
| | - Hanine Bou Hadir
- Department of Radiation Oncology, American University of Beirut Medical Center, Beirut, Lebanon
| | - Tala Mobayed
- Department of Radiation Oncology, American University of Beirut Medical Center, Beirut, Lebanon
| | - Tarek Al Bitar
- Department of Radiation Oncology, American University of Beirut Medical Center, Beirut, Lebanon
| | - Mohammad Harbi
- Department of Anatomy, Cell Biology and Physiological Sciences, American University of Beirut, Beirut, Lebanon
| | - Hala Ghorayeb
- Department of Anatomy, Cell Biology and Physiological Sciences, American University of Beirut, Beirut, Lebanon
| | - Rana El-Hassan
- Department of Anatomy, Cell Biology and Physiological Sciences, American University of Beirut, Beirut, Lebanon
| | - Larry Bodgi
- Department of Radiation Oncology, American University of Beirut Medical Center, Beirut, Lebanon
- Department of Anatomy, Cell Biology and Physiological Sciences, American University of Beirut, Beirut, Lebanon
- U1296 Unit, “Radiation: Defense, Health and Environment”, Centre Léon-Bérard, Inserm, Lyon, France
| |
Collapse
|
3
|
Park ES, Hwang YS, Ryu HW, Yoon HR, Kim JT, Lim JS, Cho HJ, Lee HG. Paulownin elicits anti-tumor effects by enhancing NK cell cytotoxicity through JNK pathway activation. Front Pharmacol 2024; 15:1439079. [PMID: 39295927 PMCID: PMC11408334 DOI: 10.3389/fphar.2024.1439079] [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: 05/27/2024] [Accepted: 08/22/2024] [Indexed: 09/21/2024] Open
Abstract
Paulownin, a natural compound derived from Paulownia tomentosa wood, exhibits various physiological functions, including anti-bacterial and anti-fungal effects. However, the impact of paulownin on natural killer (NK) cell immune activity remains largely unknown. In this study, we investigated the effect of paulownin on NK cell activity both in vitro and in vivo, and explored its potential mechanisms. NK-92 cells were used for in vitro experiments and a BALB/c mouse model with B16F10 cells injected subcutaneously were used for in vivo anti-tumor analysis. We found that paulownin enhanced the cytolytic activity of NK-92 cells against leukemia, human colon, and human lung cancer cell lines. Paulownin treatment increased the expression of the degranulation marker protein CD107a and cytolytic granules, including granzyme B and perforin in NK-92 cells. Moreover, these enhancements of cytotoxicity and the expression of cytolytic granules induced by paulownin were also observed in human primary NK cells. Signaling studies showed that paulownin promoted the phosphorylation of JNK. The increased perforin expression and elevated cytotoxic activity induced by paulownin were effectively inhibited by pre-treatment with a JNK inhibitor. In vivo studies demonstrated that the administration of paulownin suppressed the growth of B16F10 melanoma cells allografted into mice. Paulownin administration promoted the activation of NK cells in the spleen of mice, resulting in enhanced cytotoxicity against YAC-1 cells. Moreover, the anti-tumor effects of paulownin were reduced upon the depletion of NK cells. Therefore, these results suggest that paulownin enhances NK cell cytotoxicity by activating the JNK signaling pathway and provide significant implications for developing new strategies for cancer immunotherapy.
Collapse
Affiliation(s)
- Eun Sun Park
- Immunotherapy Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea
- Department of Biomolecular Science, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon, Republic of Korea
| | - Yo Sep Hwang
- Immunotherapy Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea
| | - Hyung Won Ryu
- Natural Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheong-ju, Republic of Korea
| | - Hyang Ran Yoon
- Immunotherapy Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea
| | - Jong-Tae Kim
- Immunotherapy Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea
| | - Jong-Seok Lim
- Department of Biological Science and the Cellular Heterogeneity Research Center, Research Institute of Women's Health, Sookmyung Women's University, Seoul, Republic of Korea
| | - Hee Jun Cho
- Immunotherapy Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea
- Department of Biomolecular Science, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon, Republic of Korea
| | - Hee Gu Lee
- Immunotherapy Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea
- Department of Biomolecular Science, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon, Republic of Korea
| |
Collapse
|
4
|
Kim YS, Jeong YS, Bae GH, Kang JH, Lee M, Zabel BA, Bae YS. CD200R high neutrophils with dysfunctional autophagy establish systemic immunosuppression by increasing regulatory T cells. Cell Mol Immunol 2024; 21:349-361. [PMID: 38311677 PMCID: PMC10978921 DOI: 10.1038/s41423-024-01136-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 12/21/2023] [Accepted: 01/13/2024] [Indexed: 02/06/2024] Open
Abstract
Distinct neutrophil populations arise during certain pathological conditions. The generation of dysfunctional neutrophils during sepsis and their contribution to septicemia-related systemic immune suppression remain unclear. In this study, using an experimental sepsis model that features immunosuppression, we identified a novel population of pathogenic CD200Rhigh neutrophils that are generated during the initial stages of sepsis and contribute to systemic immune suppression by enhancing regulatory T (Treg) cells. Compared to their CD200Rlow counterparts, sepsis-generated CD200Rhigh neutrophils exhibit impaired autophagy and dysfunction, with reduced chemotactic migration, superoxide anion production, and TNF-α production. Increased soluble CD200 blocks autophagy and neutrophil maturation in the bone marrow during experimental sepsis, and recombinant CD200 treatment in vitro can induce neutrophil dysfunction similar to that observed in CD200Rhigh neutrophils. The administration of an α-CD200R antibody effectively reversed neutrophil dysfunction by enhancing autophagy and protecting against a secondary infection challenge, leading to increased survival. Transcriptome analysis revealed that CD200Rhigh neutrophils expressed high levels of Igf1, which elicits the generation of Treg cells, while the administration of an α-CD200R antibody inhibited Treg cell generation in a secondary infection model. Taken together, our findings revealed a novel CD200Rhigh neutrophil population that mediates the pathogenesis of sepsis-induced systemic immunosuppression by generating Treg cells.
Collapse
Affiliation(s)
- Ye Seon Kim
- Department of Biological Sciences, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Yu Sun Jeong
- Department of Biological Sciences, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Geon Ho Bae
- Department of Biological Sciences, Sungkyunkwan University, Suwon, 16419, Republic of Korea
- Division of Immunology, Boston Children's Hospital, Boston, MA, USA
| | - Ji Hyeon Kang
- Department of Biological Sciences, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Mingyu Lee
- Department of Health Science and Technology, SAIHST, Sungkyunkwan University, Seoul, 06351, Republic of Korea
| | - Brian A Zabel
- Palo Alto Veterans Institute for Research, Veterans Affairs Hospital, Palo Alto, CA, 94304, USA
| | - Yoe-Sik Bae
- Department of Biological Sciences, Sungkyunkwan University, Suwon, 16419, Republic of Korea.
- Department of Health Science and Technology, SAIHST, Sungkyunkwan University, Seoul, 06351, Republic of Korea.
| |
Collapse
|
5
|
Gurung P, Lim J, Shrestha R, Kim YW. Chlorin e6-associated photodynamic therapy enhances abscopal antitumor effects via inhibition of PD-1/PD-L1 immune checkpoint. Sci Rep 2023; 13:4647. [PMID: 36944686 PMCID: PMC10030802 DOI: 10.1038/s41598-023-30256-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 02/20/2023] [Indexed: 03/23/2023] Open
Abstract
We hypothesized that photodynamic therapy (PDT) with Chlorin e6 (Ce6) enhances antitumor abscopal effects via inhibition of the programmed cell death-1/programmed death-ligand 1 (PD-1/PD-L1) immune checkpoint. By using syngeneic melanoma and pancreatic tumor mouse models, we studied the Ce6-PDT-induced immune responses in local and distant tumor microenvironments. In addition, the Ce6-PDT's target in the PD-1/PD-L1 interaction was analyzed in MC38-hPD-L1 colon cancer and PD-1 expressing Jurkat T cell coculture. The tumors in the irradiated and non-irradiated sites in the abscopal effective (Abseff) group of both mouse models were regressed, proving the abscopal effect. The immunogenic effect in the Abseff group was associated with an expansion of T cell and other immune cells infiltration without changes in the CD39+ population in either the right or left tumors compared to control group. Furthermore, the abscopal ineffective (Absineff) group demonstrated lesser increase of T cells, decreased immune cell infiltration, and increased CD39-expressing Treg cells without suppression of tumor growth. In the coculture with PD-1-expressing Jurkat T cell, Ce6-PDT efficiently suppressed the PD-1/PD-L1 interactions by increasing the proliferation and cytotoxic activity of CD8+ T cells while decreasing CD39-expressing Treg cells in a dose-dependent manner. Likewise, the inhibition of PD-1/PD-L1 interactions was also correlated with the increased production of IL-2 and Granzyme B. Our findings imply that Ce6-PDT is a promising immunotherapy with the potential to improve the abscopal effect.
Collapse
Grants
- (NTIS Number: 1711174319, RS-2020-KD000106), (S3034405) The Korea Medical Device Development Fund grant funded by the Korea government (the Ministry of Science and ICT, the Ministry of Trade, Industry and Energy, the Ministry of Health & Welfare, the Ministry of Food and Drug Safety) (NTIS Number: 1711174319, RS-2020-KD000106), and the Technology development Program (S3034405) funded by the Ministry of SMEs and Startups(MSS, Korea).
- (NTIS Number: 1711174319, RS-2020-KD000106), (S3034405) The Korea Medical Device Development Fund grant funded by the Korea government (the Ministry of Science and ICT, the Ministry of Trade, Industry and Energy, the Ministry of Health & Welfare, the Ministry of Food and Drug Safety) (NTIS Number: 1711174319, RS-2020-KD000106), and the Technology development Program (S3034405) funded by the Ministry of SMEs and Startups(MSS, Korea).
- (NTIS Number: 1711174319, RS-2020-KD000106), (S3034405) The Korea Medical Device Development Fund grant funded by the Korea government (the Ministry of Science and ICT, the Ministry of Trade, Industry and Energy, the Ministry of Health & Welfare, the Ministry of Food and Drug Safety) (NTIS Number: 1711174319, RS-2020-KD000106), and the Technology development Program (S3034405) funded by the Ministry of SMEs and Startups(MSS, Korea).
- (NTIS Number: 1711174319, RS-2020-KD000106), (S3034405) The Korea Medical Device Development Fund grant funded by the Korea government (the Ministry of Science and ICT, the Ministry of Trade, Industry and Energy, the Ministry of Health & Welfare, the Ministry of Food and Drug Safety) (NTIS Number: 1711174319, RS-2020-KD000106), and the Technology development Program (S3034405) funded by the Ministry of SMEs and Startups(MSS, Korea).
Collapse
Affiliation(s)
- Pallavi Gurung
- Dongsung Cancer Center, Dongsung Biopharmaceutical, Daegu, 41061, South Korea
| | - Junmo Lim
- Dongsung Cancer Center, Dongsung Biopharmaceutical, Daegu, 41061, South Korea
| | - Rajeev Shrestha
- Dongsung Cancer Center, Dongsung Biopharmaceutical, Daegu, 41061, South Korea
| | - Yong-Wan Kim
- Dongsung Cancer Center, Dongsung Biopharmaceutical, Daegu, 41061, South Korea.
| |
Collapse
|
6
|
Aru B, Pehlivanoğlu C, Dal Z, Dereli-Çalışkan NN, Gürlü E, Yanıkkaya-Demirel G. A potential area of use for immune checkpoint inhibitors: Targeting bone marrow microenvironment in acute myeloid leukemia. Front Immunol 2023; 14:1108200. [PMID: 36742324 PMCID: PMC9895857 DOI: 10.3389/fimmu.2023.1108200] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Accepted: 01/03/2023] [Indexed: 01/22/2023] Open
Abstract
Acute myeloid leukemia (AML) arises from the cells of myeloid lineage and is the most frequent leukemia type in adulthood accounting for about 80% of all cases. The most common treatment strategy for the treatment of AML includes chemotherapy, in rare cases radiotherapy and stem cell and bone marrow transplantation are considered. Immune checkpoint proteins involve in the negative regulation of immune cells, leading to an escape from immune surveillance, in turn, causing failure of tumor cell elimination. Immune checkpoint inhibitors (ICIs) target the negative regulation of the immune cells and support the immune system in terms of anti-tumor immunity. Bone marrow microenvironment (BMM) bears various blood cell lineages and the interactions between these lineages and the noncellular components of BMM are considered important for AML development and progression. Administration of ICIs for the AML treatment may be a promising option by regulating BMM. In this review, we summarize the current treatment options in AML treatment and discuss the possible application of ICIs in AML treatment from the perspective of the regulation of BMM.
Collapse
Affiliation(s)
- Başak Aru
- Immunology Department, Faculty of Medicine, Yeditepe University, Istanbul, Türkiye
| | - Cemil Pehlivanoğlu
- Immunology Department, Faculty of Medicine, Yeditepe University, Istanbul, Türkiye
| | - Zeynep Dal
- School of Medicine, Yeditepe University, Istanbul, Türkiye
| | | | - Ege Gürlü
- School of Medicine, Yeditepe University, Istanbul, Türkiye
| | - Gülderen Yanıkkaya-Demirel
- Immunology Department, Faculty of Medicine, Yeditepe University, Istanbul, Türkiye,*Correspondence: Gülderen Yanıkkaya-Demirel,
| |
Collapse
|
7
|
Bleve S, Cursano MC, Casadei C, Schepisi G, Menna C, Urbini M, Gianni C, De Padova S, Filograna A, Gallà V, Rosti G, Barone D, Chovanec M, Mego M, De Giorgi U. Inflammatory Biomarkers for Outcome Prediction in Patients With Metastatic Testicular Cancer. Front Oncol 2022; 12:910087. [PMID: 35756636 PMCID: PMC9226315 DOI: 10.3389/fonc.2022.910087] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 04/26/2022] [Indexed: 11/13/2022] Open
Abstract
Germ cell tumors are the most common malignant tumors in male young adults. Platinum-based chemotherapy has dramatically improved the outcome of metastatic germ cell tumor patients and overall cure rates now exceed 80%. The choice of medical treatment can be guided by the prognosis estimation which is an important step during the decision-making process. IGCCCG classification plays a pivotal role in the management of advanced disease. However, histological and clinical parameters are the available factors that condition the prognosis, but they do not reflect the tumor's molecular and pathological features and do not predict who will respond to chemotherapy. After first-line chemotherapy 20%-30% of patients relapse and for these patients, the issue of prognostic factors is far more complex. Validated biomarkers and a molecular selection of patients that reflect the pathogenesis are highly needed. The association between cancer-related systemic inflammation, tumorigenesis, and cancer progression has been demonstrated. In the last years, several studies have shown the prognostic utility of immune-inflammation indexes in different tumor types. This review analyzed the prognostic impact of inflammatory markers retrieved from routine blood draws in GCT patients.
Collapse
Affiliation(s)
- Sara Bleve
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Maria Concetta Cursano
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Chiara Casadei
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Giuseppe Schepisi
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Cecilia Menna
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Milena Urbini
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Caterina Gianni
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Silvia De Padova
- Psycho-Oncology Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Alessia Filograna
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Valentina Gallà
- Unit of Biostatistics and Clinical Trials, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Giovanni Rosti
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Domenico Barone
- Radiology Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Michal Chovanec
- 2nd Department of Oncology, Comenius University, Faculty of Medicine, National Cancer Institute, Bratislava, Slovakia
| | - Michal Mego
- 2nd Department of Oncology, Comenius University, Faculty of Medicine, National Cancer Institute, Bratislava, Slovakia
| | - Ugo De Giorgi
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| |
Collapse
|
8
|
Wang Y, Shang K, Zhang N, Zhao J, Cao B. Tumor-Associated Macrophage-Derived Exosomes Promote the Progression of Gastric Cancer by Regulating the P38MAPK Signaling Pathway and the Immune Checkpoint PD-L1. Cancer Biother Radiopharm 2021. [PMID: 34698510 DOI: 10.1089/cbr.2021.0218] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Objective: To investigate the effects of M2 macrophage-derived exosomes (M2-Exos) on proliferation, migration, and apoptosis of gastric cancer cells in the tumor microenvironment and to further explore their possible molecular mechanism. Materials and Methods: M2 macrophages were induced from THP-1 cells and identified by qRT-PCR. Exosomes were extracted by ultracentrifugation and identified by transmission electron microscopy (TEM), nanoparticle tracking analysis (NTA), and Western blot analysis. Fluorescence labeling was used to detect the internalization of exosomes in receptors. The proliferation, migration, and invasion of AGS and HGC27 cells were determined by EdU and MTS, wound healing and Transwell assay, and flow cytometry, respectively. Proteins in the related pathway of M2-Exos affecting the progression of gastric cancer were detected by Western blot analysis. Results: In this study, M2 macrophages and M2-Exos were successfully obtained. The purified M2-Exos were observed as small round vesicles with diameters of 50-90 nm and positive expression of CD63, CD9, and TSG101. Besides, M2-Exos can be effectively taken up and internalized by AGS and HGC27 cells. Cell behavior studies showed that M2-Exos promoted proliferation and migration and inhibited the apoptosis of AGS and HGC27. Further research illustrated that M2-Exos promoted the phosphorylation of P38 and high expression of programmed death ligand 1 (PD-L1). Conclusions: This study demonstrated that M2-Exos promoted proliferation and migration and inhibited the apoptosis of gastric cancer cells. Mechanically, M2-Exos may promote gastric cancer progression through the P38MAPK signaling pathway and achieve immune escape through elevating the expression of PD-L1.
Collapse
Affiliation(s)
- Yusheng Wang
- Cancer Center, Capital Medical University-Affiliated Beijing Friendship Hospital, Beijing, People's Republic of China
| | - Kun Shang
- Cancer Center, Capital Medical University-Affiliated Beijing Friendship Hospital, Beijing, People's Republic of China
| | - Ninggang Zhang
- Department of Gastrointestinal Oncology, Shanxi Provincial Cancer Hospital, Affiliated to Shanxi Medical University, Taiyuan, People's Republic of China
| | - Jian Zhao
- Department of Gastrointestinal Oncology, Shanxi Provincial Cancer Hospital, Affiliated to Shanxi Medical University, Taiyuan, People's Republic of China
| | - Bangwei Cao
- Cancer Center, Capital Medical University-Affiliated Beijing Friendship Hospital, Beijing, People's Republic of China
| |
Collapse
|
9
|
Liu YS, Huang BR, Lin CJ, Shen CK, Lai SW, Chen CW, Lin HJ, Lin CH, Hsieh YC, Lu DY. Paliperidone Inhibits Glioblastoma Growth in Mouse Brain Tumor Model and Reduces PD-L1 Expression. Cancers (Basel) 2021; 13:cancers13174357. [PMID: 34503167 PMCID: PMC8430966 DOI: 10.3390/cancers13174357] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 08/24/2021] [Indexed: 12/24/2022] Open
Abstract
Simple Summary The present study showed that a prescribed psychotropic medicine paliperidone inhibits GBM growth and prolongs survival in mouse brain tumor model and decreased the programmed death ligand 1 expression. Using the 3D co-culture also found that dopamine receptor D2 regulates the interaction of GBM-macrophage-induced PD-L1 expression in GBMs. In addition, the expression of DRD2 and PD-L1 in GBM modulates tumor-associated macrophage polarization. Our results also indicated that there is a contact-independent mechanism of PD-L1 induction in GBM upon interaction between GBM and monocytes. The present study also found that the interaction of GBM-macrophage-enhanced PD-L1 expression in GBM occurred by modulating the ERK and STAT3 signaling pathways. In addition, the inhibition of DRD2 reduces the upregulation of PD-1 expression, and it is regulating signaling in GBM. Abstract A previous study from our group reported that monocyte adhesion to glioblastoma (GBM) promoted tumor growth and invasion activity and increased tumor-associated macrophages (TAMs) proliferation and inflammatory mediator secretion as well. The present study showed that prescribed psychotropic medicine paliperidone reduced GBM growth and immune checkpoint protein programmed death ligand (PD-L)1 expression and increased survival in an intracranial xenograft mouse model. An analysis of the database of patients with glioma showed that the levels of PD-L1 and dopamine receptor D (DRD)2 were higher in the GBM group than in the low grade astrocytoma and non-tumor groups. In addition, GFP expressing GBM (GBM-GFP) cells co-cultured with monocytes-differentiated macrophage enhanced PD-L1 expression in GBM cells. The enhancement of PD-L1 in GBM was antagonized by paliperidone and risperidone as well as DRD2 selective inhibitor L741426. The expression of CD206 (M2 phenotype marker) was observed to be markedly increased in bone marrow-derived macrophages (BMDMs) co-cultured with GBM. Importantly, treatment with paliperidone effectively decreased CD206 and also dramatically increased CD80 (M1 phenotype marker) in BMDMs. We have previously established a PD-L1 GBM-GFP cell line that stably expresses PD-L1. Experiments showed that the expressions of CD206 was increased and CD80 was mildly decreased in the BMDMs co-cultured with PD-L1 GBM-GFP cells. On the other hands, knockdown of DRD2 expression in GBM cells dramatically decreased the expression of CD206 but markedly increased CD80 expressions in BMDMs. The present study suggests that DRD2 may be involved in regulating the PD-L1 expression in GBM and the microenvironment of GBM. Our results provide a valuable therapeutic strategy and indicate that treatments combining DRD2 antagonist paliperidone with standard immunotherapy may be beneficial for GBM treatment.
Collapse
Affiliation(s)
- Yu-Shu Liu
- Department of Pharmacology, School of Medicine, China Medical University, Taichung 404, Taiwan; (Y.-S.L.); (S.-W.L.); (H.-J.L.)
| | - Bor-Ren Huang
- Department of Neurosurgery, Taichung Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Taichung 404, Taiwan;
- School of Medicine, Tzu Chi University, Taichung 404, Taiwan
| | - Ching-Ju Lin
- Department of Physiology, School of Medicine, China Medical University, Taichung 404, Taiwan;
| | - Ching-Kai Shen
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 404, Taiwan;
| | - Sheng-Wei Lai
- Department of Pharmacology, School of Medicine, China Medical University, Taichung 404, Taiwan; (Y.-S.L.); (S.-W.L.); (H.-J.L.)
| | - Chao-Wei Chen
- Institute of New Drug Development, China Medical University, Taichung 404, Taiwan;
| | - Hui-Jung Lin
- Department of Pharmacology, School of Medicine, China Medical University, Taichung 404, Taiwan; (Y.-S.L.); (S.-W.L.); (H.-J.L.)
| | - Chia-Huei Lin
- Department of Pharmacy, China Medical University, Taichung 404, Taiwan; (C.-H.L.); (Y.-C.H.)
| | - Yun-Chen Hsieh
- Department of Pharmacy, China Medical University, Taichung 404, Taiwan; (C.-H.L.); (Y.-C.H.)
| | - Dah-Yuu Lu
- Department of Pharmacology, School of Medicine, China Medical University, Taichung 404, Taiwan; (Y.-S.L.); (S.-W.L.); (H.-J.L.)
- Department of Photonics and Communication Engineering, Asia University, Taichung 404, Taiwan
- Correspondence: ; Tel.: +886-422-053-366 (ext. 2253)
| |
Collapse
|
10
|
Farias A, Soto A, Puttur F, Goldin CJ, Sosa S, Gil C, Goldbaum FA, Berguer PM. A TLR4 agonist improves immune checkpoint blockade treatment by increasing the ratio of effector to regulatory cells within the tumor microenvironment. Sci Rep 2021; 11:15406. [PMID: 34321536 PMCID: PMC8319313 DOI: 10.1038/s41598-021-94837-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Accepted: 07/15/2021] [Indexed: 12/18/2022] Open
Abstract
Brucella lumazine synthase (BLS) is a homodecameric protein that activates dendritic cells via toll like receptor 4, inducing the secretion of pro-inflammatory cytokines and chemokines. We have previously shown that BLS has a therapeutic effect in B16 melanoma-bearing mice only when administered at early stages of tumor growth. In this work, we study the mechanisms underlying the therapeutic effect of BLS, by analyzing the tumor microenvironment. Administration of BLS at early stages of tumor growth induces high levels of serum IFN-γ, as well as an increment of hematopoietic immune cells within the tumor. Moreover, BLS-treatment increases the ratio of effector to regulatory cells. However, all treated mice eventually succumb to the tumors. Therefore, we combined BLS administration with anti-PD-1 treatment. Combined treatment increases the outcome of both monotherapies. In conclusion, we show that the absence of the therapeutic effect at late stages of tumor growth correlates with low levels of serum IFN-γ and lower infiltration of immune cells in the tumor, both of which are essential to delay tumor growth. Furthermore, the combined treatment of BLS and PD-1 blockade shows that BLS could be exploited as an essential immunomodulator in combination therapy with an immune checkpoint blockade to treat skin cancer.
Collapse
Affiliation(s)
- A Farias
- Fundación Instituto Leloir, IIBBA, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - A Soto
- Fundación Instituto Leloir, IIBBA, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - F Puttur
- Inflammation, Repair and Development, National Heart and Lung Institute, Imperial College London, London, UK
| | - C J Goldin
- Fundación Instituto Leloir, IIBBA, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - S Sosa
- Fundación Instituto Leloir, IIBBA, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - C Gil
- Fundación Instituto Leloir, IIBBA, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - F A Goldbaum
- Fundación Instituto Leloir, IIBBA, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - P M Berguer
- Fundación Instituto Leloir, IIBBA, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina.
| |
Collapse
|
11
|
Fejza A, Polano M, Camicia L, Poletto E, Carobolante G, Toffoli G, Mongiat M, Andreuzzi E. The Efficacy of Anti-PD-L1 Treatment in Melanoma Is Associated with the Expression of the ECM Molecule EMILIN2. Int J Mol Sci 2021; 22:ijms22147511. [PMID: 34299131 PMCID: PMC8306837 DOI: 10.3390/ijms22147511] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 07/07/2021] [Accepted: 07/08/2021] [Indexed: 12/14/2022] Open
Abstract
The use of immune checkpoint inhibitors has revolutionized the treatment of melanoma patients, leading to remarkable improvements in the cure. However, to ensure a safe and effective treatment, there is the need to develop markers to identify the patients that would most likely respond to the therapies. The microenvironment is gaining attention in this context, since it can regulate both the immunotherapy efficacyand angiogenesis, which is known to be affected by treatment. Here, we investigated the putative role of the ECM molecule EMILIN-2, a tumor suppressive and pro-angiogenic molecule. We verified that the EMILIN2 expression is variable among melanoma patients and is associated with the response to PD-L1 inhibitors. Consistently, in preclinical settings, the absence of EMILIN-2 is associated with higher PD-L1 expression and increased immunotherapy efficacy. We verified that EMILIN-2 modulates PD-L1 expression in melanoma cells through indirect immune-dependent mechanisms. Notably, upon PD-L1 blockage, Emilin2−/− mice displayed improved intra-tumoral vessel normalization and decreased tumor hypoxia. Finally, we provide evidence indicating that the inclusion of EMILIN2 in a number of gene expression signatures improves their predictive potential, a further indication that the analysis of this molecule may be key for the development of new markers to predict immunotherapy efficacy.
Collapse
Affiliation(s)
- Albina Fejza
- Division of Molecular Oncology, Department of Research and Diagnosis, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano, Italy; (A.F.); (L.C.); (E.P.); (G.C.)
| | - Maurizio Polano
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano, Italy; (M.P.); (G.T.)
| | - Lucrezia Camicia
- Division of Molecular Oncology, Department of Research and Diagnosis, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano, Italy; (A.F.); (L.C.); (E.P.); (G.C.)
| | - Evelina Poletto
- Division of Molecular Oncology, Department of Research and Diagnosis, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano, Italy; (A.F.); (L.C.); (E.P.); (G.C.)
| | - Greta Carobolante
- Division of Molecular Oncology, Department of Research and Diagnosis, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano, Italy; (A.F.); (L.C.); (E.P.); (G.C.)
| | - Giuseppe Toffoli
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano, Italy; (M.P.); (G.T.)
| | - Maurizio Mongiat
- Division of Molecular Oncology, Department of Research and Diagnosis, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano, Italy; (A.F.); (L.C.); (E.P.); (G.C.)
- Correspondence: (M.M.); (E.A.)
| | - Eva Andreuzzi
- Division of Molecular Oncology, Department of Research and Diagnosis, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano, Italy; (A.F.); (L.C.); (E.P.); (G.C.)
- Correspondence: (M.M.); (E.A.)
| |
Collapse
|
12
|
Lu Z, Long Y, Wang Y, Wang X, Xia C, Li M, Zhang Z, He Q. Phenylboronic acid modified nanoparticles simultaneously target pancreatic cancer and its metastasis and alleviate immunosuppression. Eur J Pharm Biopharm 2021; 165:164-173. [PMID: 34020022 DOI: 10.1016/j.ejpb.2021.05.014] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 05/05/2021] [Accepted: 05/12/2021] [Indexed: 12/24/2022]
Abstract
Pancreatic ductal adenocarcinoma is one of the most lethal malignant tumors, its drug resistance, immunosuppression and metastasis makes the traditional chemotherapy and immunotherapy inefficient. Here we confirmed a 3-aminophenylboronic acid-modified low molecular weight heparin-D-α-tocopheryl succinate micellar nanoparticle (PBA-LMWH-TOS NP, PLT NP) could inhibit orthotopic pancreatic tumor and its spontaneous metastases. The small particle size and high affinity of PBA to sialic acid residue (SA) made PLT/PTX NPs significantly targeted and accumulated in both pancreatic tumor tissues and metastases. The immunosuppressive microenvironment of pancreatic tumor was most caused by the infiltration of immunosuppressive cells, mainly myeloid-derived suppressor cells (MDSCs). We first reported that P-selectin glycoprotein ligand-1 (PSGL-1) was expressed on the surfaces of MDSCs in pancreatic tumor tissues. Meanwhile, we found that LMWH could inhibit the early stage of adhesion cascade between vascular endothelial cells (VECs) and MDSCs by interfering with P-selectin/PSGL-1 binding, thus inhibiting MDSC recruitment to pancreatic tumor tissues. The therapeutic results indicated that PLT/PTX NPs could significantly improve the immune microenvironment of pancreatic tumor and inhibit spontaneous metastases. This nanosystem provides a new immune microenvironment regulation mechanism based on carrier materials in pancreatic tumor, and has high clinical application potential.
Collapse
Affiliation(s)
- Zhengze Lu
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, People's Republic of China
| | - Yang Long
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, People's Republic of China
| | - Yashi Wang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, People's Republic of China
| | - Xuhui Wang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, People's Republic of China
| | - Chunyu Xia
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, People's Republic of China
| | - Man Li
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, People's Republic of China
| | - Zhirong Zhang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, People's Republic of China
| | - Qin He
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, People's Republic of China.
| |
Collapse
|
13
|
Trailblazing perspectives on targeting breast cancer stem cells. Pharmacol Ther 2021; 223:107800. [PMID: 33421449 DOI: 10.1016/j.pharmthera.2021.107800] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 12/30/2020] [Accepted: 01/04/2021] [Indexed: 12/12/2022]
Abstract
Breast cancer (BCa) is one of the most prevalent malignant tumors affecting women's health worldwide. The recurrence and metastasis of BCa have made it a long-standing challenge to achieve remission-persistent or disease-undetectable clinical outcomes. Cancer stem cells (CSCs) possess the ability to self-renew and generate heterogeneous tumor bulk. The existence of CSCs has been found to be vital in the initiation, metastasis, therapy resistance, and recurrence of tumors across cancer types. Because CSCs grow slowly in their dormant state, they are insensitive to conventional chemotherapies; however, when CSCs emerge from their dormant state and become clinically evident, they usually acquire genetic traits that make them resistant to existing therapies. Moreover, CSCs also show evidence of acquired drug resistance in synchrony with tumor relapses. The concept of CSCs provides a new treatment strategy for BCa. In this review, we highlight the recent advances in research on breast CSCs and their association with epithelial-mesenchymal transition (EMT), circulating tumor cells (CTCs), plasticity of tumor cells, tumor microenvironment (TME), T-cell modulatory protein PD-L1, and non-coding RNAs. On the basis that CSCs are associated with multiple dysregulated biological processes, we envisage that increased understanding of disease sub-classification, selected combination of conventional treatment, molecular aberration directed therapy, immunotherapy, and CSC targeting/sensitizing strategy might improve the treatment outcome of patients with advanced BCa. We also discuss novel perspectives on new drugs and therapeutics purposing the potent and selective expunging of CSCs.
Collapse
|
14
|
Liu BL, Cao QL, Zhao X, Liu HZ, Zhang YQ. Inhibition of TRPV1 by SHP-1 in nociceptive primary sensory neurons is critical in PD-L1 analgesia. JCI Insight 2020; 5:137386. [PMID: 32960817 PMCID: PMC7605531 DOI: 10.1172/jci.insight.137386] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Accepted: 09/10/2020] [Indexed: 12/15/2022] Open
Abstract
Recently programmed death-ligand 1 (PD-L1) receptor PD-1 was found in dorsal root ganglion (DRG) neurons, and PD-L1 activates PD-1 to inhibit inflammatory and neuropathic pain by modulating neuronal excitability. However, the downstream signaling of PD-1 in sensory neurons remains unclear. Here, we show that PD-L1 activated Src homology 2 domain-containing tyrosine phosphatase-1 (SHP-1) to downregulate transient receptor potential vanilloid 1 (TRPV1) in DRG neurons and inhibit bone cancer pain in mice. Local injection of PD-L1 produced analgesia. PD-1 in DRG neurons colocalized with TRPV1 and SHP-1. PD-L1 induced the phosphorylation of SHP-1 in DRG TRPV1 neurons and inhibited TRPV1 currents. Loss of TRPV1 in mice abolished bone cancer–induced thermal hyperalgesia and PD-L1 analgesia. Conditioned deletion of SHP-1 in NaV1.8+ neurons aggravated bone cancer pain and diminished the inhibition of PD-L1 on TRPV1 currents and pain. Together, our findings suggest that PD-L1/PD-1 signaling suppresses bone cancer pain via inhibition of TRPV1 activity. Our results also suggest that SHP-1 in sensory neurons is an endogenous pain inhibitor and delays the development of bone cancer pain via suppressing TRPV1 function. PD-L1/PD-1 signaling suppresses TRPV1 activity and alleviates pain-like behaviors via phosphorylation of SHP-1 in nociceptive primary sensory neurons in a mouse bone cancer model.
Collapse
Affiliation(s)
- Ben-Long Liu
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Qi-Lai Cao
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Xin Zhao
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Hui-Zhu Liu
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Yu-Qiu Zhang
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, Institutes of Brain Science, Fudan University, Shanghai, China.,Institutes of Integrative Medicine, Fudan University, Shanghai, China
| |
Collapse
|
15
|
Kalavska K, Schmidtova S, Chovanec M, Mego M. Immunotherapy in Testicular Germ Cell Tumors. Front Oncol 2020; 10:573977. [PMID: 33072608 PMCID: PMC7542989 DOI: 10.3389/fonc.2020.573977] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 08/24/2020] [Indexed: 12/21/2022] Open
Abstract
Testicular germ cell tumors (TGCTs) are malignancies with very high curative potential even in metastatic settings, mainly due to the introduction of cisplatin in the treatment of this disease. However, in a group of patients with cisplatin-refractory disease or with progressive disease despite high-dose salvage chemotherapy treatment, the prognosis is typically dismal. The triple combination of gemcitabine, oxaliplatin, and paclitaxel (GOP) has reasonable efficacy and is considered to be standard care for this group of patients. It remains to be seen, however, whether refractory TGCTs may represent a potential target for immune checkpoint inhibition. This review will focus on the rationale of the use of immunotherapy for platinum-refractory TGCTs and summarize data reporting experiences with immune checkpoint inhibitor treatment for this malignancy.
Collapse
Affiliation(s)
- Katarina Kalavska
- Translational Research Unit, Faculty of Medicine, Comenius University and National Cancer Institute, Bratislava, Slovakia.,Department of Molecular Oncology, Cancer Research Institute, Biomedical Research Center, University Science Park for Biomedicine, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Silvia Schmidtova
- Translational Research Unit, Faculty of Medicine, Comenius University and National Cancer Institute, Bratislava, Slovakia.,Department of Molecular Oncology, Cancer Research Institute, Biomedical Research Center, University Science Park for Biomedicine, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Michal Chovanec
- Second Department of Oncology, Faculty of Medicine, Comenius University and National Cancer Institute, Bratislava, Slovakia
| | - Michal Mego
- Translational Research Unit, Faculty of Medicine, Comenius University and National Cancer Institute, Bratislava, Slovakia.,Second Department of Oncology, Faculty of Medicine, Comenius University and National Cancer Institute, Bratislava, Slovakia
| |
Collapse
|
16
|
Bridgwater C, Geller A, Hu X, Burlison JA, Zhang HG, Yan J, Guo H. 89Zr-Labeled Anti-PD-L1 Antibody Fragment for Evaluating In Vivo PD-L1 Levels in Melanoma Mouse Model. Cancer Biother Radiopharm 2020; 35:549-557. [PMID: 32315549 DOI: 10.1089/cbr.2019.3056] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The rise of programmed death-1 (PD-1)/PD-L1 immune checkpoint inhibitor therapy has been one of the most promising developments in melanoma research. However, not all the melanoma patients respond to such immune checkpoint blockade. There is a great need of biomarkers for appropriate melanoma patient selection and therapeutic efficacy monitoring. The objective of this study is to develop a novel radiolabeled anti-PD-L1 antibody fragment, as an imaging biomarker, for evaluating the in vivo PD-L1 levels in melanoma. The Df-conjugated F(ab')2 fragment of the anti-mouse PD-L1 antibody was successfully synthesized and radiolabeled with 89Zr. Both Df-F(ab')2 and 89Zr-Df-F(ab')2 maintained the nano-molar murine PD-L1 targeting specificity and affinity. 89Zr-Df-F(ab')2 showed less uptake in normal liver tissue in mice compared with its full antibody counterpart 89Zr-Df-anti-PD-L1. Positron emission tomography (PET)/computed tomography images clearly showed that 89Zr-Df-F(ab')2 possessed superior pharmacokinetics and imaging contrast over the radiolabeled full antibody, with much earlier and higher tumor uptake (5.5 times more at 2 h post injection) and much lower liver background (51% reduction at 2 h post injection). The specific and high murine PD-L1-targeting uptake at tumor foci coupled with fast clearance of 89Zr-Df-F(ab')2 highlighted its potential for in vivo PET imaging of murine PD-L1 levels and future development of radiolabeled anti-human PD-L1 fragment for potential application in melanoma patients.
Collapse
Affiliation(s)
- Caleb Bridgwater
- Department of Radiology, University of Louisville, Louisville, Kentucky, USA
| | - Anne Geller
- Department of Microbiology and Immunology, University of Louisville, Louisville, Kentucky, USA
| | - Xiaoling Hu
- James Graham Brown Cancer Center, University of Louisville, Louisville, Kentucky, USA
| | - Joe A Burlison
- James Graham Brown Cancer Center, University of Louisville, Louisville, Kentucky, USA
| | - Huang-Ge Zhang
- Department of Microbiology and Immunology, University of Louisville, Louisville, Kentucky, USA.,James Graham Brown Cancer Center, University of Louisville, Louisville, Kentucky, USA
| | - Jun Yan
- Department of Microbiology and Immunology, University of Louisville, Louisville, Kentucky, USA.,James Graham Brown Cancer Center, University of Louisville, Louisville, Kentucky, USA.,Department of Surgery, University of Louisville, Louisville, Kentucky, USA
| | - Haixun Guo
- Department of Radiology, University of Louisville, Louisville, Kentucky, USA.,Center for Predictive Medicine, University of Louisville, Louisville, Kentucky, USA
| |
Collapse
|
17
|
Kang R, Zeh H, Lotze M, Tang D. The Multifaceted Effects of Autophagy on the Tumor Microenvironment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1225:99-114. [PMID: 32030650 DOI: 10.1007/978-3-030-35727-6_7] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The tumor microenvironment is composed of cancer cells, noncancer cells (e.g., immune cells, stromal cells, endothelial cells, and adipocytes), and various mediators (e.g., cytokines, chemokines, growth factors, and humoral factors) that work together to support cancer growth, progression, and resistance to therapies. Autophagy is an evolutionarily conserved degradation mechanism by which various cytosolic cargos (e.g., damaged organelles, unused molecules, or invaded pathogens) are engulfed by double-membrane autophagosomes, and then delivered into the lysosome for degradation and recycling. The level of autophagy is a crucial threshold to either promote cell survival or induce cell death in response to environmental stresses. Autophagy plays a context-dependent role in tumorigenesis and anticancer therapy via shaping the inflammatory, hypoxic, immunosuppressive, and metabolic tumor microenvironment. In particular, impaired autophagy flux is associated with chronic inflammation, immunosuppression, stromal formation, cancer stemness, angiogenesis, metastasis, and metabolic reprogramming in the tumor microenvironment. Understanding the molecular machinery of autophagy and its communication with hallmarks of cancer could lead to potential new anticancer strategies or drugs.
Collapse
Affiliation(s)
- Rui Kang
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX, USA.
| | - Herbert Zeh
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX, USA
| | - Michael Lotze
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Daolin Tang
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
18
|
Kalavska K, Kucerova L, Schmidtova S, Chovanec M, Mego M. Cancer Stem Cell Niche and Immune-Active Tumor Microenvironment in Testicular Germ Cell Tumors. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1226:111-121. [DOI: 10.1007/978-3-030-36214-0_9] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
19
|
Yang J, Liu J, Sheng M, Zhang X, Liu M. Programmed cell death protein 1 promotes hepatitis B virus transmission through the regulation of ERK1/2-mediated trophoblasts differentiation. Arch Gynecol Obstet 2019; 301:551-558. [PMID: 31792623 DOI: 10.1007/s00404-019-05401-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Accepted: 11/22/2019] [Indexed: 10/25/2022]
Abstract
PURPOSE The purpose of our research is to evaluate the mechanism of PD-1 in the promotion of HBV transmission. METHODS HBV was used to infect two human choriocarcinoma cell line, including JEG-3, as well as BeWo. We used PCR and western blotting to detect PD-1 gene and protein expression levels in cells. Stable knockdown of the PD-1 gene in JEG-3 cells was obtained by lentiviral transfection. Trophoblast cell proliferation was evaluated using CCK8 and flow cytometry. The concentration of HBV antibody in the cell supernatant was measured by ELISA. DNA was then extracted from the cells and the copy number of the HBV virus was detected by PCR. Finally, ERK1/2 expression was detected by western blot. RESULTS High PD-1 gene expression in HBV-infected trophoblasts and the knockdown of PD-1 gene can, respectively, improve the proliferation of HBV-infected trophoblasts and reduce viral replication in trophoblasts. In addition, PD-1 and ERK1/2 proteins were co-expressed in HBV-infected trophoblasts and inhibited the activation of ERK1/2 pathway in HBV-infected trophoblasts. ERK1/2 expression significantly increased after PD-1 knockdown. Therefore, PD-1 might be an important protein in trophoblast cells infected with HBV. CONCLUSIONS PD-1 promoted HBV transmission through regulating ERK1/2-mediated trophoblasts differentiation. Therefore, our research may provide new ideas and methods for preventing mother-to-child transmission of HBV infection during pregnancy.
Collapse
Affiliation(s)
- Jielian Yang
- Department of OB/GYN, Shanghai Public Health Clinical Center, No. 2901, Caolang Road, Jinshan District, Shanghai, 201508, China
| | - Jinghua Liu
- Department of OB/GYN, Shanghai Public Health Clinical Center, No. 2901, Caolang Road, Jinshan District, Shanghai, 201508, China
| | - Minmin Sheng
- Department of OB/GYN, Shanghai Public Health Clinical Center, No. 2901, Caolang Road, Jinshan District, Shanghai, 201508, China
| | - Xiaohong Zhang
- Department of OB/GYN, Shanghai Public Health Clinical Center, No. 2901, Caolang Road, Jinshan District, Shanghai, 201508, China
| | - Min Liu
- Department of OB/GYN, Shanghai Public Health Clinical Center, No. 2901, Caolang Road, Jinshan District, Shanghai, 201508, China.
| |
Collapse
|
20
|
Hsieh TC, Wu JM. Tumor PD-L1 Induction by Resveratrol/Piceatannol May Function as a Search, Enhance, and Engage ("SEE") Signal to Facilitate the Elimination of "Cold, Non-Responsive" Low PD-L1-Expressing Tumors by PD-L1 Blockade. Int J Mol Sci 2019; 20:ijms20235969. [PMID: 31783532 PMCID: PMC6929199 DOI: 10.3390/ijms20235969] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 11/22/2019] [Accepted: 11/25/2019] [Indexed: 12/26/2022] Open
Abstract
Programmed cell death ligand 1 (PD-L1) is an immune regulatory protein that facilitates tumor escape from host immune surveillance. In the clinic, tumors with high level of PD-L1 have been used to identify patients who might respond favorably to treatment by anti-PD-L1 antibodies (PD-L1 blockade, PLB). Typically, a progression-free response of 9–20% to PLB has been observed, the basis for the low success rate is largely unknown. Recently, we show upregulation of PD-L1 in cancer cells by ≥IC50 supra-pharmacological dose of grape polyphenol resveratrol and piceatannol, alone and combined. Herein, we summarize recent published studies on the regulation of tumor PD-L1 by flavonoids and grape polyphenols. We hypothesize that the induced tumor PD-L1 by resveratrol and/or piceatannol may serve as a Search, Enhance, and Engage (“SEE”) signal to sensitize and augment the recognition and detection of low PD-L1-expressing “cold, non-responsive” tumors. The “SEE” strategy enhances the “visibility” of previously unidentified tumor cells for targeting and eventual eradication by the host antitumor activity. This strategy expands the selection criteria for patients with improved sensitivity and potential responsiveness when used in combination with PLB. The modulation of tumor PD-L1 by flavonoids or polyphenols is proposed to improve the response to PLB in low PD-L1 tumors.
Collapse
|
21
|
Chen X, Chen F, Ren Y, Weng G, Xu L, Xue X, Keng PC, Lee SO, Chen Y. IL-6 signaling contributes to radioresistance of prostate cancer through key DNA repair-associated molecules ATM, ATR, and BRCA 1/2. J Cancer Res Clin Oncol 2019; 145:1471-1484. [PMID: 31020420 DOI: 10.1007/s00432-019-02917-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 04/15/2019] [Indexed: 12/17/2022]
Abstract
PURPOSE To study an association between IL-6 signaling and resistance to radiotherapy of prostate cancer (PCa) and explore the molecular mechanisms involved. METHODS IL-6 expressing C4-2 and CWR22Rv1 (C4-2IL-6/CWRIL-6) and vector control (C4-2vec/CWRvec) cell lines were developed. Radiation-sensitivities of these cells were compared in clonogenic assay, Comet assay, and γH2AX staining. In xenograft animal studies, radiation-sensitivity of C4-2IL-6 cell-derived tumors vs. C4-2vec cell-derived tumors was investigated. To reveal IL-6 downstream molecules involved in DNA repair after radiation, qPCR and Western blot analyses as well as immunofluorescence staining were performed. Transcriptional control of IL-6 on ATM and ATR molecules was also investigated. RESULTS We found C4-2IL-6 and CWRIL-6 cells survived better than their vector control cells after irradiation, and animal studies confirmed such in vitro results. We discovered that DNA repair-related molecules such as ATM, ATR, BRCA1, and BRCA2 were significantly upregulated in irradiated IL-6 expressing cells compared with vector control cells. We further defined that IL-6 signaling regulated cellular expressions of ATM and ATR at the transcriptional level through the activation of Stat3 signaling pathway. CONCLUSIONS IL-6 leads to PCa resistance to radiation through upregulation of DNA repair associated molecules ATM, ATR, BRCA1, and BRCA2.
Collapse
Affiliation(s)
- Xiaodong Chen
- Department of Radiation Oncology, James P. Wilmot Cancer Center, University of Rochester School of Medicine and Dentistry, 601 Elmwood Ave., Box 647, Rochester, NY, 14642, USA
- Department of Urology, Ningbo Urology and Nephrology Hospital, Ningbo, 315100, Zhejiang, People's Republic of China
| | - Feng Chen
- Department of Radiation Oncology, James P. Wilmot Cancer Center, University of Rochester School of Medicine and Dentistry, 601 Elmwood Ave., Box 647, Rochester, NY, 14642, USA
- Department of Urology, Ningbo Urology and Nephrology Hospital, Ningbo, 315100, Zhejiang, People's Republic of China
| | - Yu Ren
- Department of Urology, Ningbo Urology and Nephrology Hospital, Ningbo, 315100, Zhejiang, People's Republic of China
| | - Guobin Weng
- Department of Urology, Ningbo Urology and Nephrology Hospital, Ningbo, 315100, Zhejiang, People's Republic of China
| | - Lijun Xu
- Department of Urology, Ningbo Urology and Nephrology Hospital, Ningbo, 315100, Zhejiang, People's Republic of China
| | - Xiang Xue
- Department of Radiation Oncology, James P. Wilmot Cancer Center, University of Rochester School of Medicine and Dentistry, 601 Elmwood Ave., Box 647, Rochester, NY, 14642, USA
| | - Peter C Keng
- Department of Radiation Oncology, James P. Wilmot Cancer Center, University of Rochester School of Medicine and Dentistry, 601 Elmwood Ave., Box 647, Rochester, NY, 14642, USA
| | - Soo Ok Lee
- Department of Radiation Oncology, James P. Wilmot Cancer Center, University of Rochester School of Medicine and Dentistry, 601 Elmwood Ave., Box 647, Rochester, NY, 14642, USA.
| | - Yuhchyau Chen
- Department of Radiation Oncology, James P. Wilmot Cancer Center, University of Rochester School of Medicine and Dentistry, 601 Elmwood Ave., Box 647, Rochester, NY, 14642, USA.
| |
Collapse
|
22
|
Shevtsov M, Sato H, Multhoff G, Shibata A. Novel Approaches to Improve the Efficacy of Immuno-Radiotherapy. Front Oncol 2019; 9:156. [PMID: 30941308 PMCID: PMC6433964 DOI: 10.3389/fonc.2019.00156] [Citation(s) in RCA: 122] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Accepted: 02/25/2019] [Indexed: 12/31/2022] Open
Abstract
Radiotherapy (RT) has been applied for decades as a treatment modality in the management of various types of cancer. Ionizing radiation induces tumor cell death, which in turn can either elicit protective anti-tumor immune responses or immunosuppression in the tumor micromilieu that contributes to local tumor recurrence. Immunosuppression is frequently accompanied by the attraction of immunosuppressive cells such as myeloid-derived suppressor cells (MDSCs), M2 tumor-associated macrophages (TAMs), T regulatory cells (Tregs), N2 neutrophils, and by the release of immunosuppressive cytokines (TGF-β, IL-10) and chemokines. Immune checkpoint pathways, particularly of the PD-1/PD-L1 axis, have been determined as key regulators of cancer immune escape. While IFN-dependent upregulation of PD-L1 has been extensively investigated, up-to-date studies indicated the importance of DNA damage signaling in the regulation of PD-L1 expression following RT. DNA damage dependent PD-L1 expression is upregulated by ATM/ATR/Chk1 kinase activities and cGAS/STING-dependent pathway, proving the role of DNA damage signaling in PD-L1 induced expression. Checkpoint blockade immunotherapies (i.e., application of anti-PD-1 and anti-PD-L1 antibodies) combined with RT were shown to significantly improve the objective response rates in therapy of various primary and metastatic malignancies. Further improvements in the therapeutic potential of RT are based on combinations of RT with other immunotherapeutic approaches including vaccines, cytokines and cytokine inducers, and an adoptive immune cell transfer (DCs, NK cells, T cells). In the current review we provide immunological rationale for a combination of RT with various immunotherapies as well as analysis of the emerging preclinical evidences for these therapies.
Collapse
Affiliation(s)
- Maxim Shevtsov
- Center for Translational Cancer Research, Technical University of Munich, Klinikum rechts der Isar, Munich, Germany.,Institute of Cytology, Russian Academy of Sciences (RAS), St. Petersburg, Russia.,First Pavlov State Medical University of St. Petersburg, St. Petersburg, Russia.,Almazov National Medical Research Centre, Polenov Russian Scientific Research Institute of Neurosurgery, St. Petersburg, Russia
| | - Hiro Sato
- Department of Radiation Oncology, Graduate School of Medicine, Gunma University, Maebashi, Japan
| | - Gabriele Multhoff
- Center for Translational Cancer Research, Technical University of Munich, Klinikum rechts der Isar, Munich, Germany
| | - Atsushi Shibata
- Education and Research Support Center, Graduate School of Medicine, Gunma University, Maebashi, Japan
| |
Collapse
|
23
|
Escors D, Gato-Cañas M, Zuazo M, Arasanz H, García-Granda MJ, Vera R, Kochan G. The intracellular signalosome of PD-L1 in cancer cells. Signal Transduct Target Ther 2018; 3:26. [PMID: 30275987 PMCID: PMC6160488 DOI: 10.1038/s41392-018-0022-9] [Citation(s) in RCA: 203] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Revised: 05/07/2018] [Accepted: 05/21/2018] [Indexed: 12/18/2022] Open
Abstract
Programmed cell death-1 ligand-1 (PD-L1) overexpression in cancer cells accelerates tumor progression. PD-L1 possesses two main pro-oncogenic functions. First, PD-L1 is a strong immunosuppressive molecule that inactivates tumor-specific T cells by binding to the inhibitory receptor PD-1. Second, PD-L1 function relies on the delivery of intrinsic intracellular signals that enhance cancer cell survival, regulate stress responses and confer resistance toward pro-apoptotic stimuli, such as interferons. Here, we review the current knowledge on intracellular signal transduction pathways regulated by PD-L1, describe its associated signalosome and discuss potential combinations of targeted therapies against the signalosome with PD-L1/PD-1 blockade therapies.
Collapse
Affiliation(s)
- David Escors
- Navarrabiomed, Complejo Hospitalario de Navarra, IdISNA, Irunlarrea 3, 31008 Pamplona, Navarra Spain
- Rayne Institute, Division of Infection and Immunity, University College London, 5 University Street, WC1E 6JF London, UK
| | - María Gato-Cañas
- Navarrabiomed, Complejo Hospitalario de Navarra, IdISNA, Irunlarrea 3, 31008 Pamplona, Navarra Spain
| | - Miren Zuazo
- Navarrabiomed, Complejo Hospitalario de Navarra, IdISNA, Irunlarrea 3, 31008 Pamplona, Navarra Spain
| | - Hugo Arasanz
- Navarrabiomed, Complejo Hospitalario de Navarra, IdISNA, Irunlarrea 3, 31008 Pamplona, Navarra Spain
- Oncology Department, Complejo Hospitalario de Navarra, IdISNA, Irunlarrea 3, 31008 Pamplona, Navarra Spain
| | - María Jesus García-Granda
- Navarrabiomed, Complejo Hospitalario de Navarra, IdISNA, Irunlarrea 3, 31008 Pamplona, Navarra Spain
| | - Ruth Vera
- Oncology Department, Complejo Hospitalario de Navarra, IdISNA, Irunlarrea 3, 31008 Pamplona, Navarra Spain
| | - Grazyna Kochan
- Navarrabiomed, Complejo Hospitalario de Navarra, IdISNA, Irunlarrea 3, 31008 Pamplona, Navarra Spain
| |
Collapse
|
24
|
Lucas J, Hsieh TC, Halicka HD, Darzynkiewicz Z, Wu JM. Upregulation of PD‑L1 expression by resveratrol and piceatannol in breast and colorectal cancer cells occurs via HDAC3/p300‑mediated NF‑κB signaling. Int J Oncol 2018; 53:1469-1480. [PMID: 30066852 PMCID: PMC6086626 DOI: 10.3892/ijo.2018.4512] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Accepted: 06/05/2018] [Indexed: 12/24/2022] Open
Abstract
Programmed cell death ligand 1 (PD-L1) expressed in cancer cells interacting with its receptor programmed cell death 1 (PD-1) expressed in immune cells represents a regulatory axis linked to the suppression and evasion of host immune functions. The blockade of PD-1/PD-L1 interaction using monoclonal antibodies has emerged as an effective therapy for several solid tumors; however, durable response has been observed in a subset of patients with PD-L1-positive tumors. Thus, the understanding of the mechanisms responsible for the expression of PD-L1 in tumor cells may help to improve the response to PD-L1 blockade therapies. In this study, we investigated whether resveratrol, a grape-derived stilbenoid with immunoregulatory activity, modulates the expression of PD-L1 in breast and colorectal cancer cells. The surface expression of PD-L1 was determined by flow cytometry in cancer cells treated with resveratrol and/or piceatannol. Each stilbenoid alone induced PD-L1 and when used in combination, elicited a synergistic upregulation of PD-L1 in some cell lines. The induction of PD-L1 by the combined use of stilbenoids was most pronounced in the Cal51 triple-negative breast cancer (TNBC) and SW620 colon cancer cells. The observed induction of PD-L1 was transcriptionally mediated by nuclear factor (NF)-κB, as shown by NF-κB reporter assays, the nuclear accumulation of the p65 subunit of NF-κB, inhibition by the IKK inhibitor, BMS-345541, and histone the modification inhibitors, resminostat, entinostat or anacardic acid. Combined treatment with resveratrol and piceatannol also decreased tumor cell survival as indicated by the upregulation of the DNA damaging marker, γH2AX, the cleavage of caspase 3, the downregulation of the survival markers, p38-MAPK/c-Myc, and G1-to-S cell cycle arrest.
Collapse
Affiliation(s)
- Justin Lucas
- Department of Biochemistry and Molecular Biology, New York Medical College, Valhalla, NY 10595, USA
| | - Tze-Chen Hsieh
- Department of Biochemistry and Molecular Biology, New York Medical College, Valhalla, NY 10595, USA
| | - H Dorota Halicka
- Brander Cancer Research Institute, Department of Pathology, New York Medical College, Valhalla, NY 10595, USA
| | - Zbigniew Darzynkiewicz
- Brander Cancer Research Institute, Department of Pathology, New York Medical College, Valhalla, NY 10595, USA
| | - Joseph M Wu
- Department of Biochemistry and Molecular Biology, New York Medical College, Valhalla, NY 10595, USA
| |
Collapse
|
25
|
Shen M, Chen Y, Xu L, Zhu R, Xue X, Tsai Y, Keng PC, Lee SO, Chen Y. Increased infiltration of macrophages to radioresistant lung cancer cells contributes to the development of the additional resistance of tumor cells to the cytotoxic effects of NK cells. Int J Oncol 2018; 53:317-328. [PMID: 29750425 DOI: 10.3892/ijo.2018.4394] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Accepted: 04/02/2018] [Indexed: 11/06/2022] Open
Abstract
In this study, in order to investigate the effects of increased macrophage infiltration to radioresistant lung tumors in regulating natural killer (NK) cell-mediated immunity, we examined whether the treatment of radioresistant cells with conditioned medium (CM) from phorbol myristate acetate (PMA)/interleukin (IL)-4 treated THP-1 cells (used as a tumor-associated macrophage source) leads to the development of the additional resistance of tumor cells to NK cell cytotoxicity. We found that the susceptibility of THP-1 CM-treated radioresistant cells to NK cell cytotoxicity was decreased compared to the non-treated cells. In addition, it was found that such a decreased susceptibility was associated with increased programmed death receptor ligand 1 (PD-L1) and decreased natural killer group 2D (NKG2D) ligand levels in tumor cells. We further discovered that the THP-1 cells secreted a high level of IL-6, and that blocking IL-6 action by the addition of a neutralizing antibody (Ab) for IL-6 into the THP-1 CM decreased the resistance of THP-1 CM-treated radioresistant cells to NK cell cytotoxicity. Moreover, we discovered that MEK/Erk was the most critical IL-6 downstream signaling pathway in triggering the THP-1 CM effect; thus, the addition of MEK/Erk inhibitor to THP-1 CM enhanced the susceptibility of the THP-1 CM-treated radioresistant cells to NK cell cytotoxicity. On the whole, the findings of this study suggest the existence of a malignant loop characterized by increased macrophage infiltration into radioresistant cells which, in turn, promotes the development of the additional resistance of these cells to NK cell cytotoxicity.
Collapse
Affiliation(s)
- Mingjing Shen
- Department of Radiation Oncology, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA
| | - Yongbing Chen
- Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, P.R. China
| | - Lijun Xu
- Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, P.R. China
| | - Rongying Zhu
- Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, P.R. China
| | - Xiang Xue
- Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, P.R. China
| | - Ying Tsai
- Department of Radiation Oncology, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA
| | - Peter C Keng
- Department of Radiation Oncology, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA
| | - Soo Ok Lee
- Department of Radiation Oncology, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA
| | - Yuhchyau Chen
- Department of Radiation Oncology, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA
| |
Collapse
|
26
|
Xu L, Shen M, Chen X, Zhu R, Yang DR, Tsai Y, Keng PC, Chen Y, Lee SO. Adipocytes affect castration-resistant prostate cancer cells to develop the resistance to cytotoxic action of NK cells with alterations of PD-L1/NKG2D ligand levels in tumor cells. Prostate 2018; 78:353-364. [PMID: 29330929 DOI: 10.1002/pros.23479] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Accepted: 12/14/2017] [Indexed: 12/29/2022]
Abstract
BACKGROUND Obesity affects prostate cancer (PCa) progression, and the periprostatic adipose tissue adjacent to the prostate is considered a driving force of disease progression. Adipocytes are the main cell population in adipose tissues and their paracrine role contributes to PCa progression, however its implication in modulating immune reactions remains largely unknown. We investigated the adipocyte role in controlling the susceptibility of castration-resistant PCa (CRPC) cells to the cytotoxic action of natural killer (NK) cells. METHODS Using primary NK cells as the NK cell source, NK cell cytotoxicities to CRPC cells, either control media treated or adipocyte-conditioned media (CM) treated, were tested in lactate dehydrogenase (LDH) release-based assays. The levels of programmed death receptor ligand (PD-L1) and NK group 2D (NKG2D) ligands in adipocyte CM-treated CRPC cells were analyzed in qPCR analyses. Effects of blocking adipocyte action on altering PD-L1/NKG2D ligand levels and the susceptibility of CRPC cells to NK cell cytotoxicity were investigated. RESULTS We found NK cell cytotoxicity to CRPC cells decreases when tumor cells are treated with adipocyte CM associated with PD-L1 and NKG2D ligand level alterations. Further, we discovered that the JAK/Stat3 signaling pathway was responsible for the adipocyte CM effect. Two adipokine molecules, IL-6 and leptin, were shown to be important in activation of the JAK/Stat3 signaling in CRPC cells to modulate the PD-L1/NKG2D ligand level alteration. Adding the inhibitors of JAK/Stat3 signaling or neutralizing antibodies of IL-6 or leptin increased the susceptibility of CRPC cells to NK cell action. CONCLUSIONS Blocking the adipocyte effect by inhibiting the IL-6/leptin-JAK/Stat3 signaling axis may enhance NK cell mediated immunity to CRPC cells and this strategy may help to develop future therapeutics to treat obese PCa patients.
Collapse
Affiliation(s)
- Lijun Xu
- Department of Radiation Oncology, University of Rochester School of Medicine and Dentistry, Rochester, New York
- Dep, artment of Urology, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, P.R. China
| | - Mingjing Shen
- Department of Radiation Oncology, University of Rochester School of Medicine and Dentistry, Rochester, New York
- Dep, artment of Urology, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, P.R. China
| | - Xiaodong Chen
- Department of Radiation Oncology, University of Rochester School of Medicine and Dentistry, Rochester, New York
| | - Rongying Zhu
- Dep, artment of Urology, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, P.R. China
| | - Dong-Rong Yang
- Dep, artment of Urology, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, P.R. China
| | - Ying Tsai
- Department of Radiation Oncology, University of Rochester School of Medicine and Dentistry, Rochester, New York
| | - Peter C Keng
- Department of Radiation Oncology, University of Rochester School of Medicine and Dentistry, Rochester, New York
| | - Yuhchyau Chen
- Department of Radiation Oncology, University of Rochester School of Medicine and Dentistry, Rochester, New York
| | - Soo Ok Lee
- Department of Radiation Oncology, University of Rochester School of Medicine and Dentistry, Rochester, New York
| |
Collapse
|
27
|
Immune-Related Concepts in Biology and Treatment of Germ-Cell Tumors. Adv Urol 2018; 2018:3718165. [PMID: 29725351 PMCID: PMC5872660 DOI: 10.1155/2018/3718165] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Revised: 01/22/2018] [Accepted: 02/04/2018] [Indexed: 12/22/2022] Open
Abstract
Germ-cell tumors (GCTs) are highly curable with chemotherapy. Salvage chemotherapy or surgery can cure a proportion of patients, but the ones failing these treatments will die of their disease in the young age. Immune checkpoint pathways are emerging as powerful targetable biomarkers, and a significant preclinical and clinical research is underway to widen our knowledge and expand the treatment possibilities with immune therapy. The concept of immune modulation that was currently adopted in many solid tumors is understudied in GCTs. Herein, we summarize the current knowledge of published literature discussing the immune mechanisms and immune therapy in GCTs.
Collapse
|
28
|
Xu L, Shen M, Chen X, Yang DR, Tsai Y, Keng PC, Lee SO, Chen Y. In vitro-induced M2 type macrophages induces the resistance of prostate cancer cells to cytotoxic action of NK cells. Exp Cell Res 2018; 364:113-123. [PMID: 29408565 DOI: 10.1016/j.yexcr.2018.01.041] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Revised: 01/29/2018] [Accepted: 01/30/2018] [Indexed: 01/02/2023]
Abstract
Previous reports, including our experimental results, showed that macrophages migrate to prostate cancer (PCa) cells. We tested whether the migrated macrophages affect the susceptibility of castration-resistant PCa (CRPC) cells to cytotoxic actions of natural killer (NK) cells. We found treatment of tumor cells with the conditioned media (CM) of the PMA/IL-4 treated THP-1 cells (M2 type macrophages) (THP-1 CM) decreased the susceptibility of tumor cells to NK cell cytotoxicity, as a result of increased programmed death receptor ligand 1 (PD-L1) and decreased NK group 2D (NKG2D) ligands in CRPC cells. Meanwhile, the decreased susceptibility of tumor cells was also detected when NK cells were treated with THP-1 CM and used in NK cell cytotoxicity tests. Therefore, we observed higher resistance of CRPC cells when both tumor and NK cells were treated with THP-1 CM than when tumor cells or NK cells were individually treated. We further discovered that the PMA/IL-4 treated THP-1 cells secrete a high level of IL-6, so blocking the IL-6 action significantly decreased the PD-L1 level while recovering the NKG2D ligands, thus increasing the susceptibility of CRPC cells to NK cell action. Moreover, we discovered that JAK-Stat3 is the most critical IL-6 downstream signaling in triggering the THP-1 CM effect. Consequently, we found the susceptibility of CRPC cells to NK cells was increased when either JAK or Stat 3 inhibitor was added when tumor cells were treated with THP-1 CM, and that the best effect was observed when the JAK inhibitor and PD-L1 Ab were added together.
Collapse
Affiliation(s)
- Lijun Xu
- Department of Radiation Oncology, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA; Department of Urology, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, PR China
| | - Mingjing Shen
- Department of Radiation Oncology, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA; Department of Urology, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, PR China
| | - Xiaodong Chen
- Department of Radiation Oncology, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA
| | - Dong-Rong Yang
- Department of Urology, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, PR China
| | - Ying Tsai
- Department of Radiation Oncology, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA
| | - Peter C Keng
- Department of Radiation Oncology, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA
| | - Soo Ok Lee
- Department of Radiation Oncology, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA.
| | - Yuhchyau Chen
- Department of Radiation Oncology, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA.
| |
Collapse
|
29
|
Xu L, Chen X, Shen M, Yang DR, Fang L, Weng G, Tsai Y, Keng PC, Chen Y, Lee SO. Inhibition of IL-6-JAK/Stat3 signaling in castration-resistant prostate cancer cells enhances the NK cell-mediated cytotoxicity via alteration of PD-L1/NKG2D ligand levels. Mol Oncol 2018; 12:269-286. [PMID: 28865178 PMCID: PMC5830627 DOI: 10.1002/1878-0261.12135] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Revised: 08/04/2017] [Accepted: 08/19/2017] [Indexed: 12/18/2022] Open
Abstract
To investigate whether IL‐6 signaling affects the susceptibility of castration‐resistant prostate cancer (CRPC) cells to cytotoxic action of natural killer (NK) cells, CRPC cell lines (having different IL‐6 levels) were developed by lentiviral transduction. While observing no secreted IL‐6 level in parental C4‐2 and CWR22Rv1 cells, we found the IL‐6 expression/secretion in these cells was induced after the transduction process and the IL‐6 level difference in C4‐2siIL‐6/sc and CWR22siIL‐6/sc cell CRPC cell sets could be detected. We then found that IL‐6‐knockdown cells were more susceptible to NK cell cytotoxicity than control cells due to lowered programmed death receptor ligand 1 (PD‐L1) and increased NK group 2D (NKG2D) ligand levels. In animal studies, to concur with the in vitro results, we found that IL‐6‐expressing cell‐derived tumors were more resistant to NK cell action than the tumors of IL‐6‐knockdown cells. Further, we discovered that JAK‐Stat3 is the most critical IL‐6 downstream signaling that modulates PD‐L1/NKG2D ligand levels in CRPC cells. Furthermore, inhibition of the JAK or Stat3 signaling effectively increased the susceptibility of C4‐2sc and CWRsc cells to NK cell cytotoxicity. We observed the most effective cytotoxicity when the PD‐L1 Ab and JAK inhibitor (or Stat 3 inhibitor) were used together. These results suggest that the strategy of targeting IL‐6 signaling (or its downstream signaling) may enhance the NK cell‐mediated immune action to CRPC tumors, thus yielding clinical implications in developing future immunotherapeutics of exploiting this strategy to treat patients with CRPC.
Collapse
Affiliation(s)
- LiJun Xu
- Department of Radiation Oncology, School of Medicine and Dentistry, University of Rochester, NY, USA.,Department of Urology, The Second Affiliated Hospital, Soochow University, Suzhou, China
| | - XiaoDong Chen
- Department of Radiation Oncology, School of Medicine and Dentistry, University of Rochester, NY, USA.,Department of Urology, Ningbo Urology and Nephrology Hospital, China
| | - MingJing Shen
- Department of Radiation Oncology, School of Medicine and Dentistry, University of Rochester, NY, USA.,Department of Urology, The Second Affiliated Hospital, Soochow University, Suzhou, China
| | - Dong-Rong Yang
- Department of Urology, The Second Affiliated Hospital, Soochow University, Suzhou, China
| | - Laifu Fang
- Department of Pathology, Ningbo Yin Zhou Hospital, China
| | - Guobin Weng
- Department of Urology, Ningbo Urology and Nephrology Hospital, China
| | - Ying Tsai
- Department of Radiation Oncology, School of Medicine and Dentistry, University of Rochester, NY, USA
| | - Peter C Keng
- Department of Radiation Oncology, School of Medicine and Dentistry, University of Rochester, NY, USA
| | - Yuhchyau Chen
- Department of Radiation Oncology, School of Medicine and Dentistry, University of Rochester, NY, USA
| | - Soo Ok Lee
- Department of Radiation Oncology, School of Medicine and Dentistry, University of Rochester, NY, USA
| |
Collapse
|
30
|
Yang YQ, Dong WJ, Yin XF, Xu YN, Yang Y, Wang JJ, Yuan SJ, Xiao J, DeLong JH, Chu L, Xu HN, Zhou XM, Wang RW, Fang L, Liu XY, Zhang KJ. Interferon-related secretome from direct interaction between immune cells and tumor cells is required for upregulation of PD-L1 in tumor cells. Protein Cell 2018; 7:538-43. [PMID: 27295261 PMCID: PMC4930771 DOI: 10.1007/s13238-016-0281-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Affiliation(s)
- Yuan-Qin Yang
- Xinyuan Institute of Medicine and Biotechnology, Zhejiang Sci-Tech University, Hangzhou, 310018, China
| | - Wen-Jie Dong
- Xinyuan Institute of Medicine and Biotechnology, Zhejiang Sci-Tech University, Hangzhou, 310018, China
| | - Xiao-Fei Yin
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Yan-Ni Xu
- College of Life Sciences, Northwest Agriculture and Forestry University, Yangling, 712100, China
| | - Yu Yang
- Central China Normal University, Wuhan, 430079, China
| | - Jiao-Jiao Wang
- Xinyuan Institute of Medicine and Biotechnology, Zhejiang Sci-Tech University, Hangzhou, 310018, China
| | - Su-Jing Yuan
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Jing Xiao
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Jonathan Howard DeLong
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Liang Chu
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Hai-Neng Xu
- Department of Radiation Oncology, University of Pennsylvania Perelman School of Medicine, 3400 Civic Center Blvd., Philadelphia, PA, 19104, USA
| | - Xiu-Mei Zhou
- Xinyuan Institute of Medicine and Biotechnology, Zhejiang Sci-Tech University, Hangzhou, 310018, China
| | - Ru-Wei Wang
- Zhejiang Conba Pharmaceutical Co., Ltd, Hangzhou, 310018, China
| | - Ling Fang
- Zhejiang Conba Pharmaceutical Co., Ltd, Hangzhou, 310018, China
| | - Xin-Yuan Liu
- Xinyuan Institute of Medicine and Biotechnology, Zhejiang Sci-Tech University, Hangzhou, 310018, China.
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, 200031, China.
| | - Kang-Jian Zhang
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, 200031, China.
- Sichuan Huiyang Life Science and Technology Corp., Chengdu, 610021, China.
| |
Collapse
|
31
|
Chovanec M, Cierna Z, Miskovska V, Machalekova K, Svetlovska D, Kalavska K, Rejlekova K, Spanik S, Kajo K, Babal P, Mardiak J, Mego M. Prognostic role of programmed-death ligand 1 (PD-L1) expressing tumor infiltrating lymphocytes in testicular germ cell tumors. Oncotarget 2017; 8:21794-21805. [PMID: 28423520 PMCID: PMC5400624 DOI: 10.18632/oncotarget.15585] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Accepted: 01/10/2017] [Indexed: 12/31/2022] Open
Abstract
PURPOSE Testicular germ cell tumors (TGCTs) are nearly universally curable malignancies. Nevertheless, standard cisplatin-based chemotherapy is not curative in a small subgroup of patients. Previously, we showed that PD-L1 overexpression is associated with worse prognosis in TGCTs, while tumor infiltrating lymphocytes (TILs) are prognostic in different types of cancer. This study aimed to evaluate the prognostic value of PD-1 and PD-L1 expressing TILs in TGCTs. RESULTS PD-L1 positive TILs were found significantly more often in seminomas (95.9% of patients) and embryonal carcinomas (91.0%) compared to yolk sac tumors (60.0%), choriocarcinomas (54.5%) or teratomas (35.7%) (All p < 0.05). TGCTs patients with high infiltration of PD-L1 positive TILs (HS ≥ 160) had significantly better progression-free survival (HR = 0.17, 95% CI 0.09 - 0.31, p = 0.0006) and overall survival (HR = 0.08, 95% CI 0.04 - 0.16, p = 0.001) opposite to patients with lower expression of PD-L1 (HS < 150). PD-1 expressing TILs were not prognostic in TGCTs. MATERIALS AND METHODS Surgical specimens from 240 patients with primary TGCTs were included into this translational study. The PD-1 and PD-L1 expression on tumor and TILs were detected by immunohistochemistry using anti-PD-1 and anti-PD-L1 monoclonal antibody. Scoring was performed semiquantitatively by weighted histoscore (HS) method. CONCLUSIONS The prognostic value of PD-L1 expressing TILs in TGCTs was demonstrated for the first time.
Collapse
Affiliation(s)
- Michal Chovanec
- nd Department of Oncology, Comenius University, Faculty of Medicine & National Cancer Institute, Bratislava, Slovak Republic.,Translational Research Unit, 2nd Department of Oncology, Comenius University, Faculty of Medicine & National Cancer Institute, Bratislava, Slovak Republic.,Department of Medical Oncology, National Cancer Institute, Bratislava, Slovak Republic
| | - Zuzana Cierna
- Department of Pathology, Comenius University, Faculty of Medicine, Bratislava, Slovak Republic
| | - Viera Miskovska
- st Department of Oncology, Comenius University, Faculty of Medicine & St. Elisabeth Cancer Institute, Bratislava, Slovak Republic
| | - Katarina Machalekova
- Department of Pathology, Slovak Medical University St. Elisabeth Cancer Institute, Bratislava, Slovak Republic
| | - Daniela Svetlovska
- Translational Research Unit, 2nd Department of Oncology, Comenius University, Faculty of Medicine & National Cancer Institute, Bratislava, Slovak Republic.,Department of Clinical Trials, National Cancer Institute, Bratislava, Slovak Republic
| | - Katarina Kalavska
- Translational Research Unit, 2nd Department of Oncology, Comenius University, Faculty of Medicine & National Cancer Institute, Bratislava, Slovak Republic.,Department of Medical Oncology, National Cancer Institute, Bratislava, Slovak Republic.,Cancer Research Institute, Slovak Academy of Sciences, Bratislava, Slovak Republic
| | - Katarina Rejlekova
- nd Department of Oncology, Comenius University, Faculty of Medicine & National Cancer Institute, Bratislava, Slovak Republic.,Department of Medical Oncology, National Cancer Institute, Bratislava, Slovak Republic
| | - Stanislav Spanik
- st Department of Oncology, Comenius University, Faculty of Medicine & St. Elisabeth Cancer Institute, Bratislava, Slovak Republic
| | - Karol Kajo
- Department of Pathology, Slovak Medical University St. Elisabeth Cancer Institute, Bratislava, Slovak Republic
| | - Pavel Babal
- Department of Pathology, Comenius University, Faculty of Medicine, Bratislava, Slovak Republic.,Faculty Hospital with Policlinics Skalica, a.s., Skalica, Slovak Republic
| | - Jozef Mardiak
- nd Department of Oncology, Comenius University, Faculty of Medicine & National Cancer Institute, Bratislava, Slovak Republic.,Department of Medical Oncology, National Cancer Institute, Bratislava, Slovak Republic
| | - Michal Mego
- nd Department of Oncology, Comenius University, Faculty of Medicine & National Cancer Institute, Bratislava, Slovak Republic.,Translational Research Unit, 2nd Department of Oncology, Comenius University, Faculty of Medicine & National Cancer Institute, Bratislava, Slovak Republic.,Department of Medical Oncology, National Cancer Institute, Bratislava, Slovak Republic
| |
Collapse
|
32
|
Cioffi M, Trabulo SM, Vallespinos M, Raj D, Kheir TB, Lin ML, Begum J, Baker AM, Amgheib A, Saif J, Perez M, Soriano J, Desco M, Gomez-Gaviro MV, Cusso L, Megias D, Aicher A, Heeschen C. The miR-25-93-106b cluster regulates tumor metastasis and immune evasion via modulation of CXCL12 and PD-L1. Oncotarget 2017; 8:21609-21625. [PMID: 28423491 PMCID: PMC5400610 DOI: 10.18632/oncotarget.15450] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Accepted: 01/10/2017] [Indexed: 12/14/2022] Open
Abstract
The stromal microenvironment controls response to injury and inflammation, and is also an important determinant of cancer cell behavior. However, our understanding of its modulation by miRNA (miR) and their respective targets is still sparse. Here, we identified the miR-25-93-106b cluster and two new target genes as critical drivers for metastasis and immune evasion of cancer cells. Using miR-25-93-106b knockout mice or antagomiRs, we demonstrated regulation of the production of the chemoattractant CXCL12 controlling bone marrow metastasis. Moreover, we identified the immune checkpoint PD-L1 (CD274) as a novel miR-93/106b target playing a central role in diminishing tumor immunity. Eventually, upregulation of miR-93 and miR-106b via miR-mimics or treatment with an epigenetic reader domain (BET) inhibitor resulted in diminished expression of CXCL12 and PD-L1. These data suggest a potential new therapeutic rationale for use of BET inhibitors for dual targeting of cancers with strong immunosuppressive and metastatic phenotypes.
Collapse
Affiliation(s)
- Michele Cioffi
- Stem Cells & Cancer Group, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Sara M Trabulo
- Stem Cells & Cancer Group, Spanish National Cancer Research Centre (CNIO), Madrid, Spain.,Stem Cells in Cancer & Ageing, Barts Cancer Institute, Queen Mary University of London, London, UK
| | - Mireia Vallespinos
- Stem Cells & Cancer Group, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Deepak Raj
- Stem Cells in Cancer & Ageing, Barts Cancer Institute, Queen Mary University of London, London, UK
| | - Tony Bou Kheir
- Stem Cells in Cancer & Ageing, Barts Cancer Institute, Queen Mary University of London, London, UK
| | - Meng-Lay Lin
- Stem Cells in Cancer & Ageing, Barts Cancer Institute, Queen Mary University of London, London, UK
| | - Julfa Begum
- Stem Cells in Cancer & Ageing, Barts Cancer Institute, Queen Mary University of London, London, UK
| | - Ann-Marie Baker
- Tumour Biology, Barts Cancer Institute, Queen Mary University of London, London, UK
| | - Ala Amgheib
- Stem Cells in Cancer & Ageing, Barts Cancer Institute, Queen Mary University of London, London, UK
| | - Jaimy Saif
- School of Clinical Sciences, University of Bristol, Bristol, UK
| | - Manuel Perez
- Confocal Microscopy Unit, Centro Nacional de Investigaciones Oncológicas, Spain
| | - Joaquim Soriano
- Confocal Microscopy Unit, Centro Nacional de Investigaciones Oncológicas, Spain
| | - Manuel Desco
- Departamento de Ingenieria Biomedica e Ingeniería Aeroespacial, Universidad Carlos III de Madrid, Leganés, Spain.,Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain.,Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Madrid, Spain
| | - Maria Victoria Gomez-Gaviro
- Departamento de Ingenieria Biomedica e Ingeniería Aeroespacial, Universidad Carlos III de Madrid, Leganés, Spain.,Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain.,Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Madrid, Spain
| | - Lorena Cusso
- Departamento de Ingenieria Biomedica e Ingeniería Aeroespacial, Universidad Carlos III de Madrid, Leganés, Spain.,Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain.,Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Madrid, Spain
| | - Diego Megias
- Confocal Microscopy Unit, Centro Nacional de Investigaciones Oncológicas, Spain
| | - Alexandra Aicher
- Stem Cells & Cancer Group, Spanish National Cancer Research Centre (CNIO), Madrid, Spain.,Stem Cells in Cancer & Ageing, Barts Cancer Institute, Queen Mary University of London, London, UK
| | - Christopher Heeschen
- Stem Cells & Cancer Group, Spanish National Cancer Research Centre (CNIO), Madrid, Spain.,Stem Cells in Cancer & Ageing, Barts Cancer Institute, Queen Mary University of London, London, UK
| |
Collapse
|
33
|
Enhancing NK cell-mediated cytotoxicity to cisplatin-resistant lung cancer cells via MEK/Erk signaling inhibition. Sci Rep 2017; 7:7958. [PMID: 28801607 PMCID: PMC5554231 DOI: 10.1038/s41598-017-08483-z] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Accepted: 07/11/2017] [Indexed: 01/15/2023] Open
Abstract
Major progress has been made clinically in inhibiting the programmed death receptor 1 (PD-1)/PD-L1 interaction to enhance T cell-mediated immune function, yet the effectiveness of anti-PD-L1/PD-1 agents in enhancing natural killer (NK) cell’s function remains largely unknown. Susceptibilities of cisplatin-resistant A549CisR and H157CisR cells vs. parental cells to the cytotoxic action of NK cells were examined. We found cisplatin-resistant cells more resistant to NK cell cytotoxicity than parental cells. There were constitutively higher expressions of PD-L1 in A549CisR and H157CisR cells than in parental cells in vitro, as well as in H157CisR cell-derived tumors than H157P cell-derived tumors. In contrast, we observed that the expression of PD-1 in NK cells was induced after co-culture with cisplatin-resistant cells. We also observed increased susceptibility of cisplatin-resistant cells to NK cell cytotoxicity when neutralizing antibody of PD-1 or PD-L1 was added. Further, we found that the NK group 2, member D (NKG2D) ligand levels were lower in A549CisR and H157CisR cells than in parental cells. Meanwhile, we discovered that the MEK/Erk signaling pathway played a significant role in this regulation, and the addition of a MEK/Erk pathway inhibitor significantly enhanced the PD-L1 Ab effect in enhancing NK cell cytotoxicity to cisplatin-resistant cells.
Collapse
|
34
|
Park IJ, An S, Kim SY, Lim HM, Hong SM, Kim MJ, Kim YJ, Yu CS. Prediction of radio-responsiveness with immune-profiling in patients with rectal cancer. Oncotarget 2017; 8:79793-79802. [PMID: 29108360 PMCID: PMC5668093 DOI: 10.18632/oncotarget.19558] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Accepted: 07/14/2017] [Indexed: 12/18/2022] Open
Abstract
We evaluate whether the tumor immune infiltrate (TIL) could be used for prediction of responsiveness to preoperative chemoradiotherapy (PCRT) in rectal cancers. Using formalin-fixed paraffin-embedded slides of pretreatment biopsies, co-stain for CD4, CD8, CD274 (PD-L1), FOXP3, cytokeratin, and DAPI was performed with Opal multi staining kit (Perkin-Elmer, Waltham, MA). Multispectral imaging and digital analysis to visualize and quantify specific immune infiltrates were performed using the Vectra imaging system (Perkin-Elmer). The density (number of cells per mm2) and proportion of total TILs and specific cell types in the stroma were calculated by inForm™ 2.2.1 software (Perkin-Elmer). The density and proportion of total TILs and specific cell types in the stroma were calculated by inForm™ 2.2.1 software (Perkin-Elmer, Waltham, MA). Patients were classified as group with total regression (TR, n = 25) and group with residual disease (near total, moderate, and minimal regression, RD, n = 50). The mean density of T cell infiltration and CD274 (PD-L1)+ lymphocyte were significantly higher in TR (p = 0.005, p = 0.001). The proportion of CD4+ lymphocyte (p=0.042) and CD274 (PD-L1)+ lymphocyte (p = 0.002) were different between 2 groups. The TR group has lower CD4+ and higher CD274 (PD-L1)+ proportions than RD group. The ratio among CD4+, CD8+, CD274 (PD-L1)+, FOXP3+ T cell was different between groups. TR group showed lower CD4/ CD274 (PD-L1) (p = 0.007), CD8/ CD274 (PD-L1) (p = 0.02), and FOXP3/ CD274 (PD-L1) (p = 0.003) ratio than RD group. The determination of the immune infiltrate in biopsies before treatment could be a valuable information for the prediction of responsiveness to PCRT.
Collapse
Affiliation(s)
- In Ja Park
- Department of Colon and Rectal Surgery, University of Ulsan College of Medicine and Asan Medical Center, Seoul, Korea
| | - Soyeon An
- Department of Pathology, Incheon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Incheon, Korea
| | - Sang-Yeob Kim
- Department of Convergence Medicine, University of Ulsan College of Medicine and Asan Medical Center, Seoul, Korea.,Asan Institute for life sciences, Asan Medical Center, Seoul, Korea
| | - Hye Min Lim
- Asan Institute for life sciences, Asan Medical Center, Seoul, Korea
| | - Seung-Mo Hong
- Department of Pathology, University of Ulsan College of Medicine and Asan Medical Center, Seoul, Korea
| | - Mi-Ju Kim
- Asan Institute for life sciences, Asan Medical Center, Seoul, Korea
| | - Yun Jae Kim
- Asan Institute for life sciences, Asan Medical Center, Seoul, Korea
| | - Chang Sik Yu
- Department of Colon and Rectal Surgery, University of Ulsan College of Medicine and Asan Medical Center, Seoul, Korea
| |
Collapse
|
35
|
Shen MJ, Xu LJ, Yang L, Tsai Y, Keng PC, Chen Y, Lee SO, Chen Y. Radiation alters PD-L1/NKG2D ligand levels in lung cancer cells and leads to immune escape from NK cell cytotoxicity via IL-6-MEK/Erk signaling pathway. Oncotarget 2017; 8:80506-80520. [PMID: 29113321 PMCID: PMC5655216 DOI: 10.18632/oncotarget.19193] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Accepted: 06/28/2017] [Indexed: 02/07/2023] Open
Abstract
We investigated whether radiation influences the susceptibility of non-small cell lung cancer (NSCLC) cells to NK cell mediated cytotoxicity. We found radiation treatment increased expression of programmed cell death ligand 1 (PD-L1), but decreased NK group 2, member D (NKG2D) ligand expressions in A549 and H157 NSCLC cells. Both types of changes would have protected tumor cells from the cytotoxic action of NK cells. Consistently, we detected similar alteration in these molecules in radioresistant A549R26-1 and H157R24-1 subline cells. Higher PD-L1 level was also observed in tumors of A549R26-1 cell-derived xenografts than tumors of parental A549 (A549P) cell-derived xenografts. Accordingly, we found radioresistant cells were more resistant to the cytotoxic action of NK cells than parental cells, and such resistance was decreased when neutralizing antibody (Ab) of PD-L1 was added to the radioresistant cell/NK cell co-cultures. In mechanism studies, we found that IL-6-MEK/Erk signaling contributed most significantly to the up-regulation of PD-L1/down-regulation of NKG2D ligands in radioresistant cells. The addition of the MEK/Erk inhibitor increased the susceptibility of A549R26-1 and H157R24-1 cells to NK-cell cytotoxicity while no significant effect was observed in parental cells. Moreover, we detected enhanced NK-cell cytotoxicity to radioresistant cells when PD-L1 Ab and MEK/Erk inhibitor were added together to co-cultures of tumor/NK cells compared to when PD-L1 Ab was used alone. We suggest that combined use of PD-L1 Ab and MEK/Erk inhibitor may offer better therapeutic benefits than PD-L1 Ab alone to treat NSCLC patients who are receiving radiotherapy or who are at the radioresistant stage.
Collapse
Affiliation(s)
- Ming Jing Shen
- Department of Radiation Oncology, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA.,Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, P.R. China
| | - Li Jun Xu
- Department of Radiation Oncology, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA.,Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, P.R. China
| | - Li Yang
- Department of Radiation Oncology, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA
| | - Ying Tsai
- Department of Radiation Oncology, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA
| | - Peter C Keng
- Department of Radiation Oncology, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA
| | - Yongbing Chen
- Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, P.R. China
| | - Soo Ok Lee
- Department of Radiation Oncology, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA
| | - Yuhchyau Chen
- Department of Radiation Oncology, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA
| |
Collapse
|
36
|
Siebert N, Zumpe M, Jüttner M, Troschke-Meurer S, Lode HN. PD-1 blockade augments anti-neuroblastoma immune response induced by anti-GD 2 antibody ch14.18/CHO. Oncoimmunology 2017; 6:e1343775. [PMID: 29123953 DOI: 10.1080/2162402x.2017.1343775] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Revised: 06/12/2017] [Accepted: 06/12/2017] [Indexed: 10/19/2022] Open
Abstract
Immunotherapy with anti-GD2 antibody (Ab) ch14.18/CHO is effective for treatment of high-risk neuroblastoma (NB) patients and is mainly based on GD2-specific Ab-dependent cellular cytotoxicity (ADCC). Strategies to further enhance the efficacy are important and currently explored in prospective clinical trials randomizing ch14.18/CHO ± IL-2. Recently, expression of programmed death 1 (PD-1) inhibitory receptor by effector cells and its ligand (PD-L1) by tumor cells has been shown. Here, we report for the first time effects of PD-1 blockade on ch14.18/CHO-based immunotherapy and mechanisms involved. Expression of PD-1 and PD-L1 on NB and effector cells was analyzed by RT-PCR and flow cytometry in the presence of ch14.18/CHO and/or IL-2. The effect of PD-1 blockade on ch14.18/CHO-mediated anti-NB immune response was evaluated using anti-PD-1 Ab both in vitro (Nivolumab) and in a syngeneic PD-L1+/GD2+ NB mouse model (anti-mouse PD-1). Culture of NB cells LA-N-1 (low PD-L1 baseline expression) with leukocytes and subtherapeutic ch14.18/CHO concentrations for 24 h induced strong upregulation of PD-L1, which was further increased by IL-2 resulting in complete inhibition of ch14.18/CHO-mediated ADCC. Importantly, blockade with Nivolumab reversed the PD-L1-dependent inhibition of ADCC. Similarly, co-incubation with anti-CD11b Ab abrogated the PD-L1 upregulation and restored ADCC. Mice treated with ch14.18/CHO in combination with PD-1 blockade showed a strong reduction of tumor growth, prolonged survival and the highest cytotoxicity against NB cells. In conclusion, ch14.18/CHO-mediated effects upregulate the inhibitory immune checkpoint PD-1/PD-L1, and combination of ch14.18/CHO with PD-1 blockade results in synergistic treatment effects in mice representing a new effective treatment strategy against GD2-positive cancers.
Collapse
Affiliation(s)
- Nikolai Siebert
- Department of Pediatric Oncology and Hematology, University Medicine Greifswald, Greifswald, Germany
| | - Maxi Zumpe
- Department of Pediatric Oncology and Hematology, University Medicine Greifswald, Greifswald, Germany
| | - Madlen Jüttner
- Department of Pediatric Oncology and Hematology, University Medicine Greifswald, Greifswald, Germany
| | - Sascha Troschke-Meurer
- Department of Pediatric Oncology and Hematology, University Medicine Greifswald, Greifswald, Germany
| | - Holger N Lode
- Department of Pediatric Oncology and Hematology, University Medicine Greifswald, Greifswald, Germany
| |
Collapse
|
37
|
Regulation of PD-L1 expression on murine tumor-associated monocytes and macrophages by locally produced TNF-α. Cancer Immunol Immunother 2017; 66:523-535. [PMID: 28184968 DOI: 10.1007/s00262-017-1955-5] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Accepted: 01/15/2017] [Indexed: 12/22/2022]
Abstract
PD-L1 is an immune checkpoint protein that has emerged as a major signaling molecule involved with tumor escape from T cell immune responses. Studies have shown that intra-tumoral expression of PD-L1 can inhibit antitumor immune responses. However, it has recently been shown that expression of PD-L1 on myeloid cells from the tumor is a stronger indicator of prognosis than tumor cell PD-L1 expression. Therefore, it is important to understand the factors that govern the regulation of PD-L1 expression on tumor-infiltrating myeloid cells. We found that immature bone marrow monocytes in tumor-bearing mice had low levels of PD-L1 expression, while higher levels of expression were observed on monocytes in circulation. In contrast, macrophages found in tumor tissues expressed much higher levels of PD-L1 than circulating monocytes, implying upregulation by the tumor microenvironment. We demonstrated that tumor-conditioned media strongly induced increased PD-L1 expression by bone marrow-derived monocytes and TNF-α to be a cytokine that causes an upregulation of PD-L1 expression by the monocytes. Furthermore, we found production of TNF-α by the monocytes themselves to be a TLR2-dependent response to versican secreted by tumor cells. Thus, PD-L1 expression by tumor macrophages appears to be regulated in a different manner than by tumor cells themselves.
Collapse
|
38
|
Black M, Barsoum IB, Truesdell P, Cotechini T, Macdonald-Goodfellow SK, Petroff M, Siemens DR, Koti M, Craig AWB, Graham CH. Activation of the PD-1/PD-L1 immune checkpoint confers tumor cell chemoresistance associated with increased metastasis. Oncotarget 2016; 7:10557-67. [PMID: 26859684 PMCID: PMC4891140 DOI: 10.18632/oncotarget.7235] [Citation(s) in RCA: 137] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Accepted: 01/25/2016] [Indexed: 12/13/2022] Open
Abstract
The ability of tumor cells to avoid immune destruction (immune escape) as well as their acquired resistance to anti-cancer drugs constitute important barriers to the successful management of cancer. Interaction between the Programmed Death Ligand 1 (PD-L1) on the surface of tumor cells with the Programmed Death-1 (PD-1) receptor on cytotoxic T lymphocytes leads to inactivation of these immune effectors and, consequently, immune escape. Here we show that the PD-1/PD-L1 axis also leads to tumor cell resistance to conventional chemotherapeutic agents. Using a panel of PD-L1-expressing human and mouse breast and prostate cancer cell lines, we found that incubation of breast and prostate cancer cells in the presence of purified recombinant PD-1 resulted in resistance to doxorubicin and docetaxel as determined using clonogenic survival assays. Co-culture with PD-1-expressing Jurkat T cells also promoted chemoresistance and this was prevented by antibody blockade of either PD-L1 or PD-1 or by silencing of the PD-L1 gene. Moreover, inhibition of the PD-1/PD-L1 axis using anti-PD-1 antibody enhanced doxorubicin chemotherapy to inhibit metastasis in a syngeneic mammary orthotopic mouse model of metastatic breast cancer. To further investigate the mechanism of tumor cell survival advantage upon PD-L1 ligation, we show that exposure to rPD-1 promoted ERK and mTOR growth and survival pathways leading to increased cell proliferation. Overall, the findings of this study indicate that combinations of chemotherapy and immune checkpoint blockade may limit chemoresistance and progression to metastatic disease.
Collapse
Affiliation(s)
- Madison Black
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Ivraym B Barsoum
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada.,Department of Urology, Queen's University, Kingston, Ontario, Canada
| | - Peter Truesdell
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada.,Cancer Biology and Genetics, Queen's Cancer Research Institute, Kingston, Ontario, Canada
| | - Tiziana Cotechini
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | | | - Margaret Petroff
- Department of Pathobiology and Diagnostic Investigation, Michigan State University, East Lansing, Michigan, USA
| | - D Robert Siemens
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada.,Department of Urology, Queen's University, Kingston, Ontario, Canada
| | - Madhuri Koti
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada.,Cancer Biology and Genetics, Queen's Cancer Research Institute, Kingston, Ontario, Canada
| | - Andrew W B Craig
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada.,Cancer Biology and Genetics, Queen's Cancer Research Institute, Kingston, Ontario, Canada
| | - Charles H Graham
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada.,Department of Urology, Queen's University, Kingston, Ontario, Canada.,Cancer Biology and Genetics, Queen's Cancer Research Institute, Kingston, Ontario, Canada
| |
Collapse
|
39
|
The Roles of Mesenchymal Stromal/Stem Cells in Tumor Microenvironment Associated with Inflammation. Mediators Inflamm 2016; 2016:7314016. [PMID: 27630452 PMCID: PMC5007366 DOI: 10.1155/2016/7314016] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Revised: 07/15/2016] [Accepted: 07/27/2016] [Indexed: 02/08/2023] Open
Abstract
State of tumor microenvironment (TME) is closely linked to regulation of tumor growth and progression affecting the final outcome, refractoriness, and relapse of disease. Interactions of tumor, immune, and mesenchymal stromal/stem cells (MSCs) have been recognized as crucial for understanding tumorigenesis. Due to their outstanding features, stem cell-like properties, capacity to regulate immune response, and dynamic functional phenotype dependent on microenvironmental stimuli, MSCs have been perceived as important players in TME. Signals provided by tumor-associated chronic inflammation educate MSCs to alter their phenotype and immunomodulatory potential in favor of tumor-biased state of MSCs. Adjustment of phenotype to TME and acquisition of tumor-promoting ability by MSCs help tumor cells in maintenance of permissive TME and suppression of antitumor immune response. Potential utilization of MSCs in treatment of tumor is based on their inherent ability to home tumor tissue that makes them suitable delivery vehicles for immune-stimulating factors and vectors for targeted antitumor therapy. Here, we review data regarding intrusive effects of inflammatory TME on MSCs capacity to affect tumor development through modification of their phenotype and interactions with immune system.
Collapse
|
40
|
Abstract
The term “immune synapse” was originally coined to highlight the similarities between the synaptic contacts between neurons in the central nervous system and the cognate, antigen-dependent interactions between T cells and antigen-presenting cells. Here, instead of offering a comprehensive molecular catalogue of molecules involved in the establishment, stabilization, function, and resolution of the immune synapse, we follow a spatiotemporal timeline that begins at the initiation of exploratory contacts between the T cell and the antigen-presenting cell and ends with the termination of the contact. We focus on specific aspects that distinguish synapses established by cytotoxic and T helper cells as well as unresolved issues and controversies regarding the formation of this intercellular structure.
Collapse
Affiliation(s)
- Alvaro Ortega-Carrion
- Immunology Section, Department of Medicine, Universidad Autonoma de Madrid School of Medicine, Madrid, Spain
| | - Miguel Vicente-Manzanares
- Immunology Section, Department of Medicine, Universidad Autonoma de Madrid School of Medicine, Madrid, Spain
| |
Collapse
|
41
|
Chen MF, Chen PT, Chen WC, Lu MS, Lin PY, Lee KD. The role of PD-L1 in the radiation response and prognosis for esophageal squamous cell carcinoma related to IL-6 and T-cell immunosuppression. Oncotarget 2016; 7:7913-24. [PMID: 26761210 PMCID: PMC4884963 DOI: 10.18632/oncotarget.6861] [Citation(s) in RCA: 104] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Accepted: 01/02/2016] [Indexed: 12/21/2022] Open
Abstract
The aim of this study was to assess the significance of programmed cell death 1 ligand 1 (PD-L1) in esophageal squamous cell carcinoma (ESCC) and its association with IL-6 and radiation response. Weretrospectively enrolled 162 patients with ESCC, and examined the correlation between PD-L1 levels and clinical outcomes in esophageal cancer patients. Furthermore, the human esophageal SCC cell line CE81T and TE2 were selected for cellular experiments to investigate the role of PD-L1 in T cell functions and radiation response. Here we demonstrated that PD-L1 expression was significantly higher in esophageal cancer specimens than in non-malignant epithelium. In clinical outcome analysis, this staining of PD-L1 was positively linked to the clinical T4 stage (p=0.004), development of LN metastasis (p=0.012) and higher loco-regional failure rate (p=0.0001). In addition, the frequency of PD-L1 immunoreactivity was significantly higher in IL-6-positive esophageal cancer specimens. When IL-6 signaling was inhibited in vitro, the level of PD-L1 is significantly down-regulated. PD-L1 is a significant predictor for poor treatment response and shorter survival.As demonstrated through in vitro experiments, Irradiation increased PD-L1 expression in human esophageal cancer cells. The inhibition of T cell functions including proliferation and cytotoxicity against tumor cells might be the mechanisms responsible to the role of PD-L1 in radiation response. In conclusion, PD-L1 is important in determining the radiation response and could predict the prognosis of patients with esophageal SCC. Therefore, we suggest inhibition of PD-L1 as a potential strategy for the treatment of esophageal SCC.
Collapse
MESH Headings
- B7-H1 Antigen/metabolism
- Biomarkers, Tumor/blood
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/pathology
- Carcinoma, Squamous Cell/immunology
- Carcinoma, Squamous Cell/metabolism
- Carcinoma, Squamous Cell/secondary
- Carcinoma, Squamous Cell/therapy
- Chemoradiotherapy
- Enzyme-Linked Immunosorbent Assay
- Esophageal Neoplasms/immunology
- Esophageal Neoplasms/metabolism
- Esophageal Neoplasms/pathology
- Esophageal Neoplasms/therapy
- Female
- Flow Cytometry
- Follow-Up Studies
- Humans
- Immunoenzyme Techniques
- Interleukin-6/blood
- Lymphatic Metastasis
- Lymphocytes, Tumor-Infiltrating/immunology
- Lymphocytes, Tumor-Infiltrating/pathology
- Male
- Middle Aged
- Neoplasm Recurrence, Local/immunology
- Neoplasm Recurrence, Local/metabolism
- Neoplasm Recurrence, Local/pathology
- Neoplasm Recurrence, Local/therapy
- Neoplasm Staging
- Prognosis
- Survival Rate
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- Miao-Fen Chen
- Department of Radiation Oncology, Chang Gung Memorial Hospital at Chiayi, Chiayi, Taiwan
- College of Medicine, Chang Gung University, Chiayi, Taiwan
| | - Ping-Tsung Chen
- College of Medicine, Chang Gung University, Chiayi, Taiwan
- Department of Hematology and Oncology, Chang Gung Memorial Hospital at Chiayi, Chiayi, Taiwan
| | - Wen-Cheng Chen
- Department of Radiation Oncology, Chang Gung Memorial Hospital at Chiayi, Chiayi, Taiwan
- College of Medicine, Chang Gung University, Chiayi, Taiwan
| | - Ming-Shian Lu
- Department of Thoracic & Cardiovascular Surgery, Chang Gung Memorial Hospital at Chiayi, Chiayi, Taiwan
| | - Paul-Yang Lin
- Department of Pathology, Chang Gung Memorial Hospital at Chiayi, Chiayi, Taiwan
| | - Kuan-Der Lee
- College of Medicine, Chang Gung University, Chiayi, Taiwan
- Department of Hematology and Oncology, Chang Gung Memorial Hospital at Chiayi, Chiayi, Taiwan
| |
Collapse
|
42
|
Wu CT, Chen WC, Chang YH, Lin WY, Chen MF. The role of PD-L1 in the radiation response and clinical outcome for bladder cancer. Sci Rep 2016; 6:19740. [PMID: 26804478 PMCID: PMC4726250 DOI: 10.1038/srep19740] [Citation(s) in RCA: 136] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2015] [Accepted: 12/16/2015] [Indexed: 12/19/2022] Open
Abstract
Identification of potential factors that can stratify a tumor's response to specific therapies will aid in the selection of cancer therapy. The aim was to highlight the role of programmed cell death 1 ligand 1 (PD-L1) in bladder cancer. In this study, 92 of muscle-invasive bladder cancers and 28 of non-muscle invasive bladder cancers were selected for immunohistochemical staining analysis. Furthermore, human and murine bladder cancer cell lines were used to examine the correlation between PD-L1 and radiation response. Our data revealed that PD-L1 was overexpressed in the bladder tumor specimens compared with adjacent non-malignant specimens. Furthermore, the staining of PD-L1 was significantly linked to higher clinical stage, lower complete response rates and reduced disease-free survival rates. By in vitro and in vivo experiments, irradiation up-regulated the expression of PD-L1 in tumor cells, and its increase correlated with the irradiation dose. In immunocompetent mouse models, blocking PD-L1 induced a longer tumour growth delay following irradiation. The inhibition of T cell functions including proliferation and cytotoxicity against tumor cells was responsible to the effects of PD-L1 on radiation response. In conclusion, PD-L1 could be a significant clinical predictor for clinical stage and treatment response of bladder cancer.
Collapse
Affiliation(s)
- Chun-Te Wu
- Department of Urology, Chang Gung Memorial Hospital at Keelung, Taiwan
- Chang Gung University, College of Medicine, Taoyuan, Taiwan
| | - Wen-Cheng Chen
- Chang Gung University, College of Medicine, Taoyuan, Taiwan
- Department of Radiation Oncology, Chang Gung Memorial Hospital at Chiayi, Taiwan
| | - Ying-Hsu Chang
- Department of Urology, Chang Gung Memorial Hospital at Linko, Taiwan
| | - Wei-Yu Lin
- Chang Gung University, College of Medicine, Taoyuan, Taiwan
- Urology, Chang Gung Memorial Hospital at Chiayi, Taiwan
| | - Miao-Fen Chen
- Chang Gung University, College of Medicine, Taoyuan, Taiwan
- Department of Radiation Oncology, Chang Gung Memorial Hospital at Chiayi, Taiwan
| |
Collapse
|
43
|
Cierna Z, Mego M, Miskovska V, Machalekova K, Chovanec M, Svetlovska D, Hainova K, Rejlekova K, Macak D, Spanik S, Ondrus D, Kajo K, Mardiak J, Babal P. Prognostic value of programmed-death-1 receptor (PD-1) and its ligand 1 (PD-L1) in testicular germ cell tumors. Ann Oncol 2015; 27:300-5. [PMID: 26598537 DOI: 10.1093/annonc/mdv574] [Citation(s) in RCA: 112] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2015] [Accepted: 11/09/2015] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Testicular germ cell tumors (TGCTs) belong to the most chemosensitive solid tumors; however, a small proportion of patients fail to be cured with cisplatin-based chemotherapy. Inhibitors of PD-1/PD-L1 pathways represent a new class of promising drugs in anticancer therapy. The aim of this study was to evaluate expression and prognostic value of PD-1 and PD-L1 in TGCTs. PATIENTS AND METHODS Surgical specimens from 140 patients with TGCTs (131 with primary testicular tumor and 9 with extragonadal GCTs) were included into the translational study. PD-1 and PD-L1 expression was detected in the tumor tissue by immunohistochemistry using monoclonal antibodies, scored by the multiplicative quickscore (QS) method, compared with their expression in normal testicular tissue and correlated with clinicopathological characteristics and clinical outcome. RESULTS None of the GCTs exhibited PD-1 protein, although expression of PD-L1 was significantly higher in GCTs in comparison with normal testicular tissue (mean QS = 5.29 versus 0.32, P < 0.0001). Choriocarcinomas exhibit the highest level of PD-L1 with decreasing positivity in embryonal carcinoma, teratoma, yolk sac tumor and seminoma. PD-L1 expression was associated with poor prognostic features, including ≥3 metastatic sites, increased serum tumor markers and/or non-pulmonary visceral metastases. Patients with low PD-L1 expression had significantly better progression-free survival [hazard ratio (HR) = 0.40, 95% confidence interval (CI) 0.16-1.01, P = 0.008] and overall survival (HR = 0.43, 95% CI 0.15-1.23, P = 0.040) compared with patients with high PD-L1 expression. CONCLUSIONS In this translational study, we showed, for the first time, the prognostic value of PD-L1 expression in TGCTs and our data imply that the PD-1/PD-L1 pathway could be a novel therapeutic target in TGCTs.
Collapse
Affiliation(s)
- Z Cierna
- Department of Pathology, Faculty of Medicine
| | - M Mego
- 2nd Department of Oncology, Faculty of Medicine and National Cancer Institute Translational Research Unit, 2nd Department of Oncology, Faculty of Medicine and National Cancer Institute 1st Department of Oncology, Comenius University, Bratislava
| | - V Miskovska
- Faculty of Medicine, St Elisabeth Cancer Institute, Bratislava
| | - K Machalekova
- Department of Pathology, Slovak Medical University and St Elisabeth Cancer Institute, Bratislava
| | - M Chovanec
- 2nd Department of Oncology, Faculty of Medicine and National Cancer Institute 1st Department of Oncology, Comenius University, Bratislava
| | - D Svetlovska
- Translational Research Unit, 2nd Department of Oncology, Faculty of Medicine and National Cancer Institute
| | - K Hainova
- Translational Research Unit, 2nd Department of Oncology, Faculty of Medicine and National Cancer Institute Cancer Research Institute, Slovak, Academy of Sciences, Bratislava
| | - K Rejlekova
- 2nd Department of Oncology, Faculty of Medicine and National Cancer Institute 1st Department of Oncology, Comenius University, Bratislava
| | - D Macak
- Department of Pathology, National Cancer Institute, Bratislava
| | - S Spanik
- Faculty of Medicine, St Elisabeth Cancer Institute, Bratislava
| | - D Ondrus
- Faculty of Medicine, St Elisabeth Cancer Institute, Bratislava
| | - K Kajo
- Department of Pathology, Slovak Medical University and St Elisabeth Cancer Institute, Bratislava
| | - J Mardiak
- 2nd Department of Oncology, Faculty of Medicine and National Cancer Institute Translational Research Unit, 2nd Department of Oncology, Faculty of Medicine and National Cancer Institute 1st Department of Oncology, Comenius University, Bratislava
| | - P Babal
- Department of Pathology, Faculty of Medicine Faculty Hospital with Policlinics Skalica, a.s., Skalica, Slovak Republic
| |
Collapse
|
44
|
Guo Z, Wang H, Meng F, Li J, Zhang S. Combined Trabectedin and anti-PD1 antibody produces a synergistic antitumor effect in a murine model of ovarian cancer. J Transl Med 2015. [PMID: 26219551 PMCID: PMC4517526 DOI: 10.1186/s12967-015-0613-y] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
BACKGROUND Monoclonal antibodies (mAb) that block programmed death (PD)-1 signaling pathway hold great potential as a novel cancer immunotherapy. Recent evidence suggests that combining with conventional, targeted or other immunotherapies, these mAb can induce synergistic antitumor responses. In this study, we investigated whether Trabectedin (ET-743), a novel anticancer agent currently used for treating relapsed ovarian cancer, can synergize with anti (α)-PD-1 mAb to increase antitumor activity in the murine ID8 ovarian cancer model. METHODS Mice with established peritoneal ID8 tumor were treated with either single or combined Trabectedin and α-PD-1 mAb, their overall survival was recorded; tumor-associated immune cells and immune gene expression in tumors from treated mice were analyzed by flow cytometry and quantitative RT-PCR, respectively, and antigen-specific immunity of effector CD8(+) T cells was evaluated by ELISA and cytotoxicity assay. In addition, the effect of Trabectedin on tumoral PD-L1 expression was analyzed by both flow cytometry and immunofluorescence staining. RESULTS Though single treatment showed a modest antitumor effect in mice bearing 10-day-established ID8 tumor, combined Trabectedin and α-PD-1 mAb treatment induced a strong antitumor immune response, leading to a significant tumor regression with half of mice tumor-free 90 days after tumor inoculation. Mechanistic investigation revealed that combination treatment induces a systemic tumor-specific immunity with an indispensable role of both CD4(+) and CD8(+) T cells, and effector CD8(+) T cells exhibited the antigen-specific cytokine secretion and cytotoxicity upon tumor antigen stimulation; additionally, combination treatment increased the IFN-γ-producing effector T cells and decreased the immunosuppressive cells in peritoneal cavity; accordingly, it enhanced the expression of Th1-associated immune-stimulating genes while reducing the transcription of regulatory/suppressive immune genes, reshaping tumor microenvironment from a immunosuppressive to a stimulatory state. Finally, in vivo Trabectedin treatment has been shown to induce IFN-γ-dependent PD-L1 expression within tumor, possibly constituting a mechanistic basis for its synergistic antitumor effect with α-PD-1 mAb therapy. CONCLUSION This study provides the evidence that α-PD-1 mAb can produce a synergistic antitumor efficacy when combined with Trabectedin, a clinically available anticancer agent, supporting a direct translation of this combination strategy in clinic for the treatment of ovarian cancer.
Collapse
Affiliation(s)
- Zhiqiang Guo
- Department of Gynecology and Obstetrics, Shengjing Hospital, China Medical University, Shenyang, 110004, China.
| | - Haolin Wang
- Department of Acute Abdominal Surgery, The First Hospital of Dalian Medical University, Dalian, 116044, China.
| | - Fandong Meng
- Molecular Oncology Department of Cancer Research Institution, The First Hospital of China Medical University, Shenyang, 110004, China.
| | - Jie Li
- Department of Gynecology and Obstetrics, Shengjing Hospital, China Medical University, Shenyang, 110004, China.
| | - Shulan Zhang
- Department of Gynecology and Obstetrics, Shengjing Hospital, China Medical University, Shenyang, 110004, China.
| |
Collapse
|