1
|
Wang Y, Qu K, Xia Z, Qi M, Du X, Ke Z, Zhang R. Selenoprotein S (SELENOS) is a potential prognostic biomarker for brain lower grade glioma. J Trace Elem Med Biol 2024; 86:127539. [PMID: 39378668 DOI: 10.1016/j.jtemb.2024.127539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 09/06/2024] [Accepted: 09/26/2024] [Indexed: 10/10/2024]
Abstract
BACKGROUND Selenium, an essential micronutrient, primarily exists as selenocysteine in various selenoproteins. Selenoprotein S (SELENOS) is crucial in the development of human cancer. This study aimed to explore the correlation between SELENOS gene expression and the prognosis of brain lower-grade glioma (LGG). METHODS SELENOS protein and mRNA expression in human normal and tumor tissues were explored through the HPA database. SELENOS expression differences between normal and tumor tissues, along with its prognostic significance in gliomas, were analyzed using the TCGA, GTEx datasets, while the CGGA dataset was used to further assess its prognostic potential in a Chinese cohort. The association between SELENOS expression and tumor immune infiltration was also assessed. Multivariate and univariate Cox models were used to screen for clinicopathological parameters associated with SELENOS expression. The GDSC datasets was utilized to explore the connection between SELENOS and chemotherapeutic responses in LGG. A protein-protein interaction network for SELENOS was created. SELENOS expression in LGG cell lines were determined by Western blotting and qRT-PCR, and its functions were ascertained by routine in vitro experiments. RESULTS SELENOS was upregulated in 11 cancers and downregulated in 10 cancers relative to the corresponding normal tissues, and correlated significantly with the prognosis, especially for GBM, LGG and GBMLGG. Furthermore, It displayed a positive correlation with immune cell infiltration levels in LGG. Multivariate and Univariate Cox analyses confirmed that the impact of SELENOS on the prognosis of LGG is the combined result of factors such as age and tumor grade. The expression of SELENOS was significantly negatively correlated with temozolomide IC50 in LGG. We found that SELENOS interacts with 10 proteins, which are upregulated in LGG compared to human normal tissues. The expression of these interactors is positively correlated with SELENOS expression and LGG survival/prognosis. In vitro experiments confirmed the aberrant expression of SELENOS in LGG cell lines, and siRNA-mediated knockdown of SELENOS reduced the proliferation, viability, invasion and migration of LGG cells, and induced apoptosis. CONCLUSIONS SELENOS is a potential prognostic marker and therapeutic target for LGG, and its low expression is associated with favorable prognosis in LGG.
Collapse
Affiliation(s)
- Yuetong Wang
- Shaanxi Academy of Traditional Chinese Medicine (Shaanxi Provincial Hospital of Chinese Medicine), Xi'an 710003, PR China
| | - Kai Qu
- Shaanxi Academy of Traditional Chinese Medicine (Shaanxi Provincial Hospital of Chinese Medicine), Xi'an 710003, PR China
| | - Zengrun Xia
- Ankang R&D Center of Se-enriched Products, Ankang 725000, PR China
| | - Meng Qi
- Ankang R&D Center of Se-enriched Products, Ankang 725000, PR China
| | - Xiaoping Du
- Ankang R&D Center of Se-enriched Products, Ankang 725000, PR China
| | - Zunhua Ke
- Affiliated Hospital of Shaanxi University of Chinese Medicine, Xianyang 712046, PR China.
| | - Rongqiang Zhang
- School of Public Health, Shaanxi University of Chinese Medicine, Xianyang 712046, PR China.
| |
Collapse
|
2
|
Pisano L, Turco M, Supuran CT. Biomedical applications of tyrosinases and tyrosinase inhibitors. Enzymes 2024; 56:261-280. [PMID: 39304289 DOI: 10.1016/bs.enz.2024.05.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/22/2024]
Abstract
Tyrosinase is involved in several human diseases, among which hypopigmentation and depigmentation conditions (vitiligo, idiopathic guttate hypomelanosis, pityriasis versicolor, pityriasis alba) and hyperpigmentations (melasma, lentigines, post-inflammatory and periorbital hyperpigmentation, cervical idiopathic poikiloderma and acanthosis nigricans). There are increasing evidences that tyrosinase plays a relevant role in the formation and progression of melanoma, a difficult to treat skin tumor. Hydroquinone, azelaic acid and tretinoin (all-trans-retinoic acid) are clinically used in the management of some hyperpigmentations, whereas many novel chemotypes acting as tyrosinase inhibitors with potential antimelanoma action are being investigated. Kojic acid, hydroquinone, its glycosylated derivative arbutin, or the resorcinol derivative rucinol are used in cosmesis in creams as skin whitening agents, whereas no antimelanoma tyrosinase inhibitor reached clinical trials so far, although thiamidol is a recently approved new tyrosinase inhibitor for the treatment of melasma. Kojic acid and vitamin C are used for avoiding vegetable/food oxidative browning due to the tyrosinase-catalyzed reactions, whereas bacterial enzymes show potential in biotechnological applications, for the production of mixed melanins, for protein cross-linking reactions, for producing phenol(s) biosensors, of for the production of L-DOPA, an anti-Parkinson's disease drug.
Collapse
Affiliation(s)
- Luigi Pisano
- Section of Dermatology, Health Sciences Department, University of Florence, Florence, Italy
| | - Martina Turco
- Health Sciences Department (DSS), University of Florence, Florence, Italy
| | - Claudiu T Supuran
- Neurofarba Department, Pharmaceutical and Nutraceutical Section, University of Florence, Sesto Fiorentino, Florence, Italy.
| |
Collapse
|
3
|
Wang Y, Fu G, Chen X, Xia Z, Qi M, Du X, Liu K, Liu Q, Sun N, Shi C, Qu K, Zhang R. Selenoprotein GPX3 is a novel prognostic indicator for stomach adenocarcinoma and brain low-grade gliomas: Evidence from an integrative pan-cancer analysis. Heliyon 2024; 10:e32271. [PMID: 38873671 PMCID: PMC11170152 DOI: 10.1016/j.heliyon.2024.e32271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 05/28/2024] [Accepted: 05/30/2024] [Indexed: 06/15/2024] Open
Abstract
Background The antioxidant enzyme GPX3 is a selenoprotein that transports selenium in blood and maintains its levels in peripheral tissues. Aberrant GPX3 expression is strongly linked to the development of some tumors. However, there is a scarcity of studies examining the pan-cancer expression patterns and prognostic relevance of GPX3. Methods GPX3 expression levels in normal tissues and multiple tumors were analyzed using TCGA, CCLE, GTEx, UALCAN and HPA databases. Forest plots and KM survival curves were utilized to evaluate the correlation between GPX3 expression and the outcome of tumor patients. The prognostic value of GPX3 in LGG was assessed utilizing the CGGA datasets, and that in STAD was tested by TCGA and GEO databases. A nomogram was then constructed to predict OS in STAD using R software. Additionally, the impact of GPX3 on post-chemoradiotherapy OS in patients with LGG and STAD was evaluated using the KM method. The multiplicative interaction of GPX3 expression, chemotherapy and radiotherapy on STAD and LGG was analyzed using logistic regression models. The correlation of GPX3 with the immune infiltration, immune neoantigens and MMR genes were investigated in TCGA cohort. Results GPX3 exhibited downregulation across 21 tumor types, including STAD, with its decreased expression significantly associated with improved OS, DFS, PFS and DSS. Conversely, in LGG, low levels of GPX3 expression were indicative of a poorer prognosis. Univariate and multivariate Cox models further identified GPX3 as an independent predictor of STAD, and a nomogram based on GPX3 expression and other independent factors showed high level of predictive accuracy. Moreover, low GPX3 expression and chemotherapy prolonged the survival of STAD. In LGG patients, chemoradiotherapy, GPX3 and chemotherapy, and GPX3 and chemoradiotherapy may improve prognosis. Our observations reveal a notable connection between GPX3 and immune infiltration, immune neoantigens, and MMR genes. Conclusions The variations in GPX3 expression are linked to the controlling tumor development and could act as a promising biomarker that impacts the prognosis of specific cancers like STAD and LGG.
Collapse
Affiliation(s)
- Yuetong Wang
- Shaanxi Academy of Traditional Chinese Medicine (Shaanxi Provincial Hospital of Chinese Medicine), Xi'an, 710003, PR China
| | - Guotao Fu
- School of Public Health, Shaanxi University of Chinese Medicine, Xianyang, 712046, PR China
| | - Xueqin Chen
- School of Public Health, Shaanxi University of Chinese Medicine, Xianyang, 712046, PR China
| | - Zengrun Xia
- Ankang R&D Center of Se-enriched Products, Ankang, 725000, PR China
| | - Meng Qi
- Ankang R&D Center of Se-enriched Products, Ankang, 725000, PR China
| | - Xiaoping Du
- Ankang R&D Center of Se-enriched Products, Ankang, 725000, PR China
| | - Kun Liu
- School of Public Health and Management, Binzhou Medical University, Yantai, 246003, PR China
| | - Qiling Liu
- School of Public Health, Shaanxi University of Chinese Medicine, Xianyang, 712046, PR China
| | - Na Sun
- School of Public Health, Shaanxi University of Chinese Medicine, Xianyang, 712046, PR China
| | - Chuandao Shi
- School of Public Health, Shaanxi University of Chinese Medicine, Xianyang, 712046, PR China
| | - Kai Qu
- Shaanxi Academy of Traditional Chinese Medicine (Shaanxi Provincial Hospital of Chinese Medicine), Xi'an, 710003, PR China
| | - Rongqiang Zhang
- School of Public Health, Shaanxi University of Chinese Medicine, Xianyang, 712046, PR China
| |
Collapse
|
4
|
Sun X, Watanabe T, Oda Y, Shen W, Ahmad A, Ouda R, de Figueiredo P, Kitamura H, Tanaka S, Kobayashi KS. Targeted demethylation and activation of NLRC5 augment cancer immunogenicity through MHC class I. Proc Natl Acad Sci U S A 2024; 121:e2310821121. [PMID: 38300873 PMCID: PMC10861931 DOI: 10.1073/pnas.2310821121] [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: 06/29/2023] [Accepted: 12/30/2023] [Indexed: 02/03/2024] Open
Abstract
Impaired expression of MHC (major histocompatibility complex) class I in cancers constitutes a major mechanism of immune evasion. It has been well documented that the low level of MHC class I is associated with poor prognosis and resistance to checkpoint blockade therapies. However, there is lmited approaches to specifically induce MHC class I to date. Here, we show an approach for robust and specific induction of MHC class I by targeting an MHC class I transactivator (CITA)/NLRC5, using a CRISPR/Cas9-based gene-specific system, designated TRED-I (Targeted reactivation and demethylation for MHC-I). The TRED-I system specifically recruits a demethylating enzyme and transcriptional activators on the NLRC5 promoter, driving increased MHC class I antigen presentation and accelerated CD8+ T cell activation. Introduction of the TRED-I system in an animal cancer model exhibited tumor-suppressive effects accompanied with increased infiltration and activation of CD8+ T cells. Moreover, this approach boosted the efficacy of checkpoint blockade therapy using anti-PD1 (programmed cell death protein) antibody. Therefore, targeting NLRC5 by this strategy provides an attractive therapeutic approach for cancer.
Collapse
Affiliation(s)
- Xin Sun
- Department of Immunology, Graduate School of Medicine, Hokkaido University, Sapporo060-8638, Japan
| | - Toshiyuki Watanabe
- Department of Immunology, Graduate School of Medicine, Hokkaido University, Sapporo060-8638, Japan
| | - Yoshitaka Oda
- Department of Cancer Pathology, Graduate School of Medicine, Hokkaido University, Hokkaido, Sapporo060-8638, Japan
| | - Weidong Shen
- Division of Functional Immunology, Section of Disease Control, Institute for Genetic Medicine, Hokkaido University, Sapporo060-8638, Japan
| | - Alaa Ahmad
- Department of Immunology, Graduate School of Medicine, Hokkaido University, Sapporo060-8638, Japan
| | - Ryota Ouda
- Department of Immunology, Graduate School of Medicine, Hokkaido University, Sapporo060-8638, Japan
| | - Paul de Figueiredo
- Department of Microbial Pathogenesis and Immunology, Texas A&M Health Science Center, Bryan, TX77807
- Department of Molecular Microbiology and Immunology, University of Missouri School of Medicine, Columbia, MO65211
- Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO65211
- Department of Veterinary Pathobiology, University of MissouriSchool of Veterinary Medicine, Columbia, MO65211
| | - Hidemitsu Kitamura
- Division of Functional Immunology, Section of Disease Control, Institute for Genetic Medicine, Hokkaido University, Sapporo060-8638, Japan
- Department of Biomedical Engineering, Faculty of Science and Engineering, Toyo University, Kawagoe350-8585, Japan
| | - Shinya Tanaka
- Department of Cancer Pathology, Graduate School of Medicine, Hokkaido University, Hokkaido, Sapporo060-8638, Japan
- Institute for Chemical Reaction Design and Discovery, Hokkaido University, Sapporo001-0021, Japan
| | - Koichi S. Kobayashi
- Department of Immunology, Graduate School of Medicine, Hokkaido University, Sapporo060-8638, Japan
- Department of Microbial Pathogenesis and Immunology, Texas A&M Health Science Center, Bryan, TX77807
- Institute for Vaccine Research and Development, Hokkaido University, Sapporo060-8638, Japan
| |
Collapse
|
5
|
Douguet L, Fert I, Lopez J, Vesin B, Le Chevalier F, Moncoq F, Authié P, Nguyen T, Noirat A, Névo F, Blanc C, Bourgine M, Hardy D, Anna F, Majlessi L, Charneau P. Full eradication of pre-clinical human papilloma virus-induced tumors by a lentiviral vaccine. EMBO Mol Med 2023; 15:e17723. [PMID: 37675835 PMCID: PMC10565635 DOI: 10.15252/emmm.202317723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 08/22/2023] [Accepted: 08/23/2023] [Indexed: 09/08/2023] Open
Abstract
Human papillomavirus (HPV) infections are the cause of all cervical and numerous oropharyngeal and anogenital cancers. The currently available HPV vaccines, which induce neutralizing antibodies, have no therapeutic effect on established tumors. Here, we developed an immuno-oncotherapy against HPV-induced tumors based on a non-integrative lentiviral vector encoding detoxified forms of the Early E6 and E7 oncoproteins of HPV16 and 18 genotypes, namely, "Lenti-HPV-07". A single intramuscular injection of Lenti-HPV-07 into mice bearing established HPV-induced tumors resulted in complete tumor eradication in 100% of the animals and was also effective against lung metastases. This effect correlated with CD8+ T-cell induction and profound remodeling of the tumor microenvironment. In the intra-tumoral infiltrates of vaccinated mice, the presence of large amounts of activated effector, resident memory, and transcription factor T cell factor-1 (TCF-1)+ "stem-like" CD8+ T cells was associated with full tumor eradication. The Lenti-HPV-07-induced immunity was long-lasting and prevented tumor growth after a late re-challenge, mimicking tumor relapse. Lenti-HPV-07 therapy synergizes with an anti-checkpoint inhibitory treatment and therefore shows promise as an immuno-oncotherapy against established HPV-mediated malignancies.
Collapse
Affiliation(s)
- Laëtitia Douguet
- Virology Department, Pasteur‐TheraVectys Joint Lab, Institut PasteurUniversité de ParisParisFrance
| | - Ingrid Fert
- Virology Department, Pasteur‐TheraVectys Joint Lab, Institut PasteurUniversité de ParisParisFrance
| | - Jodie Lopez
- Virology Department, Pasteur‐TheraVectys Joint Lab, Institut PasteurUniversité de ParisParisFrance
| | - Benjamin Vesin
- Virology Department, Pasteur‐TheraVectys Joint Lab, Institut PasteurUniversité de ParisParisFrance
| | - Fabien Le Chevalier
- Virology Department, Pasteur‐TheraVectys Joint Lab, Institut PasteurUniversité de ParisParisFrance
| | - Fanny Moncoq
- Virology Department, Pasteur‐TheraVectys Joint Lab, Institut PasteurUniversité de ParisParisFrance
| | - Pierre Authié
- Virology Department, Pasteur‐TheraVectys Joint Lab, Institut PasteurUniversité de ParisParisFrance
| | - Trang‐My Nguyen
- Virology Department, Pasteur‐TheraVectys Joint Lab, Institut PasteurUniversité de ParisParisFrance
| | - Amandine Noirat
- Virology Department, Pasteur‐TheraVectys Joint Lab, Institut PasteurUniversité de ParisParisFrance
| | - Fabien Névo
- Virology Department, Pasteur‐TheraVectys Joint Lab, Institut PasteurUniversité de ParisParisFrance
| | - Catherine Blanc
- Virology Department, Pasteur‐TheraVectys Joint Lab, Institut PasteurUniversité de ParisParisFrance
| | - Maryline Bourgine
- Virology Department, Pasteur‐TheraVectys Joint Lab, Institut PasteurUniversité de ParisParisFrance
| | - David Hardy
- Histopathology Platform, Institut PasteurUniversité de ParisParisFrance
| | - François Anna
- Virology Department, Pasteur‐TheraVectys Joint Lab, Institut PasteurUniversité de ParisParisFrance
| | - Laleh Majlessi
- Virology Department, Pasteur‐TheraVectys Joint Lab, Institut PasteurUniversité de ParisParisFrance
| | - Pierre Charneau
- Virology Department, Pasteur‐TheraVectys Joint Lab, Institut PasteurUniversité de ParisParisFrance
| |
Collapse
|
6
|
Yadav R, Hakobyan N, Wang JC. Role of Next Generation Immune Checkpoint Inhibitor (ICI) Therapy in Philadelphia Negative Classic Myeloproliferative Neoplasm (MPN): Review of the Literature. Int J Mol Sci 2023; 24:12502. [PMID: 37569880 PMCID: PMC10420159 DOI: 10.3390/ijms241512502] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 07/17/2023] [Accepted: 07/26/2023] [Indexed: 08/13/2023] Open
Abstract
The Philadelphia chromosome-negative (Ph-) myeloproliferative neoplasms (MPNs), which include essential thrombocythemia (ET), polycythemia vera (PV), and myelofibrosis (MF), are enduring and well-known conditions. These disorders are characterized by the abnormal growth of one or more hematopoietic cell lineages in the body's stem cells, leading to the enlargement of organs and the manifestation of constitutional symptoms. Numerous studies have provided evidence indicating that the pathogenesis of these diseases involves the dysregulation of the immune system and the presence of chronic inflammation, both of which are significant factors. Lately, the treatment of cancer including hematological malignancy has progressed on the agents aiming for the immune system, cytokine environment, immunotherapy agents, and targeted immune therapy. Immune checkpoints are the molecules that regulate T cell function in the tumor microenvironment (TME). The first line of primary immune checkpoints are programmed cell death-1 (PD-1)/programmed cell death ligand-1 (PD-L1), and cytotoxic T-lymphocyte antigen-4 (CTLA-4). Immune checkpoint inhibitor therapy (ICIT) exerts its anti-tumor actions by blocking the inhibitory pathways in T cells and has reformed cancer treatment. Despite the impressive clinical success of ICIT, tumor internal resistance poses a challenge for oncologists leading to a low response rate in solid tumors and hematological malignancies. A Phase II trial on nivolumab for patients with post-essential thrombocythemia myelofibrosis, primary myelofibrosis, or post-polycythemia myelofibrosis was performed (Identifier: NCT02421354). This trial tested the efficacy of a PD-1 blockade agent, namely nivolumab, but was terminated prematurely due to adverse events and lack of efficacy. A multicenter, Phase II, single-arm open-label study was conducted including pembrolizumab in patients with primary thrombocythemia, post-essential thrombocythemia or post-polycythemia vera myelofibrosis that were ineligible for or were previously treated with ruxolitinib. This study showed that pembrolizumab treatment did not have many adverse events, but there were no pertinent clinical responses hence it was terminated after the first stage was completed. To avail the benefits from immunotherapy, the paradigm has shifted to new immune checkpoints in the TME such as lymphocyte activation gene-3 (LAG-3), T cell immunoglobulin and mucin domain 3 (TIM-3), T cell immunoglobulin and ITIM domain (TIGIT), V-domain immunoglobulin-containing suppressor of T cell activation (VISTA), and human endogenous retrovirus-H long terminal repeat-associating protein 2 (HHLA2) forming the basis of next-generation ICIT. The primary aim of this article is to underscore and elucidate the significance of next-generation ICIT in the context of MPN. Specifically, we aim to explore the potential of monoclonal antibodies as targeted immunotherapy and the development of vaccines targeting specific MPN epitopes, with the intent of augmenting tumor-related immune responses. It is anticipated that these therapeutic modalities rooted in immunotherapy will not only expand but also enhance the existing treatment regimens for patients afflicted with MPN. Preliminary studies from our laboratory showed over-expressed MDSC and over-expressed VISTA in MDSC, and in progenitor and immune cells directing the need for more clinical trials using next-generation ICI in the treatment of MPN.
Collapse
Affiliation(s)
- Ruchi Yadav
- Department of Internal Medicine, Brookdale University Hospital Medical Center, Brooklyn, NY 11212, USA; (R.Y.); (N.H.)
| | - Narek Hakobyan
- Department of Internal Medicine, Brookdale University Hospital Medical Center, Brooklyn, NY 11212, USA; (R.Y.); (N.H.)
| | - Jen-Chin Wang
- Department of Hematology/Oncology, Brookdale University Hospital Medical Center, Brooklyn, NY 11212, USA
| |
Collapse
|
7
|
Veerman RE, Akpinar GG, Offens A, Steiner L, Larssen P, Lundqvist A, Karlsson MCI, Gabrielsson S. Antigen-Loaded Extracellular Vesicles Induce Responsiveness to Anti-PD-1 and Anti-PD-L1 Treatment in a Checkpoint Refractory Melanoma Model. Cancer Immunol Res 2023; 11:217-227. [PMID: 36546872 PMCID: PMC9896027 DOI: 10.1158/2326-6066.cir-22-0540] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 09/23/2022] [Accepted: 11/30/2022] [Indexed: 12/24/2022]
Abstract
Extracellular vesicles (EV) are important mediators of intercellular communication and are potential candidates for cancer immunotherapy. Immune checkpoint blockade, specifically targeting the programmed cell death protein 1 (PD-1)/programmed death-ligand 1 (PD-L1) axis, mitigates T-cell exhaustion, but is only effective in a subset of patients with cancer. Reasons for therapy resistance include low primary T-cell activation to cancer antigens, poor antigen presentation, and reduced T-cell infiltration into the tumor. Therefore, combination strategies have been extensively explored. Here, we investigated whether EV therapy could induce susceptibility to anti-PD-1 or anti-PD-L1 therapy in a checkpoint-refractory B16 melanoma model. Injection of dendritic cell-derived EVs, but not checkpoint blockade, induced a potent antigen-specific T-cell response and reduced tumor growth in tumor-bearing mice. Combination therapy of EVs and anti-PD-1 or anti-PD-L1 potentiated immune responses to ovalbumin- and α-galactosylceramide-loaded EVs in the therapeutic model. Moreover, combination therapy resulted in increased survival in a prophylactic tumor model. This demonstrates that EVs can induce potent antitumor immune responses in checkpoint refractory cancer and induce anti-PD-1 or anti-PD-L1 responses in a previously nonresponsive tumor model.
Collapse
Affiliation(s)
- Rosanne E Veerman
- Division of Immunology and Allergy, Department of Medicine, Karolinska Institutet, Solna, Stockholm, Sweden.,Department of Clinical Immunology and Transfusion Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Gözde Güclüler Akpinar
- Division of Immunology and Allergy, Department of Medicine, Karolinska Institutet, Solna, Stockholm, Sweden.,Department of Clinical Immunology and Transfusion Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Annemarijn Offens
- Division of Immunology and Allergy, Department of Medicine, Karolinska Institutet, Solna, Stockholm, Sweden.,Department of Clinical Immunology and Transfusion Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Loïc Steiner
- Division of Immunology and Allergy, Department of Medicine, Karolinska Institutet, Solna, Stockholm, Sweden.,Department of Clinical Immunology and Transfusion Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Pia Larssen
- Division of Immunology and Allergy, Department of Medicine, Karolinska Institutet, Solna, Stockholm, Sweden.,Department of Clinical Immunology and Transfusion Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Andreas Lundqvist
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Mikael C I Karlsson
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Susanne Gabrielsson
- Division of Immunology and Allergy, Department of Medicine, Karolinska Institutet, Solna, Stockholm, Sweden.,Department of Clinical Immunology and Transfusion Medicine, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
8
|
Liu S, Zhang H, Yan J, Zhu J, Bai Z, Li X. FOXP3 and SQSTM1/P62 correlate with prognosis and immune infiltration in hepatocellular carcinoma. Pathol Res Pract 2023; 242:154292. [PMID: 36630868 DOI: 10.1016/j.prp.2022.154292] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 12/11/2022] [Accepted: 12/25/2022] [Indexed: 12/28/2022]
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is one of the most common highly malignant tumours worldwide. FOXP3 and SQSTM1/P62 have been shown to be abnormally expressed in tumour cells, but their function in different tumours remains controversial. The present study was designed to evaluate the expression of FOXP3 and P62 in HCC and their prognostic value as well as their relationship with immune infiltration in HCC patients. METHODS The Gene Expression Omnibus (GEO) database and TNMplot.com platform were used to analyse the expression of FOXP3 and P62. The Cancer Genome Atlas (TCGA) database and Kaplan-Meier plotter were used to assess the impacts of FOXP3 and P62 on clinical prognosis. In addition, TCGA database was also used to examine the correlation between the expression of FOXP3 and P62 and tumour immune infiltration using the CIBERSORT algorithm. Finally, immunohistochemistry (IHC) was used to determine expression levels of FOXP3 and P62 in 89 HCC and adjacent normal liver tissues, and their effects on clinicopathological features and prognosis were verified. RESULTS FOXP3 expression was downregulated in HCC tissues, while P62 expression was upregulated. FOXP3 underexpression and P62 overexpression were closely related to decreased overall survival (OS) in HCC patients. Additionally, the abnormal expression of FOXP3 and P62 was closely related to the infiltration levels of 12 types of immune cells, including regulatory T cells (Tregs), M2 macrophages, M0 macrophages, and CD8 T cells. Notably, in the validation model, abnormal FOXP3 and P62 expression was significantly associated with adverse clinicopathological factors in HCC patients, including elevated α-fetoprotein (AFP) levels, poor tumour differentiation, and increased Ki67 levels. Furthermore, low FOXP3 and high P62 expression were independent risk factors for predicting OS prognosis in HCC patients. CONCLUSION FOXP3 and P62 have been shown to be important prognostic factors in HCC patients and are associated with immune cell infiltration in HCC. These findings suggest that FOXP3 and P62 may be valuable prognostic biomarkers and potential therapeutic targets for HCC treatment.
Collapse
Affiliation(s)
- Shuohui Liu
- Department of General Surgery, Xi'an No.3 Hospital, The Affiliated Hospital of Northwest University, Xi'an, Shaanxi 710000, People's Republic of China
| | - Honglong Zhang
- The First School of Clinical Medical, Lanzhou University, Lanzhou, Gansu 730000, People's Republic of China
| | - Jun Yan
- The First School of Clinical Medical, Lanzhou University, Lanzhou, Gansu 730000, People's Republic of China; Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, Gansu 730000, People's Republic of China; Key Laboratory of Biotherapy and Regenerative Medicine of Gansu Province, Lanzhou, Gansu 730000, People's Republic of China; Hepatopancreatobiliary Surgery Institute of Gansu Province, Medical College Cancer Center of Lanzhou University, Lanzhou, Gansu 730000, People's Republic of China
| | - Jun Zhu
- Department of Pathology, Donggang District, First Hospital of Lanzhou University, Lanzhou, Gansu 730000, People's Republic of China
| | - Zhongtian Bai
- The First School of Clinical Medical, Lanzhou University, Lanzhou, Gansu 730000, People's Republic of China; Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, Gansu 730000, People's Republic of China; Key Laboratory of Biotherapy and Regenerative Medicine of Gansu Province, Lanzhou, Gansu 730000, People's Republic of China; Hepatopancreatobiliary Surgery Institute of Gansu Province, Medical College Cancer Center of Lanzhou University, Lanzhou, Gansu 730000, People's Republic of China
| | - Xun Li
- The First School of Clinical Medical, Lanzhou University, Lanzhou, Gansu 730000, People's Republic of China; Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, Gansu 730000, People's Republic of China; Key Laboratory of Biotherapy and Regenerative Medicine of Gansu Province, Lanzhou, Gansu 730000, People's Republic of China; Hepatopancreatobiliary Surgery Institute of Gansu Province, Medical College Cancer Center of Lanzhou University, Lanzhou, Gansu 730000, People's Republic of China.
| |
Collapse
|
9
|
Chocarro L, Blanco E, Arasanz H, Fernández-Rubio L, Bocanegra A, Echaide M, Garnica M, Ramos P, Fernández-Hinojal G, Vera R, Kochan G, Escors D. Clinical landscape of LAG-3-targeted therapy. IMMUNO-ONCOLOGY TECHNOLOGY 2022; 14:100079. [PMID: 35755891 PMCID: PMC9216443 DOI: 10.1016/j.iotech.2022.100079] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Lymphocyte-activated gene 3 (LAG-3) is a cell surface inhibitory receptor and a key regulator of immune homeostasis with multiple biological activities related to T-cell functions. LAG-3 is considered a next-generation immune checkpoint of clinical importance, right next to programmed cell death protein 1 (PD-1) and cytotoxic T-cell lymphocyte antigen-4 (CTLA-4). Indeed, it is the third inhibitory receptor to be exploited in human anticancer immunotherapies. Several LAG-3-antagonistic immunotherapies are being evaluated at various stages of preclinical and clinical development. In addition, combination therapies blocking LAG-3 together with other immune checkpoints are also being evaluated at preclinical and clinical levels. Indeed, the co-blockade of LAG-3 with PD-1 is demonstrating encouraging results. A new generation of bispecific PD-1/LAG-3-blocking agents have also shown strong capacities to specifically target PD-1+ LAG-3+ highly dysfunctional T cells and enhance their proliferation and effector activities. Here we identify and classify preclinical and clinical trials conducted involving LAG-3 as a target through an extensive bibliographic research. The current understanding of LAG-3 clinical applications is summarized, and most of the publically available data up to date regarding LAG-3-targeted therapy preclinical and clinical research and development are reviewed and discussed.
Collapse
Affiliation(s)
- L. Chocarro
- Oncoimmunology Research Unit, Navarrabiomed-Fundación Miguel Servet, Universidad Pública de Navarra (UPNA), Hospital Universitario de Navarra (HUN), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - E. Blanco
- Oncoimmunology Research Unit, Navarrabiomed-Fundación Miguel Servet, Universidad Pública de Navarra (UPNA), Hospital Universitario de Navarra (HUN), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
- Division of Gene Therapy and Regulation of Gene Expression, Cima Universidad de Navarra, Instituto de Investigación Sanitaria de Navarra (IdISNA), Pamplona, Spain
| | - H. Arasanz
- Oncoimmunology Research Unit, Navarrabiomed-Fundación Miguel Servet, Universidad Pública de Navarra (UPNA), Hospital Universitario de Navarra (HUN), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
- Medical Oncology Unit, Hospital Universitario de Navarra (HUN), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - L. Fernández-Rubio
- Oncoimmunology Research Unit, Navarrabiomed-Fundación Miguel Servet, Universidad Pública de Navarra (UPNA), Hospital Universitario de Navarra (HUN), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - A. Bocanegra
- Oncoimmunology Research Unit, Navarrabiomed-Fundación Miguel Servet, Universidad Pública de Navarra (UPNA), Hospital Universitario de Navarra (HUN), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - M. Echaide
- Oncoimmunology Research Unit, Navarrabiomed-Fundación Miguel Servet, Universidad Pública de Navarra (UPNA), Hospital Universitario de Navarra (HUN), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - M. Garnica
- Oncoimmunology Research Unit, Navarrabiomed-Fundación Miguel Servet, Universidad Pública de Navarra (UPNA), Hospital Universitario de Navarra (HUN), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - P. Ramos
- Oncoimmunology Research Unit, Navarrabiomed-Fundación Miguel Servet, Universidad Pública de Navarra (UPNA), Hospital Universitario de Navarra (HUN), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - G. Fernández-Hinojal
- Oncoimmunology Research Unit, Navarrabiomed-Fundación Miguel Servet, Universidad Pública de Navarra (UPNA), Hospital Universitario de Navarra (HUN), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
- Medical Oncology Department, Hospital Clínico San Carlos, Madrid, Spain
| | - R. Vera
- Medical Oncology Unit, Hospital Universitario de Navarra (HUN), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - G. Kochan
- Oncoimmunology Research Unit, Navarrabiomed-Fundación Miguel Servet, Universidad Pública de Navarra (UPNA), Hospital Universitario de Navarra (HUN), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - D. Escors
- Oncoimmunology Research Unit, Navarrabiomed-Fundación Miguel Servet, Universidad Pública de Navarra (UPNA), Hospital Universitario de Navarra (HUN), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| |
Collapse
|
10
|
Somaiah N, Chawla SP, Block MS, Morris JC, Do K, Kim JW, Druta M, Sankhala KK, Hwu P, Jones RL, Gnjatic S, Kim-Schulze S, Tuballes K, Yishak M, Lu H, Yakovich A, Ter Meulen J, Chen M, Kenney RT, Bohac C, Pollack SM. A Phase 1b Study Evaluating the Safety, Tolerability, and Immunogenicity of CMB305, a Lentiviral-Based Prime-Boost Vaccine Regimen, in Patients with Locally Advanced, Relapsed, or Metastatic Cancer Expressing NY-ESO-1. Oncoimmunology 2020; 9:1847846. [PMID: 33312760 PMCID: PMC7714520 DOI: 10.1080/2162402x.2020.1847846] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 11/03/2020] [Indexed: 02/07/2023] Open
Abstract
Preclinical data suggest that a "prime-boost" vaccine regimen using a target-expressing lentiviral vector for priming, followed by a recombinant protein boost, may be effective against cancer; however, this strategy has not been evaluated in a clinical setting. CMB305 is a prime-boost vaccine designed to induce a broad anti-NY-ESO-1 immune response. It is composed of LV305, which is an NY-ESO-1 expressing lentiviral vector, and G305, a recombinant adjuvanted NY-ESO-1 protein. This multicenter phase 1b, first-in-human trial evaluated CMB305 in patients with NY-ESO-1 expressing solid tumors. Safety was examined in a 3 + 3 dose-escalation design, followed by an expansion with CMB305 alone or in a combination with either oral metronomic cyclophosphamide or intratumoral injections of a toll-like receptor agonist (glucopyranosyl lipid A). Of the 79 patients who enrolled, 81.0% had sarcomas, 86.1% had metastatic disease, and 57.0% had progressive disease at study entry. The most common adverse events were fatigue (34.2%), nausea (26.6%), and injection-site pain (24.1%). In patients with soft tissue sarcomas, a disease control rate of 61.9% and an overall survival of 26.2 months (95% CI, 22.1-NA) were observed. CMB305 induced anti-NY-ESO-1 antibody and T-cell responses in 62.9% and 47.4% of patients, respectively. This is the first trial to test a prime-boost vaccine regimen in patients with advanced cancer. This approach is feasible, can be delivered safely, and with evidence of immune response as well as suggestion of clinical benefit.
Collapse
Affiliation(s)
- Neeta Somaiah
- Department of Sarcoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Sant P. Chawla
- Sarcoma Oncology Center, Santa Monica, CA, United States
| | - Matthew S. Block
- Department of Medical Oncology, Mayo Clinic, Rochester, MN, United States
| | - John C. Morris
- Department of Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Khanh Do
- Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, United States
| | - Joseph W. Kim
- Department of Medical Oncology, Yale School of Medicine, New Haven, CT, United States
| | - Mihaela Druta
- Medical Oncology, Moffitt Cancer Center, Tampa, FL, United States
| | - Kamalesh K. Sankhala
- Hematology/Oncology, Cedars-Sinai Medical Center, Beverly Hills, CA, United States
| | - Patrick Hwu
- Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Robin L. Jones
- Seattle Cancer Care Alliance, Seattle, WA
- Sarcoma Unit, Royal Marsden Hospital, London, UK
- Sarcoma Clinical Trials, Institute of Cancer Research, London, UK
| | - Sacha Gnjatic
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Seunghee Kim-Schulze
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Kevin Tuballes
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Mahlet Yishak
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Hailing Lu
- Immune Design Corp., South San Francisco, CA, United States
- Biomarkers and Diagnostics, Seattle Genetics, Inc, Bothell, WA, United States
| | - Adam Yakovich
- Immune Design Corp., South San Francisco, CA, United States
- Medical Affairs, Replimune Group, Inc, Woburn, MA, United States
| | - Jan Ter Meulen
- Immune Design Corp., South San Francisco, CA, United States
| | - Michael Chen
- Immune Design Corp., South San Francisco, CA, United States
- *Sangamo Therapeutics, Inc., Brisbane, CA, United States
| | - Richard T. Kenney
- Immune Design Corp., South San Francisco, CA, United States
- Clin Reg Biologics, LLC, Potomac, MD, United States
| | - Chet Bohac
- Immune Design Corp., South San Francisco, CA, United States
- Macrogenics, Inc, Rockville, MD, United States
| | - Seth M. Pollack
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States
| |
Collapse
|
11
|
Ju Q, Li XM, Zhang H, Zhao YJ. BRCA1-Associated Protein Is a Potential Prognostic Biomarker and Is Correlated With Immune Infiltration in Liver Hepatocellular Carcinoma: A Pan-Cancer Analysis. Front Mol Biosci 2020; 7:573619. [PMID: 33240929 PMCID: PMC7667264 DOI: 10.3389/fmolb.2020.573619] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 09/09/2020] [Indexed: 11/30/2022] Open
Abstract
Background BRCA1-associated protein (BRAP) is a critical gene that regulates inflammation-related signaling pathway and affects patients’ prognosis in esophageal squamous cell carcinoma (ESCC). However, its roles in different cancers remain largely unknown. Methods BRAP expression in human pan-cancer was analyzed via the Genotype-Tissue Expression (GTEx) and The Cancer Genome Atlas (TCGA) database. Pearson correlation analysis was used to evaluate the association between BRAP expression with mismatch repair (MMR) gene mutation and DNA methyltransferase. We evaluated the influence of BRAP on clinical prognosis by univariate survival analysis. Moreover, the correlation between BRAP and tumor immune infiltration was analyzed via the Tumor Immune Evaluation Resource (TIMER) database. Pearson correlation analysis was used to investigate the correlation between BRAP expression and immune checkpoint genes expression. Results BRAP is abnormally overexpressed and significantly correlated with MMR gene mutation level and DNA methyltransferase expression in human pan-cancer. Univariate survival analysis showed that BRAP was significant with patients’ overall survival (OS) in six cancer types, disease-free interval (DFI) in three cancer types, and progression-free interval (PFI) in two cancer types. Remarkably, increased BRAP expression was strongly correlated with patients’ poor prognosis in liver hepatocellular carcinoma (LIHC), whether OS (P < 0.0001, hazard ratio (HR) = 1.1), DFI (P = 0.00099, HR = 1.06), or PFI (P = 0.00025, HR = 1.07). Moreover, a positive relationship was found between BRAP expression and immune infiltrating cells including B cell, CD4 + T cell, CD8 + T cell, dendritic cell, macrophage cell, and neutrophil cell in colon adenocarcinoma (COAD), kidney renal clear cell carcinoma (KIRC), and LIHC. Additionally, BRAP expression showed strong correlations with immune checkpoint genes in LIHC. Conclusion BRAP expression is increased in human pan-cancer samples compared with normal tissues. Overexpression of BRAP is correlated with poor prognosis and immune infiltration in multiple cancers, especially in LIHC. These findings suggest that BRAP may be used as a potential molecular biomarker for determining prognosis and immune infiltration in LIHC.
Collapse
Affiliation(s)
- Qiang Ju
- Department of Blood Transfusion, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Xin-Mei Li
- School of Public Health, Qingdao University, Qingdao, China
| | - Heng Zhang
- School of Public Health, Qingdao University, Qingdao, China
| | - Yan-Jie Zhao
- School of Public Health, Qingdao University, Qingdao, China
| |
Collapse
|
12
|
Cheng R, Fontana F, Xiao J, Liu Z, Figueiredo P, Shahbazi MA, Wang S, Jin J, Torrieri G, Hirvonen JT, Zhang H, Chen T, Cui W, Lu Y, Santos HA. Recombination Monophosphoryl Lipid A-Derived Vacosome for the Development of Preventive Cancer Vaccines. ACS APPLIED MATERIALS & INTERFACES 2020; 12:44554-44562. [PMID: 32960566 PMCID: PMC7549091 DOI: 10.1021/acsami.0c15057] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Accepted: 09/09/2020] [Indexed: 05/09/2023]
Abstract
Recently, there has been an increasing interest for utilizing the host immune system to fight against cancer. Moreover, cancer vaccines, which can stimulate the host immune system to respond to cancer in the long term, are being investigated as a promising approach to induce tumor-specific immunity. In this work, we prepared an effective cancer vaccine (denoted as "vacosome") by reconstructing the cancer cell membrane, monophosphoryl lipid A as a toll-like receptor 4 agonist, and egg phosphatidylcholine. The vacosome triggered and enhanced bone marrow dendritic cell maturation as well as stimulated the antitumor response against breast cancer 4T1 cells in vitro. Furthermore, an immune memory was established in BALB/c mice after three-time preimmunization with the vacosome. After that, the immunized mice showed inhibited tumor growth and prolonged survival period (longer than 50 days). Overall, our results demonstrate that the vacosome can be a potential candidate for clinical translation as a cancer vaccine.
Collapse
Affiliation(s)
- Ruoyu Cheng
- Drug Research Program,
Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI-00014 Helsinki, Finland
| | - Flavia Fontana
- Drug Research Program,
Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI-00014 Helsinki, Finland
| | - Junyuan Xiao
- Shanghai Key Laboratory for Prevention and Treatment
of Bone and Joint Diseases, Shanghai Institute of Traumatology and
Orthopaedics, Ruijin Hospital, Shanghai
Jiao Tong University School of Medicine, 197 Ruijin Second Road, 200025 Shanghai, PR China
| | - Zehua Liu
- Drug Research Program,
Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI-00014 Helsinki, Finland
| | - Patrícia Figueiredo
- Drug Research Program,
Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI-00014 Helsinki, Finland
| | - Mohammad-Ali Shahbazi
- Drug Research Program,
Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI-00014 Helsinki, Finland
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Zanjan University of Medical Sciences, 45139-56184 Zanjan, Iran
| | - Shiqi Wang
- Drug Research Program,
Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI-00014 Helsinki, Finland
| | - Jing Jin
- Shanghai Key Laboratory for Prevention and Treatment
of Bone and Joint Diseases, Shanghai Institute of Traumatology and
Orthopaedics, Ruijin Hospital, Shanghai
Jiao Tong University School of Medicine, 197 Ruijin Second Road, 200025 Shanghai, PR China
| | - Giulia Torrieri
- Drug Research Program,
Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI-00014 Helsinki, Finland
| | - Jouni T. Hirvonen
- Drug Research Program,
Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI-00014 Helsinki, Finland
| | - Hongbo Zhang
- Shanghai Key Laboratory for Prevention and Treatment
of Bone and Joint Diseases, Shanghai Institute of Traumatology and
Orthopaedics, Ruijin Hospital, Shanghai
Jiao Tong University School of Medicine, 197 Ruijin Second Road, 200025 Shanghai, PR China
- Department of Pharmaceutical Sciences Laboratory and
Turku Center for Biotechnology, Åbo
Akademi University, FI-20520 Turku, Finland
| | - Tongtong Chen
- Radiology Department, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, 200025 Shanghai, PR China
| | - Wenguo Cui
- Shanghai Key Laboratory for Prevention and Treatment
of Bone and Joint Diseases, Shanghai Institute of Traumatology and
Orthopaedics, Ruijin Hospital, Shanghai
Jiao Tong University School of Medicine, 197 Ruijin Second Road, 200025 Shanghai, PR China
| | - Yong Lu
- Radiology Department, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, 200025 Shanghai, PR China
| | - Hélder A. Santos
- Drug Research Program,
Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI-00014 Helsinki, Finland
- Helsinki Insititute of Life Science, HiLIFE, University of Helsinki, FI-00014 Helsinki, Finland
| |
Collapse
|
13
|
Albershardt TC, Leleux J, Parsons AJ, Krull JE, Berglund P, Ter Meulen J. Intratumoral immune activation with TLR4 agonist synergizes with effector T cells to eradicate established murine tumors. NPJ Vaccines 2020; 5:50. [PMID: 32579133 PMCID: PMC7298055 DOI: 10.1038/s41541-020-0201-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 05/07/2020] [Indexed: 12/19/2022] Open
Abstract
Effective T cell-based immunotherapy of solid malignancies requires intratumoral activity of cytotoxic T cells and induction of protective immune memory. A major obstacle to intratumoral trafficking and activation of vaccine-primed or adoptively transferred tumor-specific T cells is the immunosuppressive tumor microenvironment (TME), which currently limits the efficacy of both anti-tumor vaccines and adoptive cell therapy (ACT). Combination treatments to overcome TME-mediated immunosuppression are therefore urgently needed. We combined intratumoral administration of the synthetic toll-like receptor 4 agonist glucopyranosyl lipid A (oil-in-water formulation, G100) with either active vaccination or adoptive transfer of tumor-specific CD8 T cells to mice bearing established melanomas or orthotopically inoculated glioblastomas. In combination with cancer vaccines or ACT, G100 significantly increased expression of innate immune genes, infiltration and expansion of activated effector T cells, antigen spreading, and durable immune responses. Complete tumor regression of both injected and non-injected tumors was observed only in mice receiving combination immunotherapy. TLR4-based intratumoral immune activation may be a viable approach to enhance the efficacy of therapeutic cancer vaccines and ACT in patients.
Collapse
Affiliation(s)
- Tina C Albershardt
- Immune Design, a wholly-owned subsidiary of Merck & Co., Inc, Kenilworth, NJ USA
| | - Jardin Leleux
- Immune Design, a wholly-owned subsidiary of Merck & Co., Inc, Kenilworth, NJ USA
| | - Andrea J Parsons
- Immune Design, a wholly-owned subsidiary of Merck & Co., Inc, Kenilworth, NJ USA
| | - Jordan E Krull
- Immune Design, a wholly-owned subsidiary of Merck & Co., Inc, Kenilworth, NJ USA
| | - Peter Berglund
- Immune Design, a wholly-owned subsidiary of Merck & Co., Inc, Kenilworth, NJ USA
| | - Jan Ter Meulen
- Immune Design, a wholly-owned subsidiary of Merck & Co., Inc, Kenilworth, NJ USA
| |
Collapse
|