1
|
Zheng J, Zheng Z, Zhang T, Chen X, Pang Q, Wang P, Yan C, Zhang W. Optimization of radiation target volume for locally advanced esophageal cancer in the immunotherapy era. Expert Opin Biol Ther 2024:1-12. [PMID: 39460561 DOI: 10.1080/14712598.2024.2423009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Revised: 10/19/2024] [Accepted: 10/25/2024] [Indexed: 10/28/2024]
Abstract
INTRODUCTION Locally advanced esophageal cancer (EC) has poor prognosis. Preliminary clinical studies have demonstrated the synergistic efficacy of radiotherapy combined with immunotherapy in EC. Adjusting the radiotherapy target volume to protect immune function favors immunotherapy. However, there is no clear consensus on the exact definition of the EC target volume. AREAS COVERED Preclinical studies have provided a wealth of information on immunotherapy combined with different radiotherapy modalities, and several clinical studies have evaluated the impact of immunotherapy combined with radiotherapy on locally advanced EC. Here, we illustrate the rational target volume delineation for radiotherapy in terms of patient prognosis, pattern of radiotherapy failure, treatment-related toxicities, tumor-draining lymph nodes, and systemic immunity and summarize the clinical trials of radiotherapy combined with immunotherapy in EC. EXPERT OPINION We recommend applying involved-field irradiation (IFI) instead of elective nodal irradiation (ENI) for irradiated fields when immunotherapy is combined with chemoradiotherapy (CRT) for locally advanced EC. We expect that this target design will be evaluated in clinical trials to further explore more precise diagnostic modalities, long-term toxic responses, and quality of survival, and stratification factors for personalized treatment, and to provide more treatment benefits for patients.
Collapse
Affiliation(s)
- Jian Zheng
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Zhunhao Zheng
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Tian Zhang
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Xi Chen
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Qingsong Pang
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Ping Wang
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Cihui Yan
- Department of Immunology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Immunology and Biotherapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Wencheng Zhang
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| |
Collapse
|
2
|
Singh A, Miranda Bedate A, von Richthofen HJ, Vijver SV, van der Vlist M, Kuhn R, Yermanos A, Kuball JJ, Kesmir C, Pascoal Ramos MI, Meyaard L. A novel bioinformatics pipeline for the identification of immune inhibitory receptors as potential therapeutic targets. eLife 2024; 13:RP92870. [PMID: 39377459 PMCID: PMC11460946 DOI: 10.7554/elife.92870] [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] [Indexed: 10/09/2024] Open
Abstract
Despite major successes with inhibitory receptor blockade in cancer, the identification of novel inhibitory receptors as putative drug targets is needed due to lack of durable responses, therapy resistance, and side effects. Most inhibitory receptors signal via immunoreceptor tyrosine-based inhibitory motifs (ITIMs) and previous studies estimated that our genome contains over 1600 ITIM-bearing transmembrane proteins. However, testing and development of these candidates requires increased understanding of their expression patterns and likelihood to function as inhibitory receptor. Therefore, we designed a novel bioinformatics pipeline integrating machine learning-guided structural predictions and sequence-based likelihood models to identify putative inhibitory receptors. Using transcriptomics data of immune cells, we determined the expression of these novel inhibitory receptors, and classified them into previously proposed functional categories. Known and putative inhibitory receptors were expressed across different immune cell subsets with cell type-specific expression patterns. Furthermore, putative immune inhibitory receptors were differentially expressed in subsets of tumour infiltrating T cells. In conclusion, we present an inhibitory receptor pipeline that identifies 51 known and 390 novel human inhibitory receptors. This pipeline will support future drug target selection across diseases where therapeutic targeting of immune inhibitory receptors is warranted.
Collapse
Affiliation(s)
- Akashdip Singh
- Center for Translational Immunology, University Medical Centre Utrecht, Utrecht UniversityUtrechtNetherlands
- Oncode InstituteUtrechtNetherlands
| | - Alberto Miranda Bedate
- Center for Translational Immunology, University Medical Centre Utrecht, Utrecht UniversityUtrechtNetherlands
- Department of Hematology, University Medical Center Utrecht, Utrecht UniversityUrechtNetherlands
| | - Helen J von Richthofen
- Center for Translational Immunology, University Medical Centre Utrecht, Utrecht UniversityUtrechtNetherlands
- Oncode InstituteUtrechtNetherlands
| | - Saskia V Vijver
- Center for Translational Immunology, University Medical Centre Utrecht, Utrecht UniversityUtrechtNetherlands
- Oncode InstituteUtrechtNetherlands
| | - Michiel van der Vlist
- Center for Translational Immunology, University Medical Centre Utrecht, Utrecht UniversityUtrechtNetherlands
- Oncode InstituteUtrechtNetherlands
| | - Raphael Kuhn
- Department of Biosystems Science and Engineering, ETH ZurichZurichSwitzerland
| | - Alexander Yermanos
- Center for Translational Immunology, University Medical Centre Utrecht, Utrecht UniversityUtrechtNetherlands
- Department of Biosystems Science and Engineering, ETH ZurichZurichSwitzerland
| | - Jürgen J Kuball
- Center for Translational Immunology, University Medical Centre Utrecht, Utrecht UniversityUtrechtNetherlands
- Department of Hematology, University Medical Center Utrecht, Utrecht UniversityUrechtNetherlands
| | - Can Kesmir
- Theoretical Biology and Bioinformatics, Department of Biology, Utrecht UniversityUtrechtNetherlands
| | - M Ines Pascoal Ramos
- Center for Translational Immunology, University Medical Centre Utrecht, Utrecht UniversityUtrechtNetherlands
- Oncode InstituteUtrechtNetherlands
| | - Linde Meyaard
- Center for Translational Immunology, University Medical Centre Utrecht, Utrecht UniversityUtrechtNetherlands
- Oncode InstituteUtrechtNetherlands
| |
Collapse
|
3
|
Liu Y, Zhang N, Wen Y, Wen J. Head and neck cancer: pathogenesis and targeted therapy. MedComm (Beijing) 2024; 5:e702. [PMID: 39170944 PMCID: PMC11338281 DOI: 10.1002/mco2.702] [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: 01/30/2024] [Revised: 08/02/2024] [Accepted: 08/05/2024] [Indexed: 08/23/2024] Open
Abstract
Head and neck cancer (HNC) is a highly aggressive type of tumor characterized by delayed diagnosis, recurrence, metastasis, relapse, and drug resistance. The occurrence of HNC were associated with smoking, alcohol abuse (or both), human papillomavirus infection, and complex genetic and epigenetic predisposition. Currently, surgery and radiotherapy are the standard treatments for most patients with early-stage HNC. For recurrent or metastatic (R/M) HNC, the first-line treatment is platinum-based chemotherapy combined with the antiepidermal growth factor receptor drug cetuximab, when resurgery and radiation therapy are not an option. However, curing HNC remains challenging, especially in cases with metastasis. In this review, we summarize the pathogenesis of HNC, including genetic and epigenetic changes, abnormal signaling pathways, and immune regulation mechanisms, along with all potential therapeutic strategies such as molecular targeted therapy, immunotherapy, gene therapy, epigenetic modifications, and combination therapies. Recent preclinical and clinical studies that may offer therapeutic strategies for future research on HNC are also discussed. Additionally, new targets and treatment methods, including antibody-drug conjugates, photodynamic therapy, radionuclide therapy, and mRNA vaccines, have shown promising results in clinical trials, offering new prospects for the treatment of HNC.
Collapse
Affiliation(s)
- Yan Liu
- Frontiers Medical CenterTianfu Jincheng LaboratoryChengduChina
- National Facility for Translational Medicine (Sichuan)West China Hospital of Sichuan UniversityChengduChina
| | - Nannan Zhang
- National Center for Birth Defect MonitoringKey Laboratory of Birth Defects and Related Diseases of Women and ChildrenMinistry of EducationWest China Second University HospitalSichuan UniversityChengduChina
| | - Yi Wen
- State Key Laboratory of BiotherapyWest China Hospital of Sichuan UniversityChengduChina
| | - Jiaolin Wen
- Frontiers Medical CenterTianfu Jincheng LaboratoryChengduChina
| |
Collapse
|
4
|
Yuan L, Wang Y, Shen X, Ma F, Wang J, Yan F. Soluble form of immune checkpoints in autoimmune diseases. J Autoimmun 2024; 147:103278. [PMID: 38943864 DOI: 10.1016/j.jaut.2024.103278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 05/03/2024] [Accepted: 06/19/2024] [Indexed: 07/01/2024]
Abstract
Immune checkpoints are essential regulators of immune responses, either by activating or suppressing them. Consequently, they are regarded as pivotal elements in the management of infections, cancer, and autoimmune disorders. In recent years, researchers have identified numerous soluble immune checkpoints that are produced through various mechanisms and demonstrated biological activity. These soluble immune checkpoints can be produced and distributed in the bloodstream and various tissues, with their roles in immune response dysregulation and autoimmunity extensively documented. This review aims to provide a thorough overview of the generation of various soluble immune checkpoints, such as sPD-1, sCTLA-4, sTim-3, s4-1BB, sBTLA, sLAG-3, sCD200, and the B7 family, and their importance as indicators for the diagnosis and prediction of autoimmune conditions. Furthermore, the review will investigate the potential pathological mechanisms of soluble immune checkpoints in autoimmune diseases, emphasizing their association with autoimmune diseases development, prognosis, and treatment.
Collapse
Affiliation(s)
- Li Yuan
- Geriatric Diseases Institute of Chengdu, Department of Clinical Laboratory, Chengdu Fifth People's Hospital, Chengdu, Sichuan Province, China
| | - Yuxia Wang
- Geriatric Intensive Care Unit, Sichuan Geriatric Medical Center, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan Province, China
| | - Xuxia Shen
- Geriatric Diseases Institute of Chengdu, Department of Clinical Laboratory, Chengdu Fifth People's Hospital, Chengdu, Sichuan Province, China
| | - Fujun Ma
- Department of Training, Chengdu Fifth People's Hospital, Chengdu, Sichuan Province, China
| | - Jun Wang
- Department of Respiratory and Critical Care Medicine, Chengdu Fifth People's Hospital, Chengdu, Sichuan Province, China.
| | - Fang Yan
- Geriatric Diseases Institute of Chengdu, Department of Geriatrics, Chengdu Fifth People's Hospital, Chengdu, Sichuan Province, China; Geriatric Diseases Institute of Chengdu, Department of Intensive Care Medicine, Chengdu Fifth People's Hospital, Chengdu, Sichuan Province, China; Center for Medicine Research and Translation, Chengdu Fifth People's Hospital, Chengdu, Sichuan Province, China.
| |
Collapse
|
5
|
Kaya Akca U, Sag E, Aydın B, Tasdemir NK, Kasap Cuceoglu M, Basaran O, Batu ED, Bilginer Y, Ozen S. Chronic non-bacterial osteomyelitis and immune checkpoint molecules. Clin Rheumatol 2024; 43:553-560. [PMID: 37676588 DOI: 10.1007/s10067-023-06761-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 08/18/2023] [Accepted: 08/27/2023] [Indexed: 09/08/2023]
Abstract
OBJECTIVE We aimed to investigate the plasma levels and cell surface expression of two checkpoint molecules, TIM-3 (T cell immunoglobulin and mucin domain-containing protein 3) and PD-1 (programmed cell death protein 1), in pediatric patients with chronic non-bacterial osteomyelitis (CNO). METHODS Plasma samples of CNO patients were collected at diagnosis or during biologic agent treatment. Plasma levels of TIM-3 and PD-1 were measured using the sandwich enzyme-linked immunosorbent assay method, and the expression of the two immune checkpoint molecules on the cell surface was analyzed by isolating peripheral blood mononuclear cells by density gradient centrifugation technique. RESULTS Twenty-seven patients with CNO (14 boys, 51.9%) and six healthy controls (3 boys, 50%) were enrolled in the study. There were no age differences between CNO patients and healthy controls (median age 14.5 vs. 13.5 years, respectively, p=0.762). Of the CNO patients, 18 were included at the time of diagnosis while 9 were receiving biologic treatment at enrollment. The median plasma PD-1 levels were significantly lower in the CNO group than in the healthy controls (p=0.011). However, no significant difference was found in the cellular expression of PD-1 and TIM-3 on CD3+CD4+ T cells in patients and healthy controls (p=0.083 and p=0.245, respectively). There was also no statistically significant difference in plasma TIM-3 levels of the patient and control groups (p=0.981). CONCLUSION CNO is an autoinflammatory disease, and overall, our results suggest that T cell exhaustion may not be significant in CNO. Further research is needed to find out whether the immune checkpoints are mainly associated with autoimmunity but not autoinflammation. Key Points • The median plasma PD-1 levels were significantly lower in the CNO group than in the healthy controls. • No significant difference was found in the cellular expression of PD-1 and TIM-3 on CD3+CD4+ T cells in patients and healthy controls. • Our results suggest that T cell exhaustion may not be significant in CNO pathogenesis.
Collapse
Affiliation(s)
- Ummusen Kaya Akca
- Division of Pediatric Rheumatology, Department of Pediatrics, Faculty of Medicine, Hacettepe University, Sihhiye Campus, 06100, Ankara, Turkey
| | - Erdal Sag
- Division of Pediatric Rheumatology, Department of Pediatrics, Faculty of Medicine, Hacettepe University, Sihhiye Campus, 06100, Ankara, Turkey
- Translational Medicine Laboratories, Pediatric Rheumatology Unit, Hacettepe University, Ankara, Turkey
| | - Busra Aydın
- Translational Medicine Laboratories, Pediatric Rheumatology Unit, Hacettepe University, Ankara, Turkey
| | - Nur Kubra Tasdemir
- Translational Medicine Laboratories, Pediatric Rheumatology Unit, Hacettepe University, Ankara, Turkey
| | - Muserref Kasap Cuceoglu
- Division of Pediatric Rheumatology, Department of Pediatrics, Faculty of Medicine, Hacettepe University, Sihhiye Campus, 06100, Ankara, Turkey
| | - Ozge Basaran
- Division of Pediatric Rheumatology, Department of Pediatrics, Faculty of Medicine, Hacettepe University, Sihhiye Campus, 06100, Ankara, Turkey
| | - Ezgi Deniz Batu
- Division of Pediatric Rheumatology, Department of Pediatrics, Faculty of Medicine, Hacettepe University, Sihhiye Campus, 06100, Ankara, Turkey
| | - Yelda Bilginer
- Division of Pediatric Rheumatology, Department of Pediatrics, Faculty of Medicine, Hacettepe University, Sihhiye Campus, 06100, Ankara, Turkey
| | - Seza Ozen
- Division of Pediatric Rheumatology, Department of Pediatrics, Faculty of Medicine, Hacettepe University, Sihhiye Campus, 06100, Ankara, Turkey.
- Translational Medicine Laboratories, Pediatric Rheumatology Unit, Hacettepe University, Ankara, Turkey.
| |
Collapse
|
6
|
Cieplińska K, Niedziela E, Kowalska A. Immunological Processes in the Orbit and Indications for Current and Potential Drug Targets. J Clin Med 2023; 13:72. [PMID: 38202079 PMCID: PMC10780108 DOI: 10.3390/jcm13010072] [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: 11/12/2023] [Revised: 12/17/2023] [Accepted: 12/20/2023] [Indexed: 01/12/2024] Open
Abstract
Thyroid eye disease (TED) is an extrathyroidal manifestation of Graves' disease (GD). Similar to GD, TED is caused by an autoimmune response. TED is an autoimmune inflammatory disorder of the orbit and periorbital tissues, characterized by upper eyelid retraction, swelling, redness, conjunctivitis, and bulging eyes. The pathophysiology of TED is complex, with the infiltration of activated T lymphocytes and activation of orbital fibroblasts (OFs) and autoantibodies against the common autoantigen of thyroid and orbital tissues. Better understanding of the multifactorial pathogenesis of TED contributes to the development of more effective therapies. In this review, we present current and potential drug targets. The ideal treatment should slow progression of the disease with as little interference with patient immunity as possible. In the future, TED treatment will target the immune mechanism involved in the disease and will be based on a strategy of restoring tolerance to autoantigens.
Collapse
Affiliation(s)
| | - Emilia Niedziela
- Collegium Medicum, Jan Kochanowski University in Kielce, 25-317 Kielce, Poland; (E.N.); (A.K.)
- Department of Endocrinology, Holy Cross Cancer Center, 25-734 Kielce, Poland
| | - Aldona Kowalska
- Collegium Medicum, Jan Kochanowski University in Kielce, 25-317 Kielce, Poland; (E.N.); (A.K.)
- Department of Endocrinology, Holy Cross Cancer Center, 25-734 Kielce, Poland
| |
Collapse
|
7
|
Zhao R, Jiang Y, Zhang J, Huang Y, Xiong C, Zhao Z, Huang T, Liu W, Zhou N, Li Z, Luo X, Tang Y. Development and validation of a novel necroptosis-related gene signature for predicting prognosis and therapeutic response in Ewing sarcoma. Front Med (Lausanne) 2023; 10:1239487. [PMID: 37663658 PMCID: PMC10470467 DOI: 10.3389/fmed.2023.1239487] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 08/02/2023] [Indexed: 09/05/2023] Open
Abstract
Ewing sarcoma (ES) is the second most common malignant bone tumor in children and has a poor prognosis due to early metastasis and easy recurrence. Necroptosis is a newly discovered cell death method, and its critical role in tumor immunity and therapy has attracted widespread attention. Thus, the emergence of necroptosis may provide bright prospects for the treatment of ES and deserves our further study. Here, based on the random forest algorithm, we identified 6 key necroptosis-related genes (NRGs) and used them to construct an NRG signature with excellent predictive performance. Subsequent analysis showed that NRGs were closely associated with ES tumor immunity, and the signature was also good at predicting immunotherapy and chemotherapy response. Next, a comprehensive analysis of key genes showed that RIPK1, JAK1, and CHMP7 were potential therapeutic targets. The Cancer Dependency Map (DepMap) results showed that CHMP7 is associated with ES cell growth, and the Gene Set Cancer Analysis (GSCALite) results revealed that the JAK1 mutation frequency was the highest. The expression of 3 genes was all negatively correlated with methylation and positively with copy number variation (CNV). Finally, an accurate nomogram was constructed with this signature and clinical traits. In short, this study constructed an accurate prognostic signature and identified 3 novel therapeutic targets against ES.
Collapse
Affiliation(s)
- Runhan Zhao
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Orthopedic Laboratory of Chongqing Medical University, Chongqing, China
| | - Yu Jiang
- School of Public Health, Chongqing Medical University, Chongqing, China
| | - Jun Zhang
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Orthopedic Laboratory of Chongqing Medical University, Chongqing, China
| | - Yanran Huang
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Orthopedic Laboratory of Chongqing Medical University, Chongqing, China
| | - Chuang Xiong
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Orthopedic Laboratory of Chongqing Medical University, Chongqing, China
| | - Zenghui Zhao
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Orthopedic Laboratory of Chongqing Medical University, Chongqing, China
| | - Tianji Huang
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Orthopedic Laboratory of Chongqing Medical University, Chongqing, China
| | - Wei Liu
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Orthopedic Laboratory of Chongqing Medical University, Chongqing, China
| | - Nian Zhou
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Orthopedic Laboratory of Chongqing Medical University, Chongqing, China
| | - Zefang Li
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Department of Orthopedics, Qianjiang Central Hospital of Chongqing, Chongqing, China
| | - Xiaoji Luo
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Orthopedic Laboratory of Chongqing Medical University, Chongqing, China
| | - Yongli Tang
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
8
|
Fu X, Zhou X, Ji F, He Q, Qiu X. Systematic analysis of the prognosis and immune infiltration of E2Fs in thyroid carcinoma. Front Genet 2023; 14:1215984. [PMID: 37560385 PMCID: PMC10407799 DOI: 10.3389/fgene.2023.1215984] [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: 05/03/2023] [Accepted: 07/10/2023] [Indexed: 08/11/2023] Open
Abstract
Purpose: To evaluate the diagnostic and prognostic utility of E2F transcription factors (E2Fs) in thyroid carcinoma (THCA) and their association with immune infiltration. Methods: The transcription and protein levels of E2Fs in THCA tissues were examined using the R language and the Human Protein Atlas (HPA) database in this research. We utilized the UALCAN and GEPIA2 databases to analyze the association between the level of E2Fs and the clinicopathological features of THCA. The prognostic utility of E2F expression in THCA was studied using the R language and the Gene Set Cancer Analysis (GSCA) database. Over-representation analysis (ORA) and gene set enrichment analysis (GSEA) were employed to analyze the effect of E2F family members. The TISIDB database and Tumor Immune Estimation Resource (TIMER) database were utilized to investigate the relationship between E2F expression and the level of immune infiltration in thyroid cancer. Results: E2Fs are highly conserved in thyroid carcinoma and rarely mutated. E2Fs are strongly expressed in THCA and are highly related with the clinicopathological stage of THCA. Patients with THCA have a poor prognosis when E2Fs are highly expressed. The function of E2Fs in THCA may be closely related to the renin-angiotensin system (Ras) signaling pathway, platelet-derived growth factor (PDGF) signaling pathway, apoptosis, and immune response. With regard to the immune infiltration, E2F expression and tumor-infiltrating lymphocytes exhibited a positive connection. Conclusion: The level of E2Fs is connected with the prognosis and immune infiltration level in THCA, revealing that E2Fs may be a prognostic and immune infiltration cell marker in THCA patients.
Collapse
Affiliation(s)
- Xinghao Fu
- Department of Thyroid Surgery, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Xing Zhou
- Department of Plastic and Aesthetic Surgery, Henan Provincial People’s Hospital, Zhengzhou, Henan, China
| | - Feihong Ji
- Department of Thyroid Surgery, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Qi He
- Department of Thyroid Surgery, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Xinguang Qiu
- Department of Thyroid Surgery, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
9
|
Edwards A, Chandran V, Rahman P. Investigational monoclonal antibodies in early development for psoriatic arthritis: beyond the biosimilars. Expert Opin Investig Drugs 2023; 32:741-753. [PMID: 37655430 DOI: 10.1080/13543784.2023.2254684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 08/08/2023] [Accepted: 08/30/2023] [Indexed: 09/02/2023]
Abstract
INTRODUCTION Psoriatic Arthritis (PsA) is an inflammatory arthritis that is present in approximately 25% of psoriasis patients. Currently, several targeted therapies are available to manage PsA; however, many patients fail these therapies. Several new therapeutic options, with differing mechanisms of action, are currently being evaluated. AREAS COVERED This article reviews available results from phase I to phase III trials of several investigational monoclonal antibodies that the FDA has not yet approved for PsA. The proposed mechanisms of the new therapeutic agents and their relevance to the pathogenesis of PsA will be discussed. The investigational agents' efficacy and safety will be summarized, and their potential clinical applications for managing PsA will be contemplated. EXPERT OPINION Due to recent advances in understanding psoriatic arthritis, therapeutic agents are increasingly focused on inhibiting interleukin-17 and interleukin-23 pathways. Various strategies have been used to inhibit these cytokines, demonstrating favorable efficacy and acceptable safety profile.
Collapse
Affiliation(s)
- Anna Edwards
- Faculty of Pharmacy, Memorial University of Newfoundland, St. John's, Newfoundland, Canada
| | - Vinod Chandran
- Department of Medicine, Division of Rheumatology, University of Toronto, Toronto, Ontario, Canada
| | - Proton Rahman
- Department of Medicine, Division of Rheumatology, Memorial University of Newfoundland, St. John's, Newfoundland, Canada
| |
Collapse
|
10
|
Bibi Z, Ashraf K, Shehzadi A, Rehman A, Abbas Bukhari D. Evaluation of isolated probiotics on the efficacy of immune system in male and female Wistar rats. Saudi Pharm J 2023; 31:1036-1046. [PMID: 37250360 PMCID: PMC10209143 DOI: 10.1016/j.jsps.2023.04.023] [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: 12/28/2022] [Accepted: 04/23/2023] [Indexed: 05/31/2023] Open
Abstract
Probiotics were isolated from fruits and vegetables. Microscopic, biochemical, and molecular tests were carried out for the characterization of strains of probiotics. To assess the effects of isolated probiotics on immunity, male and female (15 + 15) Wistar rats (n = 3) were randomly distributed into 5 groups: 0-day, negative control, positive control (commercially available Lactobacillus acidophilus-14), laboratory isolated probiotics with accession numbers; Lactobacillus plantarum (MZ707748) and Lactobacillus plantarum (MZ729681), respectively. After hematological investigations, the amounts of IgA and IgG in male and female groups were significantly different (p < 0.05). At the same time, the values of Alanine-transaminase (ALT) and Aspartate-aminotransferase (AST) in both genders were average, and there were no differences (p > 0.05). Male probiotic-treated groups had decreased levels of interleukin-6, bilirubin, and creatinine, but female probiotic-treated groups had a slight rise in bilirubin and creatinine values (p = 0.05). Cellular blood count levels of Hematocrit (HCT) and white blood cells (WBC) in male groups showed considerable differences (p < 0.05), while there were no differences (p > 0.05) in female groups. Levels of Red blood cells (RBC) and mean corpuscular hemoglobin concentration (MCHC) showed distinct changes (p < 0.05) in female groups, while these values were insignificant changes (p > 0.05) among male groups. There were considerable differences between the control and groups that were given probiotics. Histopathological results showed no damage to the liver and thymus. A fecal examination of rats was used to examine the viability and survival of Lactobacilli. Based on blood tests, it was observed that the immune system was boosted and improved in probiotic-treated groups compared to control groups.
Collapse
Affiliation(s)
- Zuhra Bibi
- Department of Zoology, Government College University, Lahore, Pakistan
| | - Khadija Ashraf
- Department of Zoology, Government College University, Lahore, Pakistan
| | - Areeba Shehzadi
- Department of Zoology, Government College University, Lahore, Pakistan
| | - Abdul Rehman
- Institute of Microbiology and Molecular Genetics, University of the Punjab, Quaid-e-Azam Campus, 54590 Lahore, Pakistan
| | | |
Collapse
|
11
|
Hashemi B, Abdollahi M, Abbaspour-Aghdam S, Hazrati A, Malekpour K, Meshgi S, Kafil HS, Ghazi F, Yousefi M, Roshangar L, Ahmadi M. The effect of probiotics on immune responses and their therapeutic application: A new treatment option for multiple sclerosis. Biomed Pharmacother 2023; 159:114195. [PMID: 36630847 DOI: 10.1016/j.biopha.2022.114195] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 12/10/2022] [Accepted: 12/28/2022] [Indexed: 01/12/2023] Open
Abstract
Multiple sclerosis (MS) is known as a chronic inflammatory disease (CID) that affects the central nervous system and leads to nerve demyelination. However, the exact cause of MS is unknown, but immune system regulation and inhibiting the function of inflammatory pathways may have a beneficial effect on controlling and improving the disease. Studies show that probiotics can alter the gut microbiome, thereby improving and affecting the immune system and inflammatory responses in patients with MS. The results show that probiotics have a good effect on the recovery of patients with MS in humans and animals. The present study investigated the effect of probiotics and possible therapeutic mechanisms of probiotics on immune cells and inflammatory cytokines. This review article showed that probiotics could improve immune cells and inflammatory cytokines in patients with MS and can play an effective role in disease management and control.
Collapse
Affiliation(s)
- Behnam Hashemi
- Department of Bacteriology, Faculty of Medical Science, Tarbiat Modares University, Tehran, Iran
| | - Maryam Abdollahi
- Department of Bacteriology, Faculty of Medical Science, Tarbiat Modares University, Tehran, Iran
| | - Sanaz Abbaspour-Aghdam
- Department of Clinical Biochemistry and Applied Cell Sciences, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Ali Hazrati
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Kosar Malekpour
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Shahla Meshgi
- Cardiovascular Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hossein Samadi Kafil
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Farhood Ghazi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mehdi Yousefi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Leila Roshangar
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Majid Ahmadi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
12
|
Meng L, Lu H, Li Y, Zhao J, He S, Wang Z, Shen J, Huang H, Xiao J, Sooranna SR, Song J. Human papillomavirus infection can alter the level of tumour stemness and T cell infiltration in patients with head and neck squamous cell carcinoma. Front Immunol 2022; 13:1013542. [PMID: 36420261 PMCID: PMC9676257 DOI: 10.3389/fimmu.2022.1013542] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Accepted: 10/17/2022] [Indexed: 07/20/2023] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC) usually has a poor prognosis and is associated with a high mortality rate. Its etiology is mainly the result from long-term exposure to either alcohol, tobacco or human papillomavirus (HPV) infection or a combination of these insults. However, HNSCC patients with HPV have been found to show a survival advantage over those without the virus, but the mechanism that confers this advantage is unclear. Due to the large number of HPV-independent HNSCC cases, there is a possibility that the difference in prognosis between HPV-positive (HPV+) and negative (HPV-) patients is due to different carcinogens. To clarify this, we used scRNA data and viral tracking methods in order to identify HPV+ and HPV- cells in the tumour tissues of patients infected with HPV. By comparing HPV+ and HPV- malignant cells, we found a higher level of tumour stemness in HPV- tumour cells. Using tumour stemness-related genes, we established a six-gene prognostic signature that was used to divide the patients into low- and high-risk groups. It was found that HPV patients who were at low-risk of contracting HNSCC had a higher number of CD8+ T-cells as well as a higher expression of immune checkpoint molecules. Correspondingly, we found that HPV+ tumour cells expressed higher levels of CCL4, and these were highly correlated with CD8+ T cells infiltration and immune checkpoint molecules. These data suggest that the stemness features of tumour cells are not only associated with the prognostic risk, but that it could also affect the immune cell interactions and associated signalling pathways.
Collapse
Affiliation(s)
- Lingzhang Meng
- Institute of Cardiovascular Sciences, Guangxi Academy of Medical Sciences, Nanning, China
- Center for Systemic Inflammation Research (CSIR), School of Preclinical Medicine, Youjiang Medical University for Nationalities, Baise, China
| | - Heming Lu
- Second Division of Department of Radiation Oncology, Guangxi Academy of Medical Sciences, Nanning, China
- Second Division of Department of Radiation Oncology, the People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Yueyong Li
- Department of Interventive Medicine, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
| | - Jingjie Zhao
- Life Science and Clinical Research Center, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
| | - Siyuan He
- Center for Systemic Inflammation Research (CSIR), School of Preclinical Medicine, Youjiang Medical University for Nationalities, Baise, China
| | - Zechen Wang
- Center for Systemic Inflammation Research (CSIR), School of Preclinical Medicine, Youjiang Medical University for Nationalities, Baise, China
| | - Jiajia Shen
- Center for Systemic Inflammation Research (CSIR), School of Preclinical Medicine, Youjiang Medical University for Nationalities, Baise, China
| | - Huixian Huang
- Second Division of Department of Radiation Oncology, Guangxi Academy of Medical Sciences, Nanning, China
- Second Division of Department of Radiation Oncology, the People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Jinru Xiao
- Graduate School, Guangxi University of Chinese Medicine, Nanning, China
| | - Suren Rao Sooranna
- Department of Metabolism, Digestion and Reproduction, Imperial College London, Chelsea & Westminster Hospital, London, United Kingdom
| | - Jian Song
- Institute of Cardiovascular Sciences, Guangxi Academy of Medical Sciences, Nanning, China
- Center for Systemic Inflammation Research (CSIR), School of Preclinical Medicine, Youjiang Medical University for Nationalities, Baise, China
| |
Collapse
|
13
|
Ding W, Li B, Zhang Y, He L, Su J. A neutrophil extracellular traps-associated lncRNA signature predicts the clinical outcomes in patients with lung adenocarcinoma. Front Genet 2022; 13:1047231. [PMID: 36419832 PMCID: PMC9676361 DOI: 10.3389/fgene.2022.1047231] [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: 09/17/2022] [Accepted: 10/25/2022] [Indexed: 10/24/2023] Open
Abstract
Backgrounds: Neutrophil extracellular traps (NETs) play an important role in the occurrence, metastasis, and immune escape of cancers. We aim to investigate Long non-coding RNAs (lncRNAs) that are correlated to NETs to find some potentially useful biomarkers for lung adenocarcinoma (LUAD), and to explore their correlations with immunotherapy and chemotherapy, as well as the tumor microenvironment. Methods: Based on the The Cancer Genome Atlas (TCGA) database, we identified the prognosis-related lncRNAs which are associated with NETs using cox regression. The patients were then separated into two clusters based on the expression of NETs-associated lncRNAs to perform tumor microenvironment analysis and immune-checkpoint analysis. Least absolute shrinkage and selection operator (LASSO) regression was then performed to establish a prognostic signature. Furthermore, nomogram analysis, tumor mutation burden analysis, immune infiltration analysis, as well as drug sensitivity analysis were performed to test the signature. Results: Using univariate cox regression, we found 10 NETs-associated lncRNAs that are associated with the outcomes of LUAD patients. Also, further analysis which separated the patients into 2 clusters showed that the 10 lncRNAs had significant correlations with the tumor microenvironment. Using LASSO regression, we finally constructed a signature to predict the outcomes of the patients based on 4 NETs-associated lncRNAs. The 4 NETs-associated lncRNAs were namely SIRLNT, AL365181.3, FAM83A-AS1, and AJ003147.2. Using Kaplan-Meier (K-M) analysis, we found that the risk model was strongly associated with the survival outcomes of the patients both in the training group and in the validation group 1 and 2 (p < 0.001, p = 0.026, and p < 0.01). Using receiver operating characteristic (ROC) curve, we tested the sensitivity combined with the specificity of the model and found that the risk model had a satisfactory level of 1-year, 3-year, and 5-year concordance index (C-index) (C = 0.661 in the training group, C = 0.679 in validation group 1, C = 0.692 in validation group 2). We also explored the immune microenvironment and immune checkpoint correlation of the risk model and found some significant results. Conclusion: We constructed a NETs-associated lncRNA signature to predict the outcome of patients with LUAD, which is associated with immunephenoscores and immune checkpoint-gene expression.
Collapse
Affiliation(s)
- Wencong Ding
- Department of Rheumatology, Affiliated Guangdong Hospital of Integrated Traditional Chinese and Western Medicine of Guangzhou University of Chinese Medicine, Foshan, Guangdong, China
| | - Biyi Li
- Department of Emergency Foshan Hospital of Traditional Chinese Medicine, Foshan, Guangdong, China
| | - Yuan Zhang
- Department of Emergency, Affiliated Guangdong Hospital of Integrated Traditional Chinese and Western Medicine of Guangzhou University of Chinese Medicine, Foshan, Guangdong, China
| | - Liu He
- Guangdong Hospital of Traditional Chinese Medicine, Guangzhou, Guangdong, China
| | - Junwei Su
- Department of Emergency, Affiliated Guangdong Hospital of Integrated Traditional Chinese and Western Medicine of Guangzhou University of Chinese Medicine, Foshan, Guangdong, China
| |
Collapse
|
14
|
Abstract
The concept of precision medicine is based on the identification of hallmarks of cancer to exploit them as drug targets. The basic idea was that in this way the therapeutic modalities will be more effective and the side effects will be less. Since the majority of these novel modalities are not specific for a cancer-related biological process or a cancer-specific (mutant) target protein, it is not a surprise that we had to learn new type of side effects, because these therapeutics also affect physiological or pathological processes. Even more, in cases of some of these novel therapies we were able to discover new molecular mechanisms of physiological and pathological processes. Identification of the on-target side effects of targeted drugs can help to prevent the development of them or better manage the patients when emerge during cancer therapy.
Collapse
Affiliation(s)
- József Tímár
- Departments of Pathology, Forensic and Insurance Medicine, Semmelweis University, Budapest, Hungary
- *Correspondence: József Tímár,
| | - Andrea Uhlyarik
- Internal Medicine and Oncology, Semmelweis University, Budapest, Hungary
| |
Collapse
|
15
|
Zhai Y, Dong S, Li H, Zhang Y, Shami P, Chen M. Antibody-mediated depletion of programmed death 1-positive (PD-1 +) cells. J Control Release 2022; 349:425-433. [PMID: 35820540 PMCID: PMC10699550 DOI: 10.1016/j.jconrel.2022.07.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 06/17/2022] [Accepted: 07/06/2022] [Indexed: 10/17/2022]
Abstract
PD-1 immune checkpoint has been intensively investigated in pathogenesis and treatments for cancer and autoimmune diseases. Cells that express PD-1 (PD-1+ cells) draw ever-increasing attention in cancer and autoimmune disease research although the role of PD-1+ cells in the progression and treatments of these diseases remains largely ambiguous. One definite approach to elucidate their roles is to deplete these cells in disease settings and examine how the depletion impacts disease progression and treatments. To execute the depletion, we designed and generated the first depleting antibody (D-αPD-1) that specifically ablates PD-1+ cells. D-αPD-1 has the same variable domains as an anti-mouse PD-1 blocking antibody (RMP1-14). The constant domains of D-αPD-1 were derived from mouse IgG2a heavy and κ-light chain, respectively. D-αPD-1 was verified to bind with mouse PD-1 as well as mouse FcγRIV, an immuno-activating Fc receptor. The cell depletion effect of D-αPD-1 was confirmed in vivo using a PD-1+ cell transferring model. Since transferred PD-1+ cells, EL4 cells, are tumorigenic and EL4 tumors are lethal to host mice, the depleting effect of D-αPD-1 was also manifested by an absolute survival among the antibody-treated mice while groups receiving control treatments had median survival time of merely approximately 30 days. Furthermore, we found that D-αPD-1 leads to elimination of PD-1+ cells through antibody-dependent cell-mediate phagocytosis (ADCP) and complement-dependent cytotoxicity (CDC) mechanisms. These results, altogether, confirmed the specificity and effectiveness of D-αPD-1. The results also highlighted that D-αPD-1 is a robust tool to study PD-1+ cells in cancer and autoimmune diseases and a potential therapeutic for these diseases.
Collapse
Affiliation(s)
- Yujia Zhai
- Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT 84112, USA
| | - Shuyun Dong
- Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT 84112, USA
| | - Haojia Li
- Division of Epidemiology, Department of Internal Medicine, University of Utah, Salt Lake City, UT 84132, USA
| | - Yue Zhang
- Division of Epidemiology, Department of Internal Medicine, University of Utah, Salt Lake City, UT 84132, USA
| | - Paul Shami
- Division of Hematology and Hematologic Malignancies, Department of Internal Medicine, and Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Mingnan Chen
- Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT 84112, USA.
| |
Collapse
|
16
|
Gerasimova EV, Tabakov DV, Gerasimova DA, Popkova TV. Activation Markers on B and T Cells and Immune Checkpoints in Autoimmune Rheumatic Diseases. Int J Mol Sci 2022; 23:ijms23158656. [PMID: 35955790 PMCID: PMC9368764 DOI: 10.3390/ijms23158656] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Revised: 07/27/2022] [Accepted: 08/02/2022] [Indexed: 11/17/2022] Open
Abstract
In addition to identifying the major B- and T-cell subpopulations involved in autoimmune rheumatic diseases (ARDs), in recent years special attention has been paid to studying the expression of their activation markers and immune checkpoints (ICPs). The activation markers on B and T cells are a consequence of the immune response, and these molecules are considered as sensitive specific markers of ARD activity and as promising targets for immunotherapy. ICPs regulate the activation of the immune response by preventing the initiation of autoimmune processes, and they modulate it by reducing immune cell-induced organ and tissue damage. The article considers the possible correlation of ICPs with the activity of ARDs, the efficacy of specific ARD treatments, and the prospects for the use of activation molecules and activation/blocking ICPs for the treatment of ARDs.
Collapse
Affiliation(s)
- Elena V. Gerasimova
- Department of Systemic Rheumatic Diseases, V.A. Nasonova Research Institute of Rheumatology, Kashirskoe Shosse, 115522 Moscow, Russia
- Correspondence: ; Tel.: +7-905-538-0399
| | - Dmitry V. Tabakov
- Department of Systemic Rheumatic Diseases, V.A. Nasonova Research Institute of Rheumatology, Kashirskoe Shosse, 115522 Moscow, Russia
| | - Daria A. Gerasimova
- Department of Organization and Economy of Pharmacy, Institute of Pharmacy, I.M. Sechenov First Moscow State Medical University (Sechenov University), 8/2, Trubetskaya St., 119526 Moscow, Russia
| | - Tatiana V. Popkova
- Department of Systemic Rheumatic Diseases, V.A. Nasonova Research Institute of Rheumatology, Kashirskoe Shosse, 115522 Moscow, Russia
| |
Collapse
|
17
|
Davern M, Fitzgerald MC, Buckley CE, Heeran AB, Donlon NE, McGrath J, O' Connell F, Deshpande MR, Hayes C, MacDonald J, Sheppard AD, Reynolds JV, Maher SG, Lynam-Lennon N, Murphy B, Lysaght J. PD-1 and TIGIT blockade differentially affect tumour cell survival under hypoxia and glucose deprived conditions in oesophageal adenocarcinoma; implications for overcoming resistance to PD-1 blockade in hypoxic tumours. Transl Oncol 2022; 19:101381. [PMID: 35245832 PMCID: PMC8894275 DOI: 10.1016/j.tranon.2022.101381] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 02/18/2022] [Indexed: 12/17/2022] Open
Abstract
PD-1 and TIGIT expression are highly expressed on the surface of oesophageal epithelial cells during the early stages of metaplasia. Glucose deprivation and hypoxia upregulate PD-1 and TIGIT on the surface of oesophageal adenocarcinoma (OAC) cells in vitro. PD-1 and TIGIT blockade decrease Bcl-2 and Bcl-xL expression in OAC cells. PD-1 blockade in OAC cells enhances basal respiration and glycolytic reserve and upregulates GLUT1 on the surface of a subpopulation of OAC cells. PD-1 inhibition confers a survival advantage to OAC cells under glucose deprivation and hypoxia. TIGIT blockade decreases OAC cell proliferation and induces OAC cell death under normoxia, hypoxia and nutrient deprivation. TIGIT blockade increases ECAR yet decreases a range of metabolic parameters in OAC cells.
Recent studies have demontrated that immune checkpoint receptors are expressed on the surface of oesophageal adenocarcinoma (OAC) cells and might confer a survival advantage. This study explores the role of PD-1 and TIGIT signalling in OAC cells in either promoting or inhibiting the survival of OAC cells under characteristic features of the tumour microenvironment including nutrient-deprivation and hypoxia. PD-1 and TIGIT are expressed in normal and pre-malignant oesophageal epithelial cells and this expression significantly decreases along the normal- Barrett's Oesophagus- OAC disease sequence. However, glucose-deprivation and hypoxia significantly upregulated PD-1 and TIGIT on the surface of OAC cells in vitro. PD-1 blockade decreased OAC cell proliferation under normoxia but enhanced proliferation and decreased cell death in OAC cells under hypoxia and glucose-deprivation. TIGIT blockade decreased proliferation and induced OAC cell death, an effect that was maintained under nutrient-deprivation and hypoxia. Basal respiration and glycolytic reserve were enhanced and GLUT1 was upregulated on the surface of a subpopulation of OAC cells following PD-1 blockade. In contrast, TIGIT blockade enhanced a glycolytic phenotype in OAC cells, yet decreased other metabolic parameters including oxidative phosphorylation and basal respiration. Interestingly, inhibition of oxidative phosphorylation significantly upregulated TIGIT expression and inhibition of oxidative phosphorylation and glycolysis significantly decreased PD-1 on the surface of a subpopulation of OAC cells in vitro. These findings suggest an immune-independent mechanism for PD-1 inhibitor resistance in hypoxic tumours and suggest that TIGIT might be a more effective therapeutic target in OAC compared with PD-1 for treating hypoxic tumours.
Collapse
Affiliation(s)
- Maria Davern
- Cancer Immunology and Immunotherapy Group, Department of Surgery, Trinity St. James's Cancer Institute, Trinity Translational Medicine Institute, St. James's Hospital campus, Dublin 8, Ireland
| | - Marie-Claire Fitzgerald
- Department of Physiology & Medical Physics, Royal College of Surgeons in Ireland, D02 YN77 Dublin 2, Ireland
| | - Croí E Buckley
- Translational Radiobiology and Diagnostics Group, Department of Surgery, Trinity St. James's Cancer Institute, Trinity Translational Medicine Institute, St. James's Hospital campus, Dublin 8, Ireland
| | - Aisling B Heeran
- Translational Gastrointestinal Research Group, Department of Surgery, Trinity St. James's Cancer Institute, Trinity Translational Medicine Institute, St. James's Hospital campus, Dublin 8, Ireland
| | - Noel E Donlon
- Cancer Immunology and Immunotherapy Group, Department of Surgery, Trinity St. James's Cancer Institute, Trinity Translational Medicine Institute, St. James's Hospital campus, Dublin 8, Ireland
| | - Jason McGrath
- Cancer Chemoradiation Group, Department of Surgery, Trinity St. James's Cancer Institute, Trinity Translational Medicine Institute, St. James's Hospital campus, Dublin 8, Ireland
| | - Fiona O' Connell
- Translational Gastrointestinal Research Group, Department of Surgery, Trinity St. James's Cancer Institute, Trinity Translational Medicine Institute, St. James's Hospital campus, Dublin 8, Ireland
| | - Malvika R Deshpande
- Cancer Immunology and Immunotherapy Group, Department of Surgery, Trinity St. James's Cancer Institute, Trinity Translational Medicine Institute, St. James's Hospital campus, Dublin 8, Ireland
| | - Conall Hayes
- Department of Surgery, Trinity St. James's Cancer Institute, Trinity Translational Medicine Institute, St. James's Hospital, Trinity College Dublin, Dublin, Ireland
| | - Jamie MacDonald
- Cancer Immunology and Immunotherapy Group, Department of Surgery, Trinity St. James's Cancer Institute, Trinity Translational Medicine Institute, St. James's Hospital campus, Dublin 8, Ireland
| | - Andrew D Sheppard
- Cancer Immunology and Immunotherapy Group, Department of Surgery, Trinity St. James's Cancer Institute, Trinity Translational Medicine Institute, St. James's Hospital campus, Dublin 8, Ireland
| | - John V Reynolds
- Department of Surgery, Trinity St. James's Cancer Institute, Trinity Translational Medicine Institute, St. James's Hospital, Trinity College Dublin, Dublin, Ireland
| | - Stephen G Maher
- Cancer Chemoradiation Group, Department of Surgery, Trinity St. James's Cancer Institute, Trinity Translational Medicine Institute, St. James's Hospital campus, Dublin 8, Ireland
| | - Niamh Lynam-Lennon
- Translational Radiobiology and Diagnostics Group, Department of Surgery, Trinity St. James's Cancer Institute, Trinity Translational Medicine Institute, St. James's Hospital campus, Dublin 8, Ireland
| | - Brona Murphy
- Department of Physiology & Medical Physics, Royal College of Surgeons in Ireland, D02 YN77 Dublin 2, Ireland
| | - Joanne Lysaght
- Cancer Immunology and Immunotherapy Group, Department of Surgery, Trinity St. James's Cancer Institute, Trinity Translational Medicine Institute, St. James's Hospital campus, Dublin 8, Ireland.
| |
Collapse
|
18
|
Tsai H, Wu Y, Liu X, Xu Z, Liu L, Wang C, Zhang H, Huang Y, Wang L, Zhang W, Su D, Khan FU, Zhu X, Yang R, Pang Y, Eriksson JE, Zhu H, Wang D, Jia B, Cheng F, Chen H. Engineered Small Extracellular Vesicles as a FGL1/PD-L1 Dual-Targeting Delivery System for Alleviating Immune Rejection. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2102634. [PMID: 34738731 PMCID: PMC8787398 DOI: 10.1002/advs.202102634] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 08/19/2021] [Indexed: 06/13/2023]
Abstract
There is an urgent need for developing new immunosuppressive agents due to the toxicity of long-term use of broad immunosuppressive agents after organ transplantation. Comprehensive sample analysis revealed dysregulation of FGL1/LAG-3 and PD-L1/PD-1 immune checkpoints in allogeneic heart transplantation mice and clinical kidney transplant patients. In order to enhance these two immunosuppressive signal axes, a bioengineering strategy is developed to simultaneously display FGL1/PD-L1 (FP) on the surface of small extracellular vesicles (sEVs). Among various cell sources, FP sEVs derived from mesenchymal stem cells (MSCs) not only enriches FGL1/PD-L1 expression but also maintain the immunomodulatory properties of unmodified MSC sEVs. Next, it is confirmed that FGL1 and PD-L1 on sEVs are specifically bound to their receptors, LAG-3 and PD-1 on target cells. Importantly, FP sEVs significantly inhibite T cell activation and proliferation in vitro and a heart allograft model. Furthermore, FP sEVs encapsulated with low-dose FK506 (FP sEVs@FK506) exert stronger effects on inhibiting T cell proliferation, reducing CD8+ T cell density and cytokine production in the spleens and heart grafts, inducing regulatory T cells in lymph nodes, and extending graft survival. Taken together, dual-targeting sEVs have the potential to boost the immune inhibitory signalings in synergy and slow down transplant rejection.
Collapse
Affiliation(s)
- Hsiang‐i Tsai
- School of Pharmaceutical Sciences (Shenzhen)Sun Yat‐sen UniversityShenzhen518107P. R. China
- Department of Medical ImagingThe Affiliated Hospital of Jiangsu UniversityZhenjiang212001China
| | - Yingyi Wu
- School of Pharmaceutical Sciences (Shenzhen)Sun Yat‐sen UniversityShenzhen518107P. R. China
| | - Xiaoyan Liu
- School of Pharmaceutical Sciences (Shenzhen)Sun Yat‐sen UniversityShenzhen518107P. R. China
| | - Zhanxue Xu
- School of Pharmaceutical Sciences (Shenzhen)Sun Yat‐sen UniversityShenzhen518107P. R. China
| | - Longshan Liu
- Organ Transplant CenterThe First Affiliated HospitalSun Yat‐Sen University58 Zhongshan 2nd RoadGuangzhouGuangdong510080China
| | - Changxi Wang
- Organ Transplant CenterThe First Affiliated HospitalSun Yat‐Sen University58 Zhongshan 2nd RoadGuangzhouGuangdong510080China
| | - Huanxi Zhang
- Organ Transplant CenterThe First Affiliated HospitalSun Yat‐Sen University58 Zhongshan 2nd RoadGuangzhouGuangdong510080China
| | - Yisheng Huang
- Department of Oral SurgeryStomatological HospitalSouthern Medical UniversityGuangzhouGuangdong510280P. R China
| | - Linglu Wang
- School of Pharmaceutical Sciences (Shenzhen)Sun Yat‐sen UniversityShenzhen518107P. R. China
| | - Weixian Zhang
- School of Pharmaceutical Sciences (Shenzhen)Sun Yat‐sen UniversityShenzhen518107P. R. China
| | - Dandan Su
- School of Pharmaceutical Sciences (Shenzhen)Sun Yat‐sen UniversityShenzhen518107P. R. China
| | | | - Xiaofeng Zhu
- School of Traditional Medicine Materials ResourceGuangdong Pharmaceutical University YunfuGuangdong527322China
| | - Rongya Yang
- Department of DermatologyThe Seventh Medical Center of PLA General HospitalPeking100010China
| | - Yuxin Pang
- School of Traditional Medicine Materials ResourceGuangdong Pharmaceutical University YunfuGuangdong527322China
| | - John E. Eriksson
- Cell BiologyBiosciencesFaculty of Science and EngineeringÅbo Akademi UniversityTurkuFI‐20520Finland
| | - Haitao Zhu
- Department of Medical ImagingThe Affiliated Hospital of Jiangsu UniversityZhenjiang212001China
| | - Dongqing Wang
- Department of Medical ImagingThe Affiliated Hospital of Jiangsu UniversityZhenjiang212001China
| | - Bo Jia
- Department of Oral SurgeryStomatological HospitalSouthern Medical UniversityGuangzhouGuangdong510280P. R China
| | - Fang Cheng
- School of Pharmaceutical Sciences (Shenzhen)Sun Yat‐sen UniversityShenzhen518107P. R. China
| | - Hongbo Chen
- School of Pharmaceutical Sciences (Shenzhen)Sun Yat‐sen UniversityShenzhen518107P. R. China
| |
Collapse
|