1
|
Huang M, Ji Q, Huang H, Wang X, Wang L. Gut microbiota in hepatocellular carcinoma immunotherapy: immune microenvironment remodeling and gut microbiota modification. Gut Microbes 2025; 17:2486519. [PMID: 40166981 PMCID: PMC11970798 DOI: 10.1080/19490976.2025.2486519] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 03/05/2025] [Accepted: 03/25/2025] [Indexed: 04/02/2025] Open
Abstract
Hepatocellular carcinoma (HCC) remains a leading cause of cancer-related mortality, with limited treatment options at advanced stages. The gut microbiota, a diverse community of microorganisms residing in the gastrointestinal tract, plays a pivotal role in regulating immune responses through the gut-liver axis. Emerging evidence underscores its impact on HCC progression and the efficacy of immunotherapy. This review explores the intricate interactions between gut microbiota and the immune system in HCC, with a focus on key immune cells and pathways involved in tumor immunity. Additionally, it highlights strategies for modulating the gut microbiota - such as fecal microbiota transplantation, dietary interventions, and probiotics - as potential approaches to enhancing immunotherapy outcomes. A deeper understanding of these mechanisms could pave the way for novel therapeutic strategies aimed at improving patient prognosis.
Collapse
Affiliation(s)
- Mingyao Huang
- School of Basic Medicine, Putian University, Putian, Fujian, China
- Department of Breast Surgery, Clinical Oncology School of Fujian Medical University, Fuzhou, Fujian, China
| | - Quansong Ji
- Department of Urology, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Huiyan Huang
- Ward 3, De’an Hospital, Xianyou County, Putian, Fujian, China
| | - Xiaoqian Wang
- Department of Rehabilitation Medicine, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Lin Wang
- Department of Orthopedics, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
2
|
Hu M, Lv C, Zhu J, Zhang H, Wang X, You L, Xie J, Liu W, Wei X, Chen K, Li Z, Wei S, Geng X. PD-1 deficiency disrupts in vivo neural activity in mouse Hippocampus and cortex. Neurobiol Dis 2025; 213:107002. [PMID: 40516709 DOI: 10.1016/j.nbd.2025.107002] [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: 01/21/2025] [Revised: 06/11/2025] [Accepted: 06/11/2025] [Indexed: 06/16/2025] Open
Abstract
Programmed cell death 1 (PD-1), encoded by the Pdcd1 gene, was identified as a target in cancer immunotherapy but may result in the overactivation of T cell function and emotional changes such as anxiety. The dynamic changes in neuronal activity related to the anxious status caused by Pdcd1-/- remain unclear. In this study, we addressed these physiological issues by simultaneously recording neuronal activity (spikes) and local field potentials (LFPs) in the medial prefrontal cortex (mPFC) and hippocampal CA3 region using in vivo multi-channel electrodes. Our results demonstrate that PD-1 deficiency induces anxiety-like behaviours and extensive neuronal firing disorders in the mPFC and CA3 regions of mice. The key finding was that in pyramidal neuron and interneurons in the CA3 region, the in vivo firing and spike-LFP encoding was disordered in the opposite direction by Pdcd1-/-. These changes leaded to abnormal oscillations in mPFC and CA3 and disturbed mPFC neuronal firing. Targeting the activation of excitatory neurons in CA3 regions could rescue anxiety-like behaviours in Pdcd1-/- mice. This study provides physiological insights into the dynamic cooperation mechanisms between the mPFC and CA3 circuits in anxiety-like behaviours caused by Pdcd1-/- and other mental disorders associated with autoimmune problems.
Collapse
Affiliation(s)
- Minghui Hu
- Key Laboratory of Traditional Chinese Medicine Classical Theory, Ministry of Education, Shandong University of Traditional Chinese Medicine, Jinan, China; Shandong Provincial Engineering Research Center for the Prevention and Treatment of Major Brain Diseases with Traditional Chinese Medicine (PTMBD), Shandong University of Traditional Chinese Medicine, Jinan, China; Chinese Medicine and Brain Science Interdisciplinary Research Center (CMBS), Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Cui Lv
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Jianping Zhu
- College of Life Science, Shandong Normal University, Jinan, PR China
| | - Hao Zhang
- Key Laboratory of Traditional Chinese Medicine Classical Theory, Ministry of Education, Shandong University of Traditional Chinese Medicine, Jinan, China; Shandong Provincial Engineering Research Center for the Prevention and Treatment of Major Brain Diseases with Traditional Chinese Medicine (PTMBD), Shandong University of Traditional Chinese Medicine, Jinan, China; Chinese Medicine and Brain Science Interdisciplinary Research Center (CMBS), Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xinyu Wang
- Key Laboratory of Traditional Chinese Medicine Classical Theory, Ministry of Education, Shandong University of Traditional Chinese Medicine, Jinan, China; Shandong Provincial Engineering Research Center for the Prevention and Treatment of Major Brain Diseases with Traditional Chinese Medicine (PTMBD), Shandong University of Traditional Chinese Medicine, Jinan, China; Chinese Medicine and Brain Science Interdisciplinary Research Center (CMBS), Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Luyan You
- Chinese Medicine and Brain Science Interdisciplinary Research Center (CMBS), Shandong University of Traditional Chinese Medicine, Jinan, China; Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jinlu Xie
- Key Laboratory of Vector Biology and Pathogen Control of Zhejiang Province, School of Medicine, Huzhou University, Huzhou Central Hospital, Huzhou 313000, China
| | - Wei Liu
- Key Laboratory of Traditional Chinese Medicine Classical Theory, Ministry of Education, Shandong University of Traditional Chinese Medicine, Jinan, China; Shandong Provincial Engineering Research Center for the Prevention and Treatment of Major Brain Diseases with Traditional Chinese Medicine (PTMBD), Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xiaocen Wei
- Key Laboratory of New Material Research Institute, Institute of Pharmaceutical Research, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Kai Chen
- Shandong Provincial Engineering Research Center for the Prevention and Treatment of Major Brain Diseases with Traditional Chinese Medicine (PTMBD), Shandong University of Traditional Chinese Medicine, Jinan, China; Shandong Innovation Center of Engineered Bacteriophage Therapeutics, Shandong Academy of Pharmaceutical Sciences, Jinan, China
| | - Zifa Li
- Key Laboratory of Traditional Chinese Medicine Classical Theory, Ministry of Education, Shandong University of Traditional Chinese Medicine, Jinan, China; Shandong Provincial Engineering Research Center for the Prevention and Treatment of Major Brain Diseases with Traditional Chinese Medicine (PTMBD), Shandong University of Traditional Chinese Medicine, Jinan, China; Chinese Medicine and Brain Science Interdisciplinary Research Center (CMBS), Shandong University of Traditional Chinese Medicine, Jinan, China.
| | - Sheng Wei
- Key Laboratory of Traditional Chinese Medicine Classical Theory, Ministry of Education, Shandong University of Traditional Chinese Medicine, Jinan, China; Shandong Provincial Engineering Research Center for the Prevention and Treatment of Major Brain Diseases with Traditional Chinese Medicine (PTMBD), Shandong University of Traditional Chinese Medicine, Jinan, China; Chinese Medicine and Brain Science Interdisciplinary Research Center (CMBS), Shandong University of Traditional Chinese Medicine, Jinan, China.
| | - Xiwen Geng
- Key Laboratory of Traditional Chinese Medicine Classical Theory, Ministry of Education, Shandong University of Traditional Chinese Medicine, Jinan, China; Shandong Provincial Engineering Research Center for the Prevention and Treatment of Major Brain Diseases with Traditional Chinese Medicine (PTMBD), Shandong University of Traditional Chinese Medicine, Jinan, China; Chinese Medicine and Brain Science Interdisciplinary Research Center (CMBS), Shandong University of Traditional Chinese Medicine, Jinan, China.
| |
Collapse
|
3
|
Cheng K, Guo J, Li Y, Kang Q, Wang R, Luo L, Wang W, Lu J. Anti-Tumor Activities of Anti-Siglec-15 Chimeric Heavy-Chain Antibodies. Int J Mol Sci 2025; 26:5068. [PMID: 40507880 PMCID: PMC12154215 DOI: 10.3390/ijms26115068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2025] [Revised: 05/21/2025] [Accepted: 05/22/2025] [Indexed: 06/16/2025] Open
Abstract
Immune checkpoint inhibitors like programmed cell death 1 (PD-1) antibodies have revolutionized cancer treatment, but patient response rates remain limited. Sialic acid-binding Ig-like lectin 15 (Siglec-15) has emerged as a promising new immune checkpoint target. Through phage display technology using a Bactrian camel immunized with recombinant human Siglec-15, we generated six anti-Siglec-15 camelid nanobodies and constructed chimeric heavy-chain antibodies by fusing the VHH domains with human IgG-Fc. Following expression in HEK293-F cells and purification, three antibodies (S1, S5, S6) demonstrated specific binding to both human and murine Siglec-15 in ELISA and biolayer interferometry assays. In a xenograft model established by subcutaneous inoculation of NCI-H157-S15 cells into BALB/c nude mice, these antibodies showed distinct tumor targeting and significant blockade of Siglec-15 interactions with CD44, MAG, sialyl-Tn, and LRR4C ligands. All three antibodies exhibited anti-tumor effects, with S1 showing the most potent activity. S1-treated mice had significantly smaller tumor volumes and weights compared to controls. The S1, S5, and S6 treatment groups showed enhanced anti-tumor immunity, with reduced TGF-β, IL-6, and IL-10 levels. Notably, S1 treatment significantly increased tumor-associated macrophages in tumor tissues (p < 0.05). In conclusion, S1 exhibits remarkable anti-tumor activity and has the potential to be developed as a cancer immunotherapy targeting Siglec-15.
Collapse
Affiliation(s)
- Kexuan Cheng
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing 100081, China; (K.C.); (J.G.); (Y.L.); (Q.K.); (R.W.)
- College of Public Health, Zhengzhou University, Zhengzhou 450001, China
| | - Jiazheng Guo
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing 100081, China; (K.C.); (J.G.); (Y.L.); (Q.K.); (R.W.)
| | - Yating Li
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing 100081, China; (K.C.); (J.G.); (Y.L.); (Q.K.); (R.W.)
- College of Public Health, Zhengzhou University, Zhengzhou 450001, China
| | - Qinglin Kang
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing 100081, China; (K.C.); (J.G.); (Y.L.); (Q.K.); (R.W.)
| | - Rong Wang
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing 100081, China; (K.C.); (J.G.); (Y.L.); (Q.K.); (R.W.)
| | - Longlong Luo
- State Key Laboratory of Toxicology and Medical Countermeasures, Institute of Pharmacology and Toxicology, 27 Taiping Road, Beijing 100850, China;
| | - Wei Wang
- College of Public Health, Zhengzhou University, Zhengzhou 450001, China
| | - Jiansheng Lu
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing 100081, China; (K.C.); (J.G.); (Y.L.); (Q.K.); (R.W.)
| |
Collapse
|
4
|
You YS, Chang WT, Hsu CL, Wang HY, Lu YF, Kim I, Tzeng SJ. Wip1 inhibitor CCT007093 alleviates immune exhaustion of lymphocytes via p65 NF-κB and YY1 in chronic hepatitis B virus infection in mice. Front Immunol 2025; 16:1548814. [PMID: 40416970 PMCID: PMC12098592 DOI: 10.3389/fimmu.2025.1548814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Accepted: 03/31/2025] [Indexed: 05/27/2025] Open
Abstract
Introduction Prolonged viral infections often lead to lymphocyte exhaustion, marked by heightened inhibitory receptor expression like PD-1, compromising host defense mechanisms. The unexplored potential of chemical checkpoint inhibitors in rejuvenating immune responses prompted our investigation. Methods We focused on CCT007093, a Wip1 inhibitor, screened for its distinctive capacity to simultaneously decrease PD-1 and FcγRIIB expression in B cells. Results In this study, we harnessed a murine model of immune exhaustion induced by chronic hepatitis B virus (HBV) infection using hydrodynamic injection. Treatment with CCT007093 resulted in decreased levels of PD-1 expression, resulting in reduced percentages of PD-1+/hi CD4+ and CD8+ T cells in circulation, spleen, and liver. The expression levels of PD-1 and FcγRIIB, along with the percentages of PD-1+/hi and FcγRIIB+/hi CD19+ B cells in these tissues, were similarly diminished. Moreover, intrahepatic lymphocytes treated with CCT007093 displayed heightened responsiveness to ex vivo activation. Consequently, mice treated with CCT007093 exhibited significantly reduced serum HBsAg levels compared to vehicle-treated mice. Our detailed analyses, spanning promoter and transcriptome evaluations, uncovered p65 NF-κB as the primary activator of T cells and B cells, while Ying Yang 1 (YY1) emerged as the key regulator, orchestrating the down-regulation of PD-1 and FcγRIIB gene transcription in response to CCT007093. Discussion Our study highlights the prowess of chemical checkpoint inhibitors, exemplified by CCT007093, in alleviating immune exhaustion in HBV-infected mice, particularly by enhancing adaptive immunity.
Collapse
Affiliation(s)
- Yu-Syuan You
- Graduate Institute of Pharmacology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Wan-Ting Chang
- Graduate Institute of Pharmacology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Chia-Lang Hsu
- Department of Medical Research, National Taiwan University Hospital, Taipei, Taiwan
| | - Hui-Ying Wang
- Graduate Institute of Pharmacology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Yan-Fong Lu
- Graduate Institute of Pharmacology, College of Medicine, National Taiwan University, Taipei, Taiwan
- Department of Obstetrics and Gynecology, Shin-Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan
| | - InKyeom Kim
- Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
- BK21 Plus KNU Biomedical Convergence Program, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
- Cardiovascular Research Institute, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Shiang-Jong Tzeng
- Graduate Institute of Pharmacology, College of Medicine, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
5
|
Huang Y, Yoh K, Szamreta EA, Xu X, Hershman DL, Wright JD. Patterns of pembrolizumab use for recurrent cervical cancer. Int J Gynecol Cancer 2025; 35:101802. [PMID: 40300438 DOI: 10.1016/j.ijgc.2025.101802] [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: 10/23/2024] [Revised: 03/18/2025] [Accepted: 03/24/2025] [Indexed: 05/01/2025] Open
Abstract
OBJECTIVE This study aimed to examine the real-world use of pembrolizumab for recurrent cervical cancer as part of first-line or recurrence treatment, and its associated health care utilization. METHODS The Merative MarketScan Research Databases were used to identify newly diagnosed patients with cervical cancer who underwent primary hysterectomy or radiation from 2017 to 2022. Systemic therapy utilization, including pembrolizumab, was assessed at first recurrence. Health care utilization (hospitalization, emergency department visits, and costs) during first-line treatments for recurrence was described for patients treated with and without pembrolizumab. Multivariable regression models explored factors associated with pembrolizumab adoption and differences in health care utilization. RESULTS A total of 2727 patients were identified, including 1259 (46.2%) who underwent primary hysterectomy and 1468 (53.8%) who received primary radiotherapy. Chemotherapy for recurrence was initiated in 339 patients (12.4%). Recurrence treatment was noted in 9.7% of patients initially treated with hysterectomy and in 14.8% of those receiving primary radiotherapy. Among patients treated for recurrence, 24.8% received platinum alone, 52.5% a platinum-based combination therapy, and 22.7% non-platinum regimens. Forty-one patients (12.1%) received pembrolizumab. The median duration of first-line chemotherapy for recurrence was 2.3 months (interquartile range; 1.0-5.8) overall, and 4.3 months (interquartile range; 2.4-9.8) for patients treated with pembrolizumab. Pembrolizumab utilization was associated with more recent years of recurrence, advanced age, and prior chemotherapy. While pembrolizumab use was not associated with increased inpatient stays or emergency department visits, it was associated with significantly higher total costs and chemotherapy-related expenses. CONCLUSIONS Platinum-based chemotherapy is the predominant treatment for recurrent cervical cancer. Pembrolizumab utilization, although increasing, remains limited, highlighting a significant opportunity to optimize guideline-recommended therapies with proven efficacy in clinical trials.
Collapse
Affiliation(s)
- Yongmei Huang
- Columbia University Vagelos College of Physicians and Surgeons, Department of Obstetrics and Gynecology, New York, NY
| | - Katherine Yoh
- Columbia University Vagelos College of Physicians and Surgeons, Department of Obstetrics and Gynecology, New York, NY
| | | | - Xiao Xu
- Columbia University Vagelos College of Physicians and Surgeons, Department of Obstetrics and Gynecology, New York, NY; Columbia University Irving Medical Center, Herbert Irving Comprehensive Cancer Center, New York, NY
| | - Dawn L Hershman
- Columbia University Irving Medical Center, Herbert Irving Comprehensive Cancer Center, New York, NY; Columbia University Vagelos College of Physicians and Surgeons, Department of Medicine, New York, NY; Columbia University Mailman School of Public Health, Department of Epidemiology, New York, NY
| | - Jason D Wright
- Columbia University Vagelos College of Physicians and Surgeons, Department of Obstetrics and Gynecology, New York, NY; Columbia University Irving Medical Center, Herbert Irving Comprehensive Cancer Center, New York, NY.
| |
Collapse
|
6
|
Maul LV, Ramelyte E, Dummer R, Mangana J. Management of metastatic melanoma with combinations including PD-1 inhibitors. Expert Opin Biol Ther 2025; 25:1-12. [PMID: 40159098 DOI: 10.1080/14712598.2025.2485315] [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: 01/09/2025] [Accepted: 03/24/2025] [Indexed: 04/02/2025]
Abstract
INTRODUCTION Melanoma is among the most immunogenic malignancies. The advent of immune checkpoint inhibitors (ICIs) has revolutionized the landscape of melanoma treatment. Long-term durable cancer control is possible in nearly 50% of non-resectable, metastatic melanoma patients with anti-CTLA4 and anti-PD-1 antibodies. AREAS COVERED This review provides a critical overview of the current data and future research directions on the management of metastatic melanoma with ICIs. We reviewed the efficacy and safety of combinations with PD-1 inhibitors through PubMed database research (Nov 2024-Mar 2025). EXPERT OPINION A decade after ipilimumab's approval, challenges remain. To cure more patients, the development of combinations is warranted. Combinations with a limited number of ipilimumab applications improve the overall survival outcome by approximately 10%, with a dramatic increase in adverse events including fatal events. Anti-LAG3/nivolumab is a promising alternative, offering similar efficacy to ipilimumab/nivolumab with better tolerability. In our opinion, ipilimumab/nivolumab combination should be the first-line therapy for high-risk patients (high LDH, brain or liver metastasis), while nivolumab/relatlimab or PD-1 monotherapy may be preferable for lower-risk cases. However, treatment decisions are increasingly complex, since most patients nowadays are pretreated in the (neo)-adjuvant setting. The key limitation today is the lack of biomarkers to guide individualized treatment strategies.
Collapse
Affiliation(s)
- Lara Valeska Maul
- Department of Dermatology, University Hospital Zurich, Zurich, Switzerland
- Faculty of Medicine, University of Zurich, Zurich, Switzerland
| | - Egle Ramelyte
- Department of Dermatology, University Hospital Zurich, Zurich, Switzerland
- Faculty of Medicine, University of Zurich, Zurich, Switzerland
| | - Reinhard Dummer
- Department of Dermatology, University Hospital Zurich, Zurich, Switzerland
- Faculty of Medicine, University of Zurich, Zurich, Switzerland
| | - Joanna Mangana
- Department of Dermatology, University Hospital Zurich, Zurich, Switzerland
- Faculty of Medicine, University of Zurich, Zurich, Switzerland
| |
Collapse
|
7
|
Allam AA, Ahmed HA, Hassan MA, Khaled SAA, Shibl A, Osman AM, Ali NMR, Ahmed NM. Programmed Cell Death-1 and Programmed Cell Death Ligand-1 in Childhood Acute B-Lymphoblastic Leukemia: Expression and Significance as Biomarker. Int J Lab Hematol 2025. [PMID: 40166836 DOI: 10.1111/ijlh.14472] [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: 10/13/2024] [Revised: 02/20/2025] [Accepted: 03/22/2025] [Indexed: 04/02/2025]
Abstract
INTRODUCTION This study aimed to assess programmed death-1 (PD-1) and programmed death ligand-1 (PDL-1) expression in newly diagnosed pediatric cases of acute B-lymphoblastic leukemia (B-ALL) and at 6 months of treatment and to explore their value as biomarkers. METHODS Fifty newly diagnosed B-ALL patients and 30 controls were recruited. Bone marrow samples or peripheral blood were obtained from children at diagnosis and 6 months after cytotoxic therapy. Flow cytometric analysis of obtained samples was done and the PD-1, PDL-1, and CD3 (cluster of differentiation) expressions were recorded. RESULTS Percentages of PD-1, PDL-1, and CD3 in the control and B-ALL groups at initial presentation were 7.9% ± 2.8% vs. 16.45% ± 7.7% (p = 0.023), 8.6% ± 3.4% vs. 19.05% ± 13.7% (p < 0.001), and 30.8% ± 1.2% vs. 11.05% ± 7.3% (p < 0.001), respectively. CD3 expression increased significantly at 6 months; PD-1 and PDL-1 expression showed insignificant decrease from initial presentation. There was a negative correlation between PD-1 and HB level (p = 0.03) and a positive correlation between PD-1 and PDL-1 at 6 months of treatment (p = 0.002). Remission rates increased significantly with the decrease of PD-1and PDL-1. CONCLUSION Initially, PD-1 and PDL-1 were higher in patients than in controls and decreased 6 months after treatment. PD-1 and PDL-1 expression was associated with increased remission rates, implicating that modulation of PD-1 and PDL-1 expression may be a therapeutic approach for B-ALL. Moreover, this study created a new method for the assessment of PD-1 and PDL-1 in B-ALL. CLINICAL TRIAL Trial Registration: NCT05428111.
Collapse
Affiliation(s)
- Ahmed Ahmed Allam
- Department of Clinical Pathology, Sohag University Faculty of Medicine, Sohag, Egypt
| | - Heba A Ahmed
- Department of Clinical Pathology, Sohag University Faculty of Medicine, Sohag, Egypt
| | | | - Safaa A A Khaled
- Department of Internal Medicine Haematology and BMT Unit Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Azza Shibl
- Assiut University South Egypt Cancer Institute, Assiut, Egypt
| | | | | | - Nesma Mokhtar Ahmed
- Department of Clinical Pathology, Sohag University Faculty of Medicine, Sohag, Egypt
| |
Collapse
|
8
|
Huang J, Min S, Hong R, Zou M, Zhou D. High-dose Vitamin C inhibits PD-L1 expression by activating AMPK in colorectal cancer. Immunobiology 2025; 230:152893. [PMID: 40139125 DOI: 10.1016/j.imbio.2025.152893] [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: 11/06/2024] [Revised: 02/27/2025] [Accepted: 03/17/2025] [Indexed: 03/29/2025]
Abstract
Vitamin C (VitC) has elicited considerable interest regarding its potential role in cancer therapy; however, its effects on tumor immunity remain unclear. In colorectal cancer (CRC), although anti-PD-1/PD-L1 therapies demonstrate promise, their efficacy is still constrained. Our prior research demonstrated that VitC can inhibit tumor growth by suppressing the Warburg effect. This study aims to explore the effects of high-dose VitC on PD-L1 expression in CRC, focusing on its underlying mechanisms and potential for enhancing immunotherapy. We found that VitC inhibits aerobic glycolysis in HCT116 cells while also downregulating PD-L1 expression. Further investigations indicated that this process is mediated by VitC's activation of AMPK, which downregulates HK2 and NF-κB, ultimately resulting in reduced PD-L1 expression and increased T cell infiltration. Notably, we observed that VitC and the PD-L1 monoclonal antibody atezolizumab exhibit comparable tumor-inhibiting abilities, and their combined use further enhances this efficacy. In conclusion, our results demonstrate that high-dose VitC activates AMPK, downregulates PD-L1 expression, mitigates immune evasion, and suppresses tumor growth. This provides a promising strategy for optimizing immunotherapy in CRC.
Collapse
Affiliation(s)
- Jia Huang
- Department of Anesthesiology, Technology Innovation Research Center, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Su Min
- Department of Anesthesiology, Technology Innovation Research Center, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China.
| | - Ruiyang Hong
- Department of Anesthesiology, Technology Innovation Research Center, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Mou Zou
- Department of Anesthesiology, Technology Innovation Research Center, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Dongyu Zhou
- Department of Anesthesiology, Technology Innovation Research Center, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| |
Collapse
|
9
|
Wen N, Lu Y, Zhuo Y, Fu B, Wang H, He Y, Wu H, Wang Z, Tan W, Qiu L. Enhancing T-Cell Infiltration and Immunity in Solid Tumors via DNA Nanolinker-Mediated Monocyte Hitchhiking. J Am Chem Soc 2025; 147:9800-9809. [PMID: 40042588 DOI: 10.1021/jacs.4c18455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/20/2025]
Abstract
Cytotoxic CD8+ T cells are one of the most powerful effectors in the antitumor immune response. However, their insufficient tumor infiltration severely limits the clinical success of immunotherapy in solid tumors. In this work, by using amphiphilic aptamer-incorporated DNA tetrahedra (aptTDN) as the intercellular nanolinker, we developed a monocyte-hitchhiked T-cell delivery strategy to actively promote the intratumoral infiltration of effector CD8+ T cells. Our results demonstrated that membrane-anchoring of aptTDN enabled the specific and stable ligation of T cells with Ly6c+ monocytes, without compromising the migratory behavior of monocytes and the antitumor activity of T cells. By leveraging the intrinsic tumor-homing capability of monocytes, the ligated T cells efficiently accumulated within tumor-associated vasculature and then deeply infiltrated the tumor bed. Additionally, the enhanced intratumoral presence of adoptively transferred effector CD8+ T cells facilitated the establishment of an immunosupportive microenvironment, that further recruited endogenous T cells and ultimately bolstered antitumor immunity. Moreover, our monocyte-hitchhiked T-cell tumor infiltration system could significantly improve the efficacy of immune checkpoint blockade therapy. Collectively, by utilizing chemically synthetic nanolinkers to modulate cellular interactions and develop a delivery system of therapeutic cells, our work presents a new paradigm for the advancement of immunotherapy against solid tumors.
Collapse
Affiliation(s)
- Nachuan Wen
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan 410082, China
- The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Yao Lu
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan 410082, China
- The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Yuting Zhuo
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan 410082, China
- The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Bo Fu
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan 410082, China
| | - Haiyuan Wang
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan 410082, China
| | - Yao He
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan 410082, China
| | - Hui Wu
- The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Zhimin Wang
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan 410082, China
| | - Weihong Tan
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan 410082, China
- The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Liping Qiu
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan 410082, China
- The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| |
Collapse
|
10
|
Xu Q, Li L, Zhu R. T Cell Exhaustion in Allergic Diseases and Allergen Immunotherapy: A Novel Biomarker? Curr Allergy Asthma Rep 2025; 25:18. [PMID: 40091122 DOI: 10.1007/s11882-025-01199-5] [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] [Accepted: 03/06/2025] [Indexed: 03/19/2025]
Abstract
PURPOSE OF REVIEW This review explores the emerging role of T cell exhaustion in allergic diseases and allergen immunotherapy (AIT). It aims to synthesize current knowledge on the mechanisms of T cell exhaustion, evaluate its potential involvement in allergic inflammation, and assess its implications as a novel biomarker for predicting and monitoring AIT efficacy. RECENT FINDINGS Recent studies highlight that T cell exhaustion, characterized by co-expression of inhibitory receptors (e.g., PD-1, CTLA-4, TIM-3), diminished cytokine production, and altered transcriptional profiles, may suppress type 2 inflammation in allergic diseases. In allergic asthma, exhausted CD4 + T cells exhibit upregulated inhibitory receptors, correlating with reduced IgE levels and airway hyperreactivity. During AIT, prolonged high-dose allergen exposure drives allergen-specific Th2 and T follicular helper (Tfh) cell exhaustion, potentially contributing to immune tolerance. Notably, clinical improvements in AIT correlate with depletion of allergen-specific Th2 cells and persistent expression of exhaustion markers (e.g., PD-1, CTLA-4) during maintenance phases. Blockade of inhibitory receptors (e.g., PD-1) enhances T cell activation, underscoring their dual regulatory role in allergy. T cell exhaustion represents a double-edged sword in allergy: it may dampen pathological inflammation in allergic diseases while serving as a mechanism for AIT-induced tolerance. The co-expression of inhibitory receptors on allergen-specific T cells emerges as a promising biomarker for AIT efficacy. Future research should clarify the transcriptional and metabolic drivers of exhaustion in allergy, validate its role across diverse allergic conditions, and optimize strategies to harness T cell exhaustion for durable immune tolerance. These insights could revolutionize therapeutic approaches and biomarker development in allergy management.
Collapse
Affiliation(s)
- Qingxiu Xu
- Department of Allergy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Le Li
- Department of Allergy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Rongfei Zhu
- Department of Allergy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
- Institute of Allergy and Clinical Immunology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
11
|
Chang E, Cavallo K, Behar SM. CD4 T cell dysfunction is associated with bacterial recrudescence during chronic tuberculosis. Nat Commun 2025; 16:2636. [PMID: 40097414 PMCID: PMC11914476 DOI: 10.1038/s41467-025-57819-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Accepted: 03/04/2025] [Indexed: 03/19/2025] Open
Abstract
While most people contain Mycobacterium tuberculosis infection, some individuals develop active disease, usually within two years of infection. Why immunity fails after initially controlling infection is unknown. C57BL/6 mice control Mycobacterium tuberculosis for up to a year but ultimately succumb to disease. We hypothesize that the development of CD4 T cell dysfunction permits bacterial recrudescence. We developed a reductionist model to assess antigen-specific T cells during chronic infection and found evidence of CD4 T cell senescence and exhaustion. In C57BL/6 mice, CD4 T cells upregulate coinhibitory receptors and lose effector cytokine production. Single cell RNAseq shows that only a small number of CD4 T cells in the lungs of chronically infected mice are polyfunctional. While the origin and causal relationship between T-cell dysfunction and recrudescence remains uncertain, we propose T cell dysfunction leads to a feed-forward loop that causes increased bacillary numbers, greater T cell dysfunction, and progressive disease.
Collapse
Affiliation(s)
- Evelyn Chang
- Immunology and Microbiology Program, Morningside Graduate School of Biomedical Sciences, Worcester, MA, USA
- Department of Microbiology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Kelly Cavallo
- Department of Microbiology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Samuel M Behar
- Immunology and Microbiology Program, Morningside Graduate School of Biomedical Sciences, Worcester, MA, USA.
- Department of Microbiology, University of Massachusetts Chan Medical School, Worcester, MA, USA.
| |
Collapse
|
12
|
Carr AS, Vonberg FW, Koay S, Young K, Shaw H, Olsson-Brown A, Willis M. Neurological complications of immune checkpoint inhibitors: a practical guide. Pract Neurol 2025; 25:116-126. [PMID: 39592208 DOI: 10.1136/pn-2024-004327] [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] [Accepted: 10/26/2024] [Indexed: 11/28/2024]
Abstract
Immune checkpoint inhibition unleashes the power of the immune system against tumour cells. Immune checkpoint inhibitors (ICIs) block the inhibitory effects of cytotoxic T-lymphocyte associated protein 4 (CTLA-4), programmed death protein 1 (PD-1), programmed death ligand 1 (PD-L1) and lymphocyte activation gene 3 (LAG-3) molecules on T-cells, and so enhance physiological cytotoxic effects. ICIs can significantly improve survival from cancers, including those previously associated with poor treatment response, such as metastatic melanoma. However, on-target off-tumour effects of ICIs result in immune-related adverse events. These toxicities are common and require new multidisciplinary expertise to manage. ICI neurotoxicity is relatively rare but ominous due to its severity, heterogenous manifestations and potential for long-term disability. Neurotoxic syndromes are novel and often present precipitously. Here, we describe ICI mechanisms of action, their impact on cancer outcomes and their frequency of immune-related adverse events. We focus particularly on neurotoxicity. We discuss the current appreciation of neurotoxic syndromes, management strategies and outcomes based on clinical expertise and consensus, multi-specialty guidance. The use of immunotherapy is expanding exponentially across multiple cancer types and so too will our approach to these cases.
Collapse
Affiliation(s)
- Aisling S Carr
- Centre for Neuromuscular Diseases, National Hospital for Neurology and Neurosurgery, London, UK
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, UK
| | - Frederick William Vonberg
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, UK
- Neurology, National Hospital for Neurology and Neurosurgery, London, UK
| | - Shiwen Koay
- Centre for Neuromuscular Diseases, National Hospital for Neurology and Neurosurgery, London, UK
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, UK
| | - Kate Young
- Renal and Melanoma Unit, Royal Marsden Hospital Chelsea, London, London, UK
| | - Heather Shaw
- Department of Oncology, University College London Hospitals NHS Foundation Trust, London, London, UK
| | - Anna Olsson-Brown
- Sussex Cancer Centre, University Hospitals Sussex NHS Foundation Trust, Brighton, UK
- Department of Clinical and Molecular Pharmacology, University of Liverpool, Liverpool, UK
| | - Mark Willis
- Department of Neurology, University Hospital of Wales, Cardiff, UK
| |
Collapse
|
13
|
Hui X, Xue M, Ren Y, Chen Y, Chen X, Farooq MA, Ji Y, Zhan W, Huang Y, Du B, Yao J, Duan Y, Jiang W. GPR132 regulates the function of NK cells through the Gαs/CSK/ZAP70/NF-κB signaling pathway as a potential immune checkpoint. SCIENCE ADVANCES 2025; 11:eadr9395. [PMID: 40043109 PMCID: PMC11881902 DOI: 10.1126/sciadv.adr9395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 01/30/2025] [Indexed: 05/13/2025]
Abstract
As a member of the proton-sensing GPCR receptors, GPR132 plays a crucial role in regulating immune cell functions, but the mechanism by which GPR132 affects natural killer (NK) cells has not yet been reported. Here, RNA-seq displayed that the expression of GPR132 was reduced in activated NK cells, and the proportion of mature NK cells in GPR132-/- mice was substantially increased compared to WT mice, with stronger anti-melanoma capabilities. Further investigation indicates that GPR132-deficient NK92 cells expressed more GzmB and IFN-γ and exhibited stronger cytotoxicity. Mechanically, GPR132 regulates NK cell function through the CSK/ZAP70/NF-κB signaling axis. Down-regulation of GPR132 weakens the inhibition of NK cell function by lactate, thereby enhancing the functional execution of CAR-NK cells against colorectal cancer. These results highlight the previously unrecognized role of GPR132 in the regulation of NK cell function and that inhibition of GPR132 provided an updated insight for NK cell therapy.
Collapse
Affiliation(s)
- Xinhui Hui
- Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai, China
| | - Min Xue
- Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai, China
| | - Yaojun Ren
- Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai, China
- College of Life Science, Xinjiang Normal University, Urumqi, China
| | - Yiran Chen
- Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai, China
| | - Xuenan Chen
- Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai, China
| | - Muhammad Asad Farooq
- The First Dongguan Affiliated Hospital, Guangdong Provincial Key Laboratory of Medical Immunology and Molecular Diagnostics, Guangdong Medical University, Dongguan, China
| | - Yuzhou Ji
- Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai, China
| | - Weirong Zhan
- Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai, China
| | - Yunhe Huang
- Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai, China
| | - Bingtan Du
- Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai, China
| | - Jie Yao
- Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai, China
| | - Yixin Duan
- Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai, China
| | - Wenzheng Jiang
- Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai, China
| |
Collapse
|
14
|
Zhang S, Wang HY, Tao X, Chen Z, Levental I, Lin X. Palmitoylation of PD-L1 Regulates Its Membrane Orientation and Immune Evasion. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2025; 41:5170-5178. [PMID: 39965093 DOI: 10.1021/acs.langmuir.4c04441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/20/2025]
Abstract
Recently identified palmitoylation of PD-L1 is essential for immune regulation. To elucidate the underlying molecular mechanism, we performed giant plasma membrane vesicle (GPMV) experiments, μs-scale all-atom molecular dynamics (MD) simulations, fluorescence resonance energy transfer (FRET) experiments, and immune killing experiments. GPMV experiments indicated that PD-L1 palmitoylation enhanced its lipid raft affinity. MD simulations revealed dramatically different membrane orientation states of PD-L1 in liquid-ordered (Lo, lipid raft) compared to liquid-disordered (Ld, nonraft) membrane environments, which was validated by FRET experiments. The Ld region promoted the "lie-down" orientation of PD-L1, which could inhibit its association with the PD-1 protein on immune cells and thus promote the immune killing of cancer cells. This hypothesis was supported by immune killing experiments using γδT cells as effector cells and NCI-H1299 lung cancer cells as target cells. In short, our study demonstrates that the palmitoylation affects PD-L1's membrane localization and then membrane orientation, which thus regulates its binding with T cell PD-1 and the immune regulation. These observations may guide therapeutic strategies by explicating the regulation of immune checkpoint proteins by post-translational modifications and membrane environments.
Collapse
Affiliation(s)
- Siya Zhang
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Engineering Medicine, Beihang University, Beijing 100191, China
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Beijing 100005, China
| | - Hong-Yin Wang
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, Virginia 22903, United States
| | - Xuan Tao
- State Key Laboratory of Membrane Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Zhongwen Chen
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 201210, China
| | - Ilya Levental
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, Virginia 22903, United States
| | - Xubo Lin
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Engineering Medicine, Beihang University, Beijing 100191, China
| |
Collapse
|
15
|
Luca VC. LAG Time in the Era of Immunotherapy-New Molecular Insights Into the Immunosuppression Mechanism of Lymphocyte Activation Gene-3. Immunol Rev 2025; 330:e70002. [PMID: 39887765 PMCID: PMC11917464 DOI: 10.1111/imr.70002] [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: 01/09/2025] [Accepted: 01/14/2025] [Indexed: 02/01/2025]
Abstract
The immune checkpoint receptor lymphocyte activation gene-3 (LAG3) inhibits T-cell activation and was recently validated as a target for cancer immunotherapy. Despite its emergence as a therapeutic target, a lack of molecular-level insight has obscured our understanding of the LAG3 immunosuppression mechanism. This review highlights a series of breakthroughs that have illuminated fundamental aspects of LAG3 molecular biology. Key discoveries include structural insights into LAG3 interactions with ligands and antibodies, mechanistic studies of LAG3 interference with T-cell receptor (TCR) signaling, and the development of novel therapeutics. A particular focus is placed on structure-function relationships for LAG3-targeting drugs, as it has become apparent that several distinct approaches to LAG3 antagonism are viable. In addition to LAG3 antagonists, agonistic LAG3 antibodies and immunostimulatory LAG3 extracellular domains (ECDs) are discussed in the context of current structural and mechanistic data. Collectively, these findings should provide an updated landscape for the design of optimal LAG3-based therapeutics for cancer and autoimmune diseases.
Collapse
Affiliation(s)
- Vincent C. Luca
- Moffitt Cancer Center, Department of Immunology. Tampa, FL 33612
| |
Collapse
|
16
|
Zhao H, Huang S, Wu J, Lu Y, Zou Y, Zeng H, Li C, Wang J, Zhang X, Duan S, Liang W. Efficacy and safety of first-line PD-1/PD-L1 inhibitor in combination with CTLA-4 inhibitor in the treatment of patients with advanced non-small cell lung cancer: a systemic review and meta-analysis. Front Immunol 2025; 16:1515027. [PMID: 39981238 PMCID: PMC11839650 DOI: 10.3389/fimmu.2025.1515027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Accepted: 01/20/2025] [Indexed: 02/22/2025] Open
Abstract
Introduction The combination of PD-1/PD-L1 inhibitor with CTLA-4 inhibitor for advanced non-small cell lung cancer(NSCLC) is presently a significant area of research, however its clinical application remains contentious. This meta-analysis aimed to assess the efficacy and safety of first-line PD-1/PD-L1 inhibitor in combination with CTLA-4 inhibitor (CP) in the treatment of patients with advanced NSCLC. Methods A systemic search was conducted in four databases (PubMed, Cochrane library, Embase, and Web of Science) from their establishment until January 17, 2024, for randomized controlled trials that investigated the use of the first-line PD-1/PD-L1 inhibitor plus CTLA-4 inhibitor in patients with advanced NSCLC. Progression-free survival (PFS), overall survival (OS), objective response rate (ORR), disease control rate (DCR), and adverse events (AEs) were subjected to meta-analyses. Results Totally 7 eligible randomized controlled trials including 4682 people were included. Two comparative analyses were performed: CP versus chemotherapy, CP versus PD-1/PD-L1 inhibitor (P). Compared with the chemotherapy group, CP improved OS (HR: 0.84, 95% CI: 0.75-0.94, p<0.05) but not PFS (HR: 0.94, 95%CI: 0.73-1.20, p = 0.63) or ORR (OR: 1.16, 95% CI: 0.79-1.71, p = 0.45). In terms of toxicity, CP had slightly fewer any AEs compared to chemotherapy (RR: 0.94, 95% CI: 0.91-0.97; p<0.05). Compared to the P group, there was no significant difference in OS (MD: -0,25, 95% CI: -2.47-1.98, p = 0.83), PFS (MD: -0.91, 95% CI: -3.19-1.36, p = 0.43), and ORR (OR:1.05, 95% CI. 0.80-1.36, p = 0.73). Subgroup analysis revealed that CP provided superior OS compared with P in patients with PD-L1 expression < 1%. Conclusion CP was a feasible and safe first-line therapy for patients with advanced NSCLC. Specifically, CP may function as a therapeutic alternative for individuals with low or negative PD-L1 expression, resulting in enhanced long-term outcomes compared to chemotherapy or P. Further randomized controlled trials with prolonged follow-up periods are necessary to validate these results, particularly focusing on efficacy in patients with differing PD-L1 expression levels, to improve the stratified implementation of immunotherapy. Systematic review registration https://www.crd.york.ac.uk/prospero/display_record.php?ID=CRD42024621116, identifier CRD42024621116.
Collapse
Affiliation(s)
- Huimin Zhao
- The First Affiliated Hospital of Guangxi University of Science and Technology, Guangxi University of Science and Technology, Liuzhou, Guangxi, China
| | - Shanshan Huang
- The First Affiliated Hospital of Guangxi University of Science and Technology, Guangxi University of Science and Technology, Liuzhou, Guangxi, China
| | - Jianyu Wu
- The First Affiliated Hospital of Guangxi University of Science and Technology, Guangxi University of Science and Technology, Liuzhou, Guangxi, China
| | - Yanlan Lu
- The First Affiliated Hospital of Guangxi University of Science and Technology, Guangxi University of Science and Technology, Liuzhou, Guangxi, China
| | - Yue Zou
- The First Affiliated Hospital of Guangxi University of Science and Technology, Guangxi University of Science and Technology, Liuzhou, Guangxi, China
| | - Haijian Zeng
- The First Affiliated Hospital of Guangxi University of Science and Technology, Guangxi University of Science and Technology, Liuzhou, Guangxi, China
| | - Chunlan Li
- The First Affiliated Hospital of Guangxi University of Science and Technology, Guangxi University of Science and Technology, Liuzhou, Guangxi, China
| | - Jin Wang
- The First Affiliated Hospital of Guangxi University of Science and Technology, Guangxi University of Science and Technology, Liuzhou, Guangxi, China
| | - Xiaochen Zhang
- The First Affiliated Hospital of Guangxi University of Science and Technology, Guangxi University of Science and Technology, Liuzhou, Guangxi, China
- Medicine College, Guangxi University of Science and Technology, Liuzhou, Guangxi, China
| | - Siliang Duan
- The First Affiliated Hospital of Guangxi University of Science and Technology, Guangxi University of Science and Technology, Liuzhou, Guangxi, China
- Medicine College, Guangxi University of Science and Technology, Liuzhou, Guangxi, China
| | - Weiming Liang
- The First Affiliated Hospital of Guangxi University of Science and Technology, Guangxi University of Science and Technology, Liuzhou, Guangxi, China
| |
Collapse
|
17
|
Sun Q, Hong S. Glycoscience in Advancing PD-1/PD-L1-Axis-Targeted Tumor Immunotherapy. Int J Mol Sci 2025; 26:1238. [PMID: 39941004 PMCID: PMC11818636 DOI: 10.3390/ijms26031238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 01/23/2025] [Accepted: 01/28/2025] [Indexed: 02/16/2025] Open
Abstract
Immune checkpoint blockade therapy, represented by anti-PD-1/PD-L1 monoclonal antibodies, has significantly changed the immunotherapy landscape. However, the treatment is still limited by unsatisfactory response rates, immune-related adverse effects, and drug resistance. Current studies have established that glycosylation, a common post-translational modification, is crucial in promoting cancer progression and immune invasion. Targeting aberrant glycosylation in cancers presents precision medicine regimens for monitoring cancer progression and developing personalized medicine. Notably, the immune checkpoints PD-1 and PD-L1 are highly glycosylated, which affects PD-1/PD-L1 interaction and the binding of anti-PD-1/PD-L1 monoclonal antibodies. Recent achievements in glycoscience to enhance patient outcomes, referred to as glycotherapy, have underscored their high potency in advancing PD-1/PD-L1 blockade therapies, i.e., glycoengineered antibodies with improved binding toward PD-1/PD-L1, pharmaceutic inhibitors for core fucosylation and sialylation, and synergistic treatment with the antibody-sialidase conjugate. This review briefly introduces the PD-1/PD-L1 axis and glycosylation and highlights the fundamental and applied advances in glycoscience that improve PD-1/PD-L1 immunoblockade therapies.
Collapse
Affiliation(s)
| | - Senlian Hong
- State Key Laboratory of Natural and Biomimetic Drugs, Chemical Biology Center, Department of Chemical Biology, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China;
| |
Collapse
|
18
|
Chang E, Cavallo K, Behar SM. CD4 T cell dysfunction is associated with bacterial recrudescence during chronic tuberculosis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.22.634376. [PMID: 39896548 PMCID: PMC11785196 DOI: 10.1101/2025.01.22.634376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
While most people contain Mycobacterium tuberculosis infection, some individuals develop active disease, usually within two years of infection. Why immunity fails after initially controlling infection is unknown. C57BL/6 mice control Mycobacterium tuberculosis for up to a year but ultimately succumb to disease. We hypothesize that the development of CD4 T cell dysfunction permits bacterial recrudescence. We developed a reductionist model to assess antigen-specific T cells during chronic infection and found evidence of CD4 T cell senescence and exhaustion. In C57BL/6 mice, CD4 T cells upregulate coinhibitory receptors and lose effector cytokine production. Single cell RNAseq shows that only a small number of CD4 T cells in the lungs of chronically infected mice are polyfunctional. While the origin and causal relationship between T-cell dysfunction and recrudescence remains uncertain, we propose T cell dysfunction leads to a feed-forward loop that causes increased bacillary numbers, greater T cell dysfunction, and progressive disease.
Collapse
Affiliation(s)
- Evelyn Chang
- Immunology and Microbiology Program, Graduate School of Biomedical Science, Worcester, Massachusetts, USA
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Kelly Cavallo
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Samuel M. Behar
- Immunology and Microbiology Program, Graduate School of Biomedical Science, Worcester, Massachusetts, USA
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| |
Collapse
|
19
|
Zhang H, Zheng H, Wang Y, Chen C, Tong Y, Xie S, Ma X, Guo L, Lu R. PD-1 suppresses human CD38 + circulating Tfr cells and regulates humoral immunity. J Immunother Cancer 2025; 13:e010026. [PMID: 39800377 PMCID: PMC11748770 DOI: 10.1136/jitc-2024-010026] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Accepted: 12/15/2024] [Indexed: 01/23/2025] Open
Abstract
BACKGROUND Anti-programmed cell death protein 1 (anti-PD-1) antibodies have achieved revolutionary success in cancer therapy. However, the impact of anti-PD-1 therapy on host humoral immunity in humans during cancer immunotherapy requires further investigation. METHODS We evaluated immunoglobulin titers by ELISA and screened the immune landscape of immune cells from 25 healthy donors and 50 cases including 25 new-onset hepatocellular carcinoma (HCC) patients prior to systemic treatment and 25 HCC patients undergoing anti-PD-1 therapy by multicolor flow cytometry. Flow or beads sorted cells were cultured ex vivo for proliferation and functional analysis. RESULTS Anti-PD-1 therapy significantly increased the levels of IgG and IgA in the periphery of HCC patients. Anti-PD-1 treatment led to an increase in plasmablasts and a notable rise in circulating T follicular regulatory (cTfr) cells, while changes in circulating B cells, T follicular helper cells, or regulatory T cells were not significant. Anti-PD-1 therapy also influenced the proliferation and function of cTfr cells, promoting the differentiation of CD38+ cTfr cells. We observed that the CD38+ Tfr cell subset in the peripheral blood can promote plasmablast differentiation, associated with altered antibody production. CONCLUSIONS Together, these data demonstrate the immunomodulatory role of PD-1 in restricting the differentiation and function of human cTfr cells and in regulating humoral immunity.
Collapse
Affiliation(s)
- Heng Zhang
- Department of Clinical Laboratory, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Hui Zheng
- Department of Clinical Laboratory, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Yanchun Wang
- Department of Clinical Laboratory, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Cuncun Chen
- Department of Clinical Laboratory, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Ying Tong
- Department of Clinical Laboratory, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Suhong Xie
- Department of Clinical Laboratory, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Xiaolu Ma
- Department of Clinical Laboratory, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Lin Guo
- Department of Clinical Laboratory, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical School,Fudan University, Shanghai, China
| | - Renquan Lu
- Department of Clinical Laboratory, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical School,Fudan University, Shanghai, China
| |
Collapse
|
20
|
Long J, Yang S, Bian Z, Zhu H, Ma M, Wang X, Li L, Zhang W, Han Y, Gershwin ME, Lian Z, Zhao Z. PD-1 +CD8 + T Cell-Mediated Hepatocyte Pyroptosis Promotes Progression of Murine Autoimmune Liver Disease. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2407284. [PMID: 39494472 PMCID: PMC11714232 DOI: 10.1002/advs.202407284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 10/10/2024] [Indexed: 11/05/2024]
Abstract
The specific mechanisms underlying effector pathways in autoimmune liver disease remain enigmatic and therefore constructing appropriate murine models to investigate disease pathogenesis becomes critical. A spontaneous severe murine model of autoimmune liver disease has been previously established in dnTGFβRII Aire-/- mice, exhibiting disease phenotypes that resemble both human primary biliary cholangitis (PBC) and autoimmune hepatitis (AIH). The data suggests that auto-reactive liver-specific CD8+ T cells are the primary pathogenic cells in liver injury. In this study, these data are advanced through the use of both single-cell sequencing and extensive in vitro analysis. The results identify a specific expanded pathogenic subset of PD-1+CD8+ T cells in the liver, exhibiting strong functional activity and cytotoxicity against target cells. Depletion of PD-1+CD8+ T cells using CAR-T cells effectively alleviates the disease. GSDMD-mediated pyroptosis is found to be aberrantly activated in the livers of model mice, and treatment with a GSDMD-specific inhibitor significantly inhibits disease progression. In vitro experiments reveal that PD-1+CD8+ T cells can induce the pyroptosis of hepatocytes through elevated production of granzyme B and perforin-1. These results provide a novel explanation for the cytotoxic activity of pathogenic liver PD-1+CD8+ T cells in autoimmune liver diseases and offer potential therapeutic targets.
Collapse
Affiliation(s)
- Jie Long
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhou510080China
| | - Si‐Yu Yang
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhou510080China
| | - Zhen‐Hua Bian
- School of Biomedical Sciences and EngineeringSouth China University of TechnologyGuangzhou International CampusGuangzhou511442China
| | - Hao‐Xian Zhu
- School of MedicineSouth China University of TechnologyGuangzhou510006China
- Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhou510080China
| | - Min Ma
- School of MedicineSouth China University of TechnologyGuangzhou510006China
- Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhou510080China
| | - Xiao‐Qing Wang
- School of MedicineSouth China University of TechnologyGuangzhou510006China
| | - Liang Li
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhou510080China
| | - Weici Zhang
- Division of Rheumatology, Allergy and Clinical ImmunologyUniversity of California DavisDavisCA95616USA
| | - Ying Han
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive DiseasesAir Force Military Medical UniversityXi'an710000China
| | - M. Eric Gershwin
- Division of Rheumatology, Allergy and Clinical ImmunologyUniversity of California DavisDavisCA95616USA
| | - Zhe‐Xiong Lian
- Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhou510080China
| | - Zhi‐Bin Zhao
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhou510080China
| |
Collapse
|
21
|
Oura K, Morishita A, Tadokoro T, Fujita K, Tani J, Kobara H. Immune Microenvironment and the Effect of Vascular Endothelial Growth Factor Inhibition in Hepatocellular Carcinoma. Int J Mol Sci 2024; 25:13590. [PMID: 39769351 PMCID: PMC11679663 DOI: 10.3390/ijms252413590] [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: 11/04/2024] [Revised: 12/12/2024] [Accepted: 12/17/2024] [Indexed: 01/03/2025] Open
Abstract
Systemic therapy for unresectable hepatocellular carcinoma (HCC) has progressed with the development of multiple kinases, such as vascular endothelial growth factor (VEGF) signaling, targeting cancer growth and angiogenesis. Additionally, the efficacy of sorafenib, regorafenib, lenvatinib, ramucirumab, and cabozantinib has been demonstrated in various clinical trials, and they are now widely used in clinical practice. Furthermore, the development of effective immune checkpoint inhibitors has progressed in systemic therapy for unresectable HCC, and atezolizumab + bevacizumab (atezo/bev) therapy and durvalumab + tremelimumab therapy are now recommended as first-line treatment. Atezo/bev therapy, which combines an anti-programmed cell death 1 ligand 1 antibody with an anti-VEGF antibody, is the first cancer immunotherapy to demonstrate efficacy against unresectable HCC. With the increasing popularity of these treatments, VEGF inhibition is attracting attention from the perspective of its anti-angiogenic effects and impact on the cancer-immune cycle. In this review, we outline the role of VEGF in the tumor immune microenvironment and cancer immune cycle in HCC and outline the potential immune regulatory mechanisms of VEGF. Furthermore, we consider the potential significance of the dual inhibition of angiogenesis and immune-related molecules by VEGF, and ultimately aim to clarify the latest treatment strategies that maximizes efficacy.
Collapse
Affiliation(s)
| | - Asahiro Morishita
- Department of Gastroenterology and Neurology, Faculty of Medicine, Kagawa University, 1750-1 Ikenobe, Miki, Kita 761-0793, Kagawa, Japan; (K.O.)
| | | | | | | | | |
Collapse
|
22
|
Ding R, Lu J, Huang X, Deng M, Wei H, Jiang G, Zhu H, Yuan H. The effect of immunotherapy PD-1 blockade on acute bone cancer pain: Insights from transcriptomic and microbiomic profiling. Int Immunopharmacol 2024; 142:113100. [PMID: 39244901 DOI: 10.1016/j.intimp.2024.113100] [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: 05/20/2024] [Revised: 08/08/2024] [Accepted: 09/03/2024] [Indexed: 09/10/2024]
Abstract
INTRODUCTION The skeletal system ranks as the third most common site for cancer metastasis, often leading to pain with nociceptive and neuropathic features. Programmed cell death protein 1 (PD-1)-targeting therapeutic antibodies offer effective cancer treatment but can cause treatment-related acute pain. Understanding the mechanisms of this pain and identifying potential interventions is still a challenge. METHODS A murine model of bone cancer pain was established using Lewis lung carcinoma (LLC) cells, followed by intravenous administration of nivolumab, a human anti-PD-1 monoclonal antibody. Pain thresholds were measured, and micro-CT images of the skeletal system were obtained. High-throughput sequencing of the spinal cord/colon transcriptome during the acute phase of bone cancer pain and gut microbiota analysis at the end of the treatment were performed. Immunofluorescence staining and western blot experiments assessed spinal cord microglia activation and acute pain-associated molecules. RESULTS PD-1 inhibition with nivolumab protected against bone degradation initiated by LLC cell administration but consistently induced acute pain during nivolumab treatment. Spinal cord and colon transcriptomics revealed an immunopathological pattern during tumor progression and the acute pain phase, with notable changes in interleukin and S100 gene families. Gut microbiota analysis post-immunotherapy showed a decline in beneficial bacteria associated with short-chain fatty acid (SCFA) production. Activation of spinal cord microglia and enhanced glycolytic metabolism were confirmed as key factors in inducing acute pain following immunotherapy. CONCLUSIONS This study reveals that nivolumab induces acute pain by activating microglia and enhancing glycolytic metabolism in the treatment of bone cancer and uncovers connections between transcriptomic changes, gut microbiota, and acute pain following immune checkpoint blockade (ICB) treatment. It offers novel insights into the relationship between immune checkpoint blockade therapies and pain management.
Collapse
Affiliation(s)
- Ruifeng Ding
- Department of Anesthesiology, Changzheng Hospital, Second Affiliated Hospital of Naval Medical University, Shanghai 200003, China
| | - Jinfang Lu
- Department of Anesthesiology, Changzheng Hospital, Second Affiliated Hospital of Naval Medical University, Shanghai 200003, China
| | - Xingshuai Huang
- Department of Anesthesiology, Changzheng Hospital, Second Affiliated Hospital of Naval Medical University, Shanghai 200003, China
| | - Mengqiu Deng
- Department of Anesthesiology, Changzheng Hospital, Second Affiliated Hospital of Naval Medical University, Shanghai 200003, China
| | - Huawei Wei
- Department of Anesthesiology, Changzheng Hospital, Second Affiliated Hospital of Naval Medical University, Shanghai 200003, China
| | - Guowei Jiang
- Department of Anesthesiology, Changzheng Hospital, Second Affiliated Hospital of Naval Medical University, Shanghai 200003, China
| | - Hongwei Zhu
- Department of Anesthesiology, Changzheng Hospital, Second Affiliated Hospital of Naval Medical University, Shanghai 200003, China
| | - Hongbin Yuan
- Department of Anesthesiology, Changzheng Hospital, Second Affiliated Hospital of Naval Medical University, Shanghai 200003, China.
| |
Collapse
|
23
|
Feng Y, Deyanat-Yazdi G, Newburn K, Potter S, Wortinger M, Ramirez M, Truhlar SME, Yachi PP. PD-1 antibody interactions with Fc gamma receptors enable PD-1 agonism to inhibit T cell activation - therapeutic implications for autoimmunity. J Autoimmun 2024; 149:103339. [PMID: 39608214 DOI: 10.1016/j.jaut.2024.103339] [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: 11/21/2023] [Revised: 08/30/2024] [Accepted: 11/12/2024] [Indexed: 11/30/2024]
Abstract
PD-1 has emerged as a central inhibitory checkpoint receptor in maintaining immune homeostasis and as a target in cancer immunotherapies. However, targeting PD-1 for the treatment of autoimmune diseases has been more challenging. We recently showed in a phase 2a trial that PD-1 could be stimulated with the PD-1 agonist antibody peresolimab to treat rheumatoid arthritis. Here, we demonstrate that PD-1 antibodies can elicit agonism and inhibit T cell activation by co-localization of PD-1 with the T cell receptor via Fcγ receptor (FcγR) engagement. Three PD-1 agonist antibodies with different antigen binding domains, including the clinically validated PD-1 blocking antibody pembrolizumab, suppressed T cell activation to a similar degree; this finding suggests that a specific PD-1-binding epitope is not required for PD-1 agonism. We next explored whether antibody-mediated clustering was an important driver of inhibition of T cell activation; however, we found that a monovalent PD-1 antibody was not inferior to a conventional bivalent antibody in its ability to suppress T cell activation. Importantly, we found that affinity to PD-1 correlated positively with inhibition of T cell activation, with higher affinity antibodies exhibiting higher levels of inhibition. Using a series of human Fc mutants with altered affinities to various FcγRs, we dissected the contributions of FcγRs and found that multiple FcγRs rather than a single receptor contribute to agonist activity. Our work reveals an important role for FcγR binding in the activity of PD-1 antibodies, which has implications for optimizing both PD-1 agonist and antagonist antibodies.
Collapse
MESH Headings
- Humans
- Programmed Cell Death 1 Receptor/immunology
- Programmed Cell Death 1 Receptor/metabolism
- Receptors, IgG/metabolism
- Receptors, IgG/immunology
- Lymphocyte Activation/immunology
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- T-Lymphocytes/drug effects
- Autoimmunity
- Protein Binding
- Immune Checkpoint Inhibitors/pharmacology
- Immune Checkpoint Inhibitors/therapeutic use
- Antibodies, Monoclonal, Humanized/pharmacology
- Antibodies, Monoclonal, Humanized/therapeutic use
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell/metabolism
- Autoimmune Diseases/immunology
- Autoimmune Diseases/drug therapy
- Autoimmune Diseases/therapy
Collapse
Affiliation(s)
- Yiqing Feng
- Biotechnology Discovery Research, Lilly Research Laboratories, Lilly Technology Center North, Indianapolis, IN, USA
| | - Gordafaried Deyanat-Yazdi
- Immunology Discovery Research, Lilly Research Laboratories, Lilly Biotechnology Center, San Diego, CA, USA
| | - Kristin Newburn
- Biotechnology Discovery Research, Lilly Research Laboratories, Lilly Technology Center North, Indianapolis, IN, USA
| | - Scott Potter
- Immunology Discovery Research, Lilly Research Laboratories, Lilly Biotechnology Center, San Diego, CA, USA
| | - Mark Wortinger
- Biotechnology Discovery Research, Lilly Research Laboratories, Lilly Corporate Center, Indianapolis, IN, USA
| | - Miriam Ramirez
- Immunology Discovery Research, Lilly Research Laboratories, Lilly Biotechnology Center, San Diego, CA, USA
| | - Stephanie M E Truhlar
- Biotechnology Discovery Research, Lilly Research Laboratories, Lilly Biotechnology Center, San Diego, CA, USA
| | - Pia P Yachi
- Immunology Discovery Research, Lilly Research Laboratories, Lilly Biotechnology Center, San Diego, CA, USA.
| |
Collapse
|
24
|
Jiang H, Ye Y, Wang M, Sun X, Sun T, Chen Y, Li P, Zhang M, Wang T. The progress on the relationship between gut microbiota and immune checkpoint blockade in tumors. Biotechnol Genet Eng Rev 2024; 40:4446-4465. [PMID: 37191003 DOI: 10.1080/02648725.2023.2212526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Accepted: 05/05/2023] [Indexed: 05/17/2023]
Abstract
Immune checkpoint blockade (ICB) has emerged as a promising immunotherapeutic approach for the treatment of various tumors. However, the efficacy of this therapy is limited in a subset of patients, and it is important to develop strategies to enhance immune responses. Studies have demonstrated a critical role of gut microbiota in regulating the therapeutic response to ICB. Gut microbiota composition, diversity, and function are mediated by metabolites, such as short-chain fatty acids and secondary bile acids, that interact with host immune cells through specific receptors. In addition, gut bacteria may translocate to the tumor site and stimulate antitumor immune responses. Therefore, maintaining a healthy gut microbiota composition, for instance through avoiding the use of antibiotics or probiotic interventions, can be an effective approach to optimize ICB therapy. This review summarizes the current understanding of the microbiota-immunity interactions in the context of ICB therapy, and discusses potential clinical implications of these findings.
Collapse
Affiliation(s)
- Haili Jiang
- Oncology Department of Integrated Traditional Chinese and Western Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Yingquan Ye
- Oncology Department of Integrated Traditional Chinese and Western Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Mingqi Wang
- Oncology Department of Integrated Traditional Chinese and Western Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Xin Sun
- Oncology Department of Integrated Traditional Chinese and Western Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Ting Sun
- Oncology Department of Integrated Traditional Chinese and Western Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Yang Chen
- Oncology Department of Integrated Traditional Chinese and Western Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Ping Li
- Oncology Department of Integrated Traditional Chinese and Western Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Mei Zhang
- Oncology Department of Integrated Traditional Chinese and Western Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Ting Wang
- Oncology Department of Integrated Traditional Chinese and Western Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| |
Collapse
|
25
|
Eren E, Das J, Tollefsbol TO. Polyphenols as Immunomodulators and Epigenetic Modulators: An Analysis of Their Role in the Treatment and Prevention of Breast Cancer. Nutrients 2024; 16:4143. [PMID: 39683540 PMCID: PMC11644657 DOI: 10.3390/nu16234143] [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: 11/05/2024] [Revised: 11/26/2024] [Accepted: 11/27/2024] [Indexed: 12/18/2024] Open
Abstract
Breast cancer poses a substantial health challenge for women globally. Recently, there has been a notable increase in scholarly attention regarding polyphenols, primarily attributed to not only the adverse effects associated with conventional treatments but also their immune-preventive impacts. Polyphenols, nature-derived substances present in vegetation, including fruits and vegetables, have received considerable attention in various fields of science due to their probable wellness merits, particularly in the treatment and hindrance of cancer. This review focuses on the immunomodulatory effects of polyphenols in breast cancer, emphasizing their capacity to influence the reaction of adaptive and innate immune cells within the tumor-associated environment. Polyphenols are implicated in the modulation of inflammation, the enhancement of antioxidant defenses, the promotion of epigenetic modifications, and the support of immune functions. Additionally, these compounds have been shown to influence the activity of critical immune cells, including macrophages and T cells. By targeting pathways involved in immune evasion, polyphenols may augment the capacity of the defensive system to detect and eliminate tumors. The findings suggest that incorporating polyphenol-rich foods into the diet could offer a promising, collaborative (integrative) approach to classical breast cancer remedial procedures by regulating how the defense mechanism interacts with the disease.
Collapse
Affiliation(s)
- Esmanur Eren
- Department of Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (E.E.); (J.D.)
| | - Jyotirmoyee Das
- Department of Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (E.E.); (J.D.)
| | - Trygve O. Tollefsbol
- Department of Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (E.E.); (J.D.)
- Integrative Center for Aging Research, University of Alabama at Birmingham, Birmingham, AL 35294, USA
- O’Neal Comprehensive Cancer Research, University of Alabama at Birmingham, Birmingham, AL 35294, USA
- Nutrition Obesity Research Center, University of Alabama at Birmingham, Birmingham, AL 35294, USA
- Comprehensive Diabetes Center, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| |
Collapse
|
26
|
Ben Saad E, Oroya A, Anto NP, Bachais M, Rudd CE. PD-1 endocytosis unleashes the cytolytic potential of checkpoint blockade in tumor immunity. Cell Rep 2024; 43:114907. [PMID: 39471174 DOI: 10.1016/j.celrep.2024.114907] [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: 10/03/2023] [Revised: 09/12/2024] [Accepted: 10/08/2024] [Indexed: 11/01/2024] Open
Abstract
PD-1 immune checkpoint blockade (ICB) is a key cancer treatment. While blocking PD-1 binding to ligand is known, the role of internalization in enhancing ICB efficacy is less explored. Our study reveals that PD-1 internalization helps unlock ICB's full potential in cancer immunotherapy. Anti-PD-1 induces 50%-60% surface PD-1 internalization from human and mouse cells, leaving low to intermediate levels of resistant receptors. Complexes then appear in early and late endosomes. Both CD4 and CD8 T cells, especially CD8+ effectors, are affected. Nivolumab outperforms pembrolizumab in human T cells, while PD-1 internalization requires crosslinking by bivalent antibody. While mono- and bivalent anti-PD-1 inhibit tumor growth with CD8 tumor-infiltrating cells expressing increased granzyme B, bivalent antibody is more effective where the combination of steric blockade and endocytosis induces greater CD8+ T cell tumor infiltration and the expression of the cytolytic pore protein, perforin. Our findings highlight an ICB mechanism that combines steric blockade and PD-1 endocytosis for optimal checkpoint immunotherapy.
Collapse
Affiliation(s)
- Elham Ben Saad
- Department of Medicine, Universite de Montréal, Montréal, QC H3C 3J7, Canada; Centre de Recherche Hopital Maisonneuve-Rosemont, Montréal, Quebec, QC H1T 2M4, Canada; Department of Biochemistry and Molecular Medicine, Universite de Montréal, Montréal, QC H3T 1J4, Canada
| | - Andres Oroya
- Department of Medicine, Universite de Montréal, Montréal, QC H3C 3J7, Canada; Centre de Recherche Hopital Maisonneuve-Rosemont, Montréal, Quebec, QC H1T 2M4, Canada; Department of Microbiology, Infection and Immunology, Universite de Montréal, Montréal, QC, Canada
| | - Nikhil Ponnoor Anto
- Department of Medicine, Universite de Montréal, Montréal, QC H3C 3J7, Canada; Centre de Recherche Hopital Maisonneuve-Rosemont, Montréal, Quebec, QC H1T 2M4, Canada; Department of Microbiology, Infection and Immunology, Universite de Montréal, Montréal, QC, Canada
| | - Meriem Bachais
- Department of Medicine, Universite de Montréal, Montréal, QC H3C 3J7, Canada; Centre de Recherche Hopital Maisonneuve-Rosemont, Montréal, Quebec, QC H1T 2M4, Canada; Department of Microbiology, Infection and Immunology, Universite de Montréal, Montréal, QC, Canada
| | - Christopher E Rudd
- Department of Medicine, Universite de Montréal, Montréal, QC H3C 3J7, Canada; Centre de Recherche Hopital Maisonneuve-Rosemont, Montréal, Quebec, QC H1T 2M4, Canada; Department of Biochemistry and Molecular Medicine, Universite de Montréal, Montréal, QC H3T 1J4, Canada; Department of Microbiology, Infection and Immunology, Universite de Montréal, Montréal, QC, Canada.
| |
Collapse
|
27
|
Huo MH, Adeerjiang Y, Abulitipu A, Khan U, Li XX, Zhang L, Tian Y, Jiang S, Xu CC, Chao XZ, Yang YF, Zhang JX, Du GL. Th17/Treg cell balance in patients with papillary thyroid carcinoma: a new potential biomarker and therapeutic target. Front Oncol 2024; 14:1325575. [PMID: 39534095 PMCID: PMC11554530 DOI: 10.3389/fonc.2024.1325575] [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: 10/21/2023] [Accepted: 10/10/2024] [Indexed: 11/16/2024] Open
Abstract
Papillary thyroid carcinoma (PTC) is the most common subtype of thyroid carcinoma. The most effective treatment for PTC is surgical resection, and patients who undergo surgery have good survival outcomes, but some patients have distant metastasis or even multiorgan metastases at the time of initial diagnosis. Distant metastasis is associated with poorer prognosis and a higher mortality rate. Helper T lymphocyte 17 (Th17) cells and regulatory T lymphocytes (Tregs) play different roles in PTC, and the Th17/Treg balance is closely related to the progression of PTC. Th17 cells play anticancer roles, whereas Tregs play cancer-promoting roles. A Th17/Treg imbalance promotes tumor progression and accelerates invasive behaviors such as tumor metastasis. Th17/Treg homeostasis can be regulated by the TGF-β/IL-2 and IL-6 cytokine axes. Immune checkpoint inhibitors contribute to Treg/Th17 cell homeostasis. For PTC, monoclonal antibodies against CTLA-4, PD-1 and PD-L1 inhibit the activation of Tregs, reversing the Th17/Treg cell imbalance and providing a new option for the prevention and treatment of PTC. This article reviews the role of Tregs and Th17 cells in PTC and their potential targets, aiming to provide better treatment options for PTC.
Collapse
Affiliation(s)
- Meng-Han Huo
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Urumqi, China
- Department of Endocrinology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
- Department of Gastroenterology and Endocrinology, Tianjin Haihe Hospital, Tianjin, China
| | - Yilinuer Adeerjiang
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Urumqi, China
- Department of Endocrinology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Ayiguzhali Abulitipu
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Urumqi, China
- Department of Endocrinology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Umair Khan
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Urumqi, China
- Department of Endocrinology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Xin-Xi Li
- Department of Endocrine Surgery, First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Lei Zhang
- Department of Endocrine Surgery, First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Ye Tian
- Department of Endocrine Surgery, First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Sheng Jiang
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Urumqi, China
- Department of Endocrinology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Can-Can Xu
- First Clinical Medical College of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Xian-Zhen Chao
- First Clinical Medical College of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Ye-Fan Yang
- First Clinical Medical College of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Jin-Xia Zhang
- First Clinical Medical College of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Guo-Li Du
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Urumqi, China
- Department of Endocrinology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
- Department of Endocrinology, Bayingolin Mongolian Autonomous Prefecture People's Hospital, Kuerle, China
| |
Collapse
|
28
|
Yuan G, Chen Y, Zhu P, Deng Q, Su K, Liu J, Wang Y, Li R, Li W, Zang M, Hu X, Wang JJ, Li Q, Du Y, Chen J. Cadonilimab (PD-1/CTLA-4) in combination with lenvatinib in unresectable hepatocellular carcinoma (uHCC): A retrospective real-world study. Heliyon 2024; 10:e37616. [PMID: 39398001 PMCID: PMC11467631 DOI: 10.1016/j.heliyon.2024.e37616] [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: 06/20/2024] [Revised: 08/23/2024] [Accepted: 09/06/2024] [Indexed: 10/15/2024] Open
Abstract
Background Previous research has shown that combining tyrosine kinase inhibitors (TKIs) with immunotherapy results in synergistic clinical efficacy. Cadonilimab, the first approved bi-specific antibody targeting PD-1 and CTLA-4, was studied to evaluate its efficacy and safety in combination with Lenvatinib as a first-line treatment for patients with unresectable hepatocellular carcinoma (uHCC). Methods A retrospective study was conducted on 29 uHCC patients diagnosed at Nanfang Hospital, Southern Medical University, between July 7, 2022, and March 3, 2023. Patients received Cadonilimab (10 mg/kg, IV, every 3 weeks) combined with Lenvatinib (8 mg, orally, daily). The primary endpoint was the objective response rate (ORR), with secondary endpoints including disease control rate (DCR), median progression-free survival (mPFS), median overall survival (mOS), median time to progression (mTTP), and safety. Results By April 2023, 29 patients had been enrolled in the study. The ORR was 37.9 %, DCR was 82.8 %, mPFS was 8.1 months, mTTP was 8.2 months, and mOS was not reached. A total of 93.1 % of patients experienced at least one treatment-related adverse event (TRAE). The most common adverse events were weight loss (51.7 %), increased aspartate aminotransferase (48.3 %), leukocytopenia (48.3 %), and neutropenia (48.3 %). TRAEs of grade 3 or higher occurred in 51.7 % of patients, with no grade 4 TRAEs observed. Conclusion This study demonstrated the efficacy and safety of this combination, potentially improving outcomes as a first-line therapy, and offering a novel therapeutic approach for advanced HCC.
Collapse
Affiliation(s)
- Guosheng Yuan
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Yongru Chen
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Peilin Zhu
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Qiong Deng
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Kaiyan Su
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Jie Liu
- Department of Infectious Diseases, Ganzhou Hospital of Nanfang Hospital, Southern Medical University, Ganzhou, Jiangxi, 341099, China
| | - Yan Wang
- Medical Center, Akeso Biopharma, Inc, Zhongshan, China
| | - Rong Li
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Wenli Li
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Mengya Zang
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Xiaoyun Hu
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Jun-Jie Wang
- Department of Infectious Diseases, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Qi Li
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Yangfeng Du
- Department of Oncology, Changde Hospital, Xiangya School of Medicine, Central South University(The First People's Hospital of Changde City), Changde, China
| | - Jinzhang Chen
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China
| |
Collapse
|
29
|
Liang B, Xing X, Storts H, Ye Z, Claybon H, Austin R, Ding R, Liu B, Wen H, Miles WO, Fishel R, Wang JJ. Antagonistic roles of cGAS/STING signaling in colorectal cancer chemotherapy. Front Oncol 2024; 14:1441935. [PMID: 39469633 PMCID: PMC11513249 DOI: 10.3389/fonc.2024.1441935] [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: 05/31/2024] [Accepted: 09/27/2024] [Indexed: 10/30/2024] Open
Abstract
FOLFOX, composed of 5-FU, oxaliplatin and leucovorin, is a first line chemotherapy regimen for colorectal cancer (CRC) treatment. In this study, we show that 5-FU and oxaliplatin induce DNA damage and activate cGAS/STING signaling leading to enhanced expression of interferon (IFN) β, IFN-stimulated genes and inflammatory cytokines in mouse and human colon cancer cells as well as increased intratumoral CD8+ T cells in mice. Crucially, 5-FU and oxaliplatin increase PD-L1 expression at the mRNA and protein levels, which has been shown to inhibit CD8+ T cell function. Depletion of cGAS, STING, IRF3, or IFNα/β receptor 1 (IFNAR1) abolishes this increase, indicating that 5-FU/oxaliplatin mediated upregulation of PD-L1 expression is dependent on tumor cell intrinsic cGAS/STING signaling. These results imply opposing roles for FOLFOX during cancer treatment. On one hand, 5-FU and oxaliplatin activate the innate immune response to facilitate anti-tumor immunity, and conversely upregulate PD-L1 expression to evade immune surveillance. Analysis of TCGA colon cancer dataset shows a positive correlation between expression of PD-L1 and components of the cGAS/STING pathway, supporting a role for cGAS/STING signaling in upregulating PD-L1 expression in colon cancer patients. Tumor studies in syngeneic immune competent mice demonstrate that the combination of 5-FU/oxaliplatin and anti-PD-1 significantly reduced tumor growth of colon cancer cells compared to 5-FU/oxaliplatin treatment alone. Taken together, our studies have identified a unique pathway leading to chemoresistance and provide a rationale to combine FOLFOX with anti-PD-1/PD-L1 as an effective CRC treatment.
Collapse
Affiliation(s)
- Beiyuan Liang
- Department of Cancer Biology and Genetics, The Ohio State University, Columbus, OH, United States
| | - Xuanxuan Xing
- Department of Cancer Biology and Genetics, The Ohio State University, Columbus, OH, United States
| | - Hayden Storts
- Department of Cancer Biology and Genetics, The Ohio State University, Columbus, OH, United States
| | - Zhen Ye
- Department of Cancer Biology and Genetics, The Ohio State University, Columbus, OH, United States
| | - Hazel Claybon
- Department of Cancer Biology and Genetics, The Ohio State University, Columbus, OH, United States
| | - Ryan Austin
- Department of Cancer Biology and Genetics, The Ohio State University, Columbus, OH, United States
| | - Rachel Ding
- Department of Cancer Biology and Genetics, The Ohio State University, Columbus, OH, United States
| | - Bei Liu
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, OH, United States
- Pelotonia Institute for Immuno-Oncology, James Comprehensive Cancer Center, Wexner Medical Center, The Ohio State University, Columbus, OH, United States
| | - Haitao Wen
- Pelotonia Institute for Immuno-Oncology, James Comprehensive Cancer Center, Wexner Medical Center, The Ohio State University, Columbus, OH, United States
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, United States
| | - Wayne O. Miles
- Department of Cancer Biology and Genetics, The Ohio State University, Columbus, OH, United States
| | - Richard Fishel
- Department of Cancer Biology and Genetics, The Ohio State University, Columbus, OH, United States
| | - Jing J. Wang
- Department of Cancer Biology and Genetics, The Ohio State University, Columbus, OH, United States
- Pelotonia Institute for Immuno-Oncology, James Comprehensive Cancer Center, Wexner Medical Center, The Ohio State University, Columbus, OH, United States
| |
Collapse
|
30
|
Zhang Y, Huang W, Jiao H, Kang L. PET radiomics in lung cancer: advances and translational challenges. EJNMMI Phys 2024; 11:81. [PMID: 39361110 PMCID: PMC11450131 DOI: 10.1186/s40658-024-00685-5] [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: 11/19/2023] [Accepted: 09/26/2024] [Indexed: 10/06/2024] Open
Abstract
Radiomics is an emerging field of medical imaging that aims at improving the accuracy of diagnosis, prognosis, treatment planning and monitoring non-invasively through the automated or semi-automated quantitative analysis of high-dimensional image features. Specifically in the field of nuclear medicine, radiomics utilizes imaging methods such as positron emission tomography (PET) and single photon emission computed tomography (SPECT) to evaluate biomarkers related to metabolism, blood flow, cellular activity and some biological pathways. Lung cancer ranks among the leading causes of cancer-related deaths globally, and radiomics analysis has shown great potential in guiding individualized therapy, assessing treatment response, and predicting clinical outcomes. In this review, we summarize the current state-of-the-art radiomics progress in lung cancer, highlighting the potential benefits and existing limitations of this approach. The radiomics workflow was introduced first including image acquisition, segmentation, feature extraction, and model building. Then the published literatures were described about radiomics-based prediction models for lung cancer diagnosis, differentiation, prognosis and efficacy evaluation. Finally, we discuss current challenges and provide insights into future directions and potential opportunities for integrating radiomics into routine clinical practice.
Collapse
Affiliation(s)
- Yongbai Zhang
- Department of Nuclear Medicine, Peking University First Hospital, No. 8 Xishiku Str., Xicheng Dist, Beijing, 100034, China
| | - Wenpeng Huang
- Department of Nuclear Medicine, Peking University First Hospital, No. 8 Xishiku Str., Xicheng Dist, Beijing, 100034, China
| | - Hao Jiao
- Department of Nuclear Medicine, Peking University First Hospital, No. 8 Xishiku Str., Xicheng Dist, Beijing, 100034, China
| | - Lei Kang
- Department of Nuclear Medicine, Peking University First Hospital, No. 8 Xishiku Str., Xicheng Dist, Beijing, 100034, China.
| |
Collapse
|
31
|
Wang SW, Zheng QY, Hong WF, Tang BF, Hsu SJ, Zhang Y, Zheng XB, Zeng ZC, Gao C, Ke AW, Du SS. Mechanism of immune activation mediated by genomic instability and its implication in radiotherapy combined with immune checkpoint inhibitors. Radiother Oncol 2024; 199:110424. [PMID: 38997092 DOI: 10.1016/j.radonc.2024.110424] [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: 04/07/2024] [Revised: 06/27/2024] [Accepted: 07/05/2024] [Indexed: 07/14/2024]
Abstract
Various genetic and epigenetic changes associated with genomic instability (GI), including DNA damage repair defects, chromosomal instability, and mitochondrial GI, contribute to development and progression of cancer. These alterations not only result in DNA leakage into the cytoplasm, either directly or through micronuclei, but also trigger downstream inflammatory signals, such as the cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) signaling pathway. Apart from directly inducing DNA damage to eliminate cancer cells, radiotherapy (RT) exerts its antitumor effects through intracellular DNA damage sensing mechanisms, leading to the activation of downstream inflammatory signaling pathways. This not only enables local tumor control but also reshapes the immune microenvironment, triggering systemic immune responses. The combination of RT and immunotherapy has emerged as a promising approach to increase the probability of abscopal effects, where distant tumors respond to treatment due to the systemic immunomodulatory effects. This review emphasizes the importance of GI in cancer biology and elucidates the mechanisms by which RT induces GI remodeling of the immune microenvironment. By elucidating the mechanisms of GI and RT-induced immune responses, we aim to emphasize the crucial importance of this approach in modern oncology. Understanding the impact of GI on tumor biological behavior and therapeutic response, as well as the possibility of activating systemic anti-tumor immunity through RT, will pave the way for the development of new treatment strategies and improve prognosis for patients.
Collapse
Affiliation(s)
- Si-Wei Wang
- Department of Radiation Oncology, Zhongshan Hospital, Fudan University, Shanghai 200030, China; Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Zhongshan Hospital, Liver Cancer Institute, Fudan University, Shanghai 200030, China
| | - Qiu-Yi Zheng
- Department of Radiation Oncology, Zhongshan Hospital, Fudan University, Shanghai 200030, China
| | - Wei-Feng Hong
- Department of Radiation Oncology, Zhongshan Hospital, Fudan University, Shanghai 200030, China
| | - Bu-Fu Tang
- Department of Radiation Oncology, Zhongshan Hospital, Fudan University, Shanghai 200030, China
| | - Shu-Jung Hsu
- Department of Radiation Oncology, Zhongshan Hospital, Fudan University, Shanghai 200030, China
| | - Yang Zhang
- Department of Radiation Oncology, Zhongshan Hospital, Fudan University, Shanghai 200030, China
| | - Xiao-Bin Zheng
- Department of Radiation Oncology, Zhongshan Hospital, Fudan University, Shanghai 200030, China
| | - Zhao-Chong Zeng
- Department of Radiation Oncology, Zhongshan Hospital, Fudan University, Shanghai 200030, China
| | - Chao Gao
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Zhongshan Hospital, Liver Cancer Institute, Fudan University, Shanghai 200030, China.
| | - Ai-Wu Ke
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Zhongshan Hospital, Liver Cancer Institute, Fudan University, Shanghai 200030, China.
| | - Shi-Suo Du
- Department of Radiation Oncology, Zhongshan Hospital, Fudan University, Shanghai 200030, China.
| |
Collapse
|
32
|
Carlomagno S, Setti C, Ortolani F, Sivori S. Pancreatic ductal adenocarcinoma microenvironment: Soluble factors and cancer associated fibroblasts as modulators of NK cell functions. Immunol Lett 2024; 269:106898. [PMID: 39019404 DOI: 10.1016/j.imlet.2024.106898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 07/13/2024] [Indexed: 07/19/2024]
Abstract
Pancreatic Ductal Adenocarcinoma (PDAC) is the most frequent pancreatic cancer and represents one of the most aggressive human neoplasms. Typically identified at advance stage disease, most PDAC tumors are unresectable and resistant to standard therapies. The immunosuppressive microenvironment in PDAC impedes tumor control but a greater understanding of the complex stromal interactions within the tumor microenvironment (TME) and the development of strategies capable of restoring antitumor effector immune responses could be crucial to fight this aggressive tumor and its spread. Natural Killer (NK) cells play a crucial role in cancer immunosurveillance and represent an attractive target for immunotherapies, both as cell therapy and as a pharmaceutical target. This review describes some crucial components of the PDAC TME (collagens, soluble factors and fibroblasts) that can influence the presence, phenotype and function of NK cells in PDAC patients tumor tissue. This focused overview highlights the therapeutic relevance of dissecting the complex stromal composition to define new strategies for NK cell-based immunotherapies to improve the treatment of PDAC.
Collapse
Affiliation(s)
- Simona Carlomagno
- Department of Medicine (DMED), University of Udine, Piazzale Kolbe 4, Udine 33100, Italy.
| | - Chiara Setti
- Department of Experimental Medicine (DIMES), University of Genoa, Via Leon Battista Alberti 2, Genoa 16132, Italy
| | - Fulvia Ortolani
- Department of Medicine (DMED), University of Udine, Piazzale Kolbe 4, Udine 33100, Italy
| | - Simona Sivori
- Department of Experimental Medicine (DIMES), University of Genoa, Via Leon Battista Alberti 2, Genoa 16132, Italy; IRCCS Ospedale Policlinico San Martino, Genoa, Italy.
| |
Collapse
|
33
|
Liu S, Lu P, Yang B, Yang Y, Zhou H, Yang M. Single-cell RNA sequencing analysis of peripheral blood mononuclear cells in PD-1-induced renal toxicity in patients with lung cancer. BMC Nephrol 2024; 25:307. [PMID: 39277735 PMCID: PMC11401319 DOI: 10.1186/s12882-024-03754-0] [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: 11/14/2023] [Accepted: 09/11/2024] [Indexed: 09/17/2024] Open
Abstract
BACKGROUND Although the patient survival rate for many malignancies has been improved with immune checkpoint inhibitors (ICIs), some patients experience various immune-related adverse events (irAEs). IrAEs impact several organ systems, including the kidney. With anti-programmed cell death protein 1 (PD-1) therapy (pembrolizumab), kidney-related adverse events occur relatively rarely compared with other irAEs. However, the occurrence of AKI usually leads to anti-PD-1 therapy interruption or discontinuation. Therefore, there is an urgent need to clarify the mechanisms of renal irAEs (R-irAEs) to facilitate early management. This study aimed to analyse the characteristics of peripheral blood mononuclear cells (PBMCs) in R-irAEs. METHODS PBMCs were collected from three patients who developed R-irAEs after anti-PD-1 therapy and three patients who did not. The PBMCs were subjected to scRNA-seq to identify cell clusters and differentially expressed genes (DEGs). Kyoto Encyclopedia of Genes and Genomes (KEGG) and gene ontology (GO) enrichment analyses were performed to investigate the most active biological processes in immune cells. RESULTS Fifteen cell clusters were identified across the two groups. FOS, RPS26, and JUN were the top three upregulated genes in CD4+ T cells. The DEGs in CD4+ T cells were enriched in Th17 differentiation, Th1 and Th2 cell differentiation, NF-kappa B, Nod-like receptor, TNF, IL-17, apoptosis, and NK cell-mediated cytotoxicity signaling pathways. RPS26, TRBV25-1, and JUN were the top three upregulated genes in CD8+ T cells. The DEGs in CD8+ T cells were enriched in Th17 cell differentiation, antigen processing and presentation, natural killer cell-mediated cytotoxicity, the intestinal immune network for IgA production, the T-cell receptor signalling pathway, Th1 and Th2 cell differentiation, the phagosome, and cell adhesion molecules. CONCLUSIONS In conclusion, R-irAEs are associated with immune cell dysfunction. DEGs and their enriched pathways identified in CD4+ T cells and CD8+ T cells play important roles in the development of renal irAEs related to anti-PD-1 therapy. These findings offer fresh perspectives on the pathogenesis of renal damage caused by anti-PD-1 therapy.
Collapse
Affiliation(s)
- Shusu Liu
- Department of Nephrology, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, China
| | - Peiyu Lu
- Department of Nephrology, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, China
| | - Bixia Yang
- Department of Nephrology, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, China
| | - Yan Yang
- Department of Nephrology, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, China
| | - Hua Zhou
- Department of Nephrology, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, China
| | - Min Yang
- Department of Nephrology, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, China.
| |
Collapse
|
34
|
Chandrasekaran V, Andersson KME, Erlandsson M, Li S, Olsson TN, Garcia-Bonete MJ, Malmhäll-Bah E, Johansson P, Katona G, Bokarewa MI. Bivalent chromatin accommodates survivin and BRG1/SWI complex to activate DNA damage response in CD4 + cells. Cell Commun Signal 2024; 22:440. [PMID: 39261837 PMCID: PMC11389452 DOI: 10.1186/s12964-024-01814-4] [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: 04/23/2024] [Accepted: 09/01/2024] [Indexed: 09/13/2024] Open
Abstract
BACKGROUND Bivalent regions of chromatin (BvCR) are characterized by trimethylated lysine 4 (H3K4me3) and lysine 27 on histone H3 (H3K27me3) deposition which aid gene expression control during cell differentiation. The role of BvCR in post-transcriptional DNA damage response remains unidentified. Oncoprotein survivin binds chromatin and mediates IFNγ effects in CD4+ cells. In this study, we explored the role of BvCR in DNA damage response of autoimmune CD4+ cells in rheumatoid arthritis (RA). METHODS We performed deep sequencing of the chromatin bound to survivin, H3K4me3, H3K27me3, and H3K27ac, in human CD4+ cells and identified BvCR, which possessed all three histone H3 modifications. Protein partners of survivin on chromatin were predicted by integration of motif enrichment analysis, computational machine-learning, and structural modeling, and validated experimentally by mass spectrometry and peptide binding array. Survivin-dependent change in BvCR and transcription of genes controlled by the BvCR was studied in CD4+ cells treated with survivin inhibitor, which revealed survivin-dependent biological processes. Finally, the survivin-dependent processes were mapped to the transcriptome of CD4+ cells in blood and in synovial tissue of RA patients and the effect of modern immunomodulating drugs on these processes was explored. RESULTS We identified that BvCR dominated by H3K4me3 (H3K4me3-BvCR) accommodated survivin within cis-regulatory elements of the genes controlling DNA damage. Inhibition of survivin or JAK-STAT signaling enhanced H3K4me3-BvCR dominance, which improved DNA damage recognition and arrested cell cycle progression in cultured CD4+ cells. Specifically, BvCR accommodating survivin aided sequence-specific anchoring of the BRG1/SWI chromatin-remodeling complex coordinating DNA damage response. Mapping survivin interactome to BRG1/SWI complex demonstrated interaction of survivin with the subunits anchoring the complex to chromatin. Co-expression of BRG1, survivin and IFNγ in CD4+ cells rendered complete deregulation of DNA damage response in RA. Such cells possessed strong ability of homing to RA joints. Immunomodulating drugs inhibited the anchoring subunits of BRG1/SWI complex, which affected arthritogenic profile of CD4+ cells. CONCLUSIONS BvCR execute DNA damage control to maintain genome fidelity in IFN-activated CD4+ cells. Survivin anchors the BRG1/SWI complex to BvCR to repress DNA damage response. These results offer a platform for therapeutic interventions targeting survivin and BRG1/SWI complex in autoimmunity.
Collapse
Affiliation(s)
- Venkataragavan Chandrasekaran
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Box 480, Gothenburg, 40530, Sweden
| | - Karin M E Andersson
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Box 480, Gothenburg, 40530, Sweden
| | - Malin Erlandsson
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Box 480, Gothenburg, 40530, Sweden
- Rheumatology Clinic, Sahlgrenska University Hospital, Gröna Stråket 16, Gothenburg, 41346, Sweden
| | - Shuxiang Li
- Computational Biology and Biophysics Lab, Queen's University, Kingston, Canada
| | - Torbjörn Nur Olsson
- Department of Chemistry and Molecular Biology, Faculty of Science, University of Gothenburg, Gothenburg, Sweden
| | - Maria-Jose Garcia-Bonete
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, University of Gothenburg, Gothenburg, SE, 405 30, Sweden
| | - Eric Malmhäll-Bah
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Box 480, Gothenburg, 40530, Sweden
| | - Pegah Johansson
- Department of Clinical Chemistry, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Gergely Katona
- Department of Chemistry and Molecular Biology, Faculty of Science, University of Gothenburg, Gothenburg, Sweden
| | - Maria I Bokarewa
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Box 480, Gothenburg, 40530, Sweden.
- Rheumatology Clinic, Sahlgrenska University Hospital, Gröna Stråket 16, Gothenburg, 41346, Sweden.
| |
Collapse
|
35
|
Zheng X, Sun R, Wei T. Immune microenvironment in papillary thyroid carcinoma: roles of immune cells and checkpoints in disease progression and therapeutic implications. Front Immunol 2024; 15:1438235. [PMID: 39290709 PMCID: PMC11405226 DOI: 10.3389/fimmu.2024.1438235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Accepted: 08/14/2024] [Indexed: 09/19/2024] Open
Abstract
Papillary thyroid cancer (PTC) is the most common type of primary thyroid cancer. Despite the low malignancy and relatively good prognosis, some PTC cases are highly aggressive and even develop refractory cancer in the thyroid. Growing evidence suggested that microenvironment in tumor affected PTC biological behavior due to different immune states. Different interconnected components in the immune system influence and participate in tumor invasion, and are closely related to PTC metastasis. Immune cells and molecules are widely distributed in PTC tissues. Their quantity and proportion vary with the host's immune status, which suggests that immunotherapy may be a very promising therapeutic modality for PTC. In this paper, we review the role of immune cells and immune checkpoints in PTC immune microenvironment based on the characteristics of the PTC tumor microenvironment.
Collapse
Affiliation(s)
- Xun Zheng
- Department of Thyroid and Parathyroid Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Ruonan Sun
- Department of Thyroid and Parathyroid Surgery, West China Hospital, Sichuan University, Chengdu, China
- West China School of Medicine, Sichuan University, Chengdu, China
| | - Tao Wei
- Department of Thyroid and Parathyroid Surgery, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
36
|
Tanabe M, Funatsu N, Akiyama M, Takaki KI, Fujii Y, Seki E, Yamana K, Yoshikawa H, Sonoda KH. Clinical features and prognosis of conjunctival melanoma in Japanese patients. Jpn J Ophthalmol 2024; 68:463-471. [PMID: 38990387 DOI: 10.1007/s10384-024-01085-z] [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: 01/17/2024] [Accepted: 05/28/2024] [Indexed: 07/12/2024]
Abstract
PURPOSE To evaluate the clinical features and prognosis of conjunctival melanoma in Japanese patients. STUDY DESIGN Retrospective observational case series. METHODS Twenty patients (8 men and 12 women) diagnosed with conjunctival melanoma at a singlehospital between 2003 and 2017 were analyzed. Data on clinical presentation, sex, age, the affected eye, tumor location, tumor origin, tumor stage according to the American Joint Committee on Cancer staging system (eighth edition), treatment, outcomes, local recurrence, metastasis, and survival were extracted from the patients' medical records and reviewed. RESULTS The mean age at diagnosis was 64.2 ± 14.8 years. Tumor locations at the first examination included the bulbar conjunctiva (n = 19), plica (n = 13), and fornix (n = 12). The tumor stage was T1 in 5 cases (25%), T2 in 12 cases (60%), T3 in 3 cases (15%), and T4 in none. The mean follow-up duration was 91.7 ± 46.0 months. The local recurrence rates at 1, 5, and 10 years were 5.0%, 18.8%, and 31.5%, respectively, whilst the metastasis rates were 5.0%, 25.6%, and 32.4%, respectively. Four of the 6 patients who experienced metastasis died; duration from metastasis to death was 17.5 months (range, 7-25). The 5-year survival rate for conjunctival melanoma was 78.8%. Tumor thickness was significantly associated with survival duration on univariate Cox regression analyses. CONCLUSION The mortality rate for conjunctival melanoma in the Japanese population was lower and higher than that reported in the Chinese and United States populations, respectively. Tumor thickness was a prognostic factor for survival in patients with conjunctival melanoma.
Collapse
Affiliation(s)
- Mika Tanabe
- Department of Ophthalmology, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan.
| | - Naohiko Funatsu
- Department of Ophthalmology, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Masato Akiyama
- Department of Ophthalmology, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
- Department of Ocular Pathology and Imaging Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Ken-Ichi Takaki
- Department of Ophthalmology, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Yuya Fujii
- Department of Ophthalmology, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Eiko Seki
- Department of Ophthalmology, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Kanako Yamana
- Department of Ophthalmology, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Hiroshi Yoshikawa
- Department of Ophthalmology, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Koh-Hei Sonoda
- Department of Ophthalmology, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| |
Collapse
|
37
|
McGale JP, Chen DL, Trebeschi S, Farwell MD, Wu AM, Cutler CS, Schwartz LH, Dercle L. Artificial intelligence in immunotherapy PET/SPECT imaging. Eur Radiol 2024; 34:5829-5841. [PMID: 38355986 DOI: 10.1007/s00330-024-10637-3] [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: 09/27/2023] [Revised: 12/12/2023] [Accepted: 01/15/2024] [Indexed: 02/16/2024]
Abstract
OBJECTIVE Immunotherapy has dramatically altered the therapeutic landscape for oncology, but more research is needed to identify patients who are likely to achieve durable clinical benefit and those who may develop unacceptable side effects. We investigated the role of artificial intelligence in PET/SPECT-guided approaches for immunotherapy-treated patients. METHODS We performed a scoping review of MEDLINE, CENTRAL, and Embase databases using key terms related to immunotherapy, PET/SPECT imaging, and AI/radiomics through October 12, 2022. RESULTS Of the 217 studies identified in our literature search, 24 relevant articles were selected. The median (interquartile range) sample size of included patient cohorts was 63 (157). Primary tumors of interest were lung (n = 14/24, 58.3%), lymphoma (n = 4/24, 16.7%), or melanoma (n = 4/24, 16.7%). A total of 28 treatment regimens were employed, including anti-PD-(L)1 (n = 13/28, 46.4%) and anti-CTLA-4 (n = 4/28, 14.3%) monoclonal antibodies. Predictive models were built from imaging features using univariate radiomics (n = 7/24, 29.2%), radiomics (n = 12/24, 50.0%), or deep learning (n = 5/24, 20.8%) and were most often used to prognosticate (n = 6/24, 25.0%) or describe tumor phenotype (n = 5/24, 20.8%). Eighteen studies (75.0%) performed AI model validation. CONCLUSION Preliminary results suggest broad potential for the application of AI-guided immunotherapy management after further validation of models on large, prospective, multicenter cohorts. CLINICAL RELEVANCE STATEMENT This scoping review describes how artificial intelligence models are built to make predictions based on medical imaging and explores their application specifically in the PET and SPECT examination of immunotherapy-treated cancers. KEY POINTS • Immunotherapy has drastically altered the cancer treatment landscape but is known to precipitate response patterns that are not accurately accounted for by traditional imaging methods. • There is an unmet need for better tools to not only facilitate in-treatment evaluation but also to predict, a priori, which patients are likely to achieve a good response with a certain treatment as well as those who are likely to develop side effects. • Artificial intelligence applied to PET/SPECT imaging of immunotherapy-treated patients is mainly used to make predictions about prognosis or tumor phenotype and is built from baseline, pre-treatment images. Further testing is required before a true transition to clinical application can be realized.
Collapse
Affiliation(s)
- Jeremy P McGale
- Department of Radiology, New York-Presbyterian Hospital, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA.
| | - Delphine L Chen
- Department of Molecular Imaging and Therapy, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Department of Radiology, University of Washington, Seattle, WA, USA
| | - Stefano Trebeschi
- Department of Radiology, Netherlands Cancer Institute, Amsterdam, The Netherlands
- GROW School of Oncology and Reproduction, Maastricht University, Maastricht, The Netherlands
| | - Michael D Farwell
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Anna M Wu
- Department of Immunology and Theranostics, Beckman Research Institute of City of Hope, Duarte, CA, USA
| | - Cathy S Cutler
- Collider Accelerator Department, Brookhaven National Laboratory, Upton, NY, USA
| | - Lawrence H Schwartz
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Laurent Dercle
- Department of Radiology, New York-Presbyterian Hospital, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA.
| |
Collapse
|
38
|
Ma Z, Chen G, Li H, Yang S, Xu Y, Pan B, Lai W, Chen G, Liao W, Zhang X. B7-H3 promotes nasopharyngeal carcinoma progression by regulating CD8+ T cell exhaustion. Immun Inflamm Dis 2024; 12:e70005. [PMID: 39267471 PMCID: PMC11393430 DOI: 10.1002/iid3.70005] [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: 11/07/2023] [Revised: 06/26/2024] [Accepted: 08/10/2024] [Indexed: 09/17/2024] Open
Abstract
BACKGROUND B7-H3 protein is an important regulator of the adaptive immune response in human tumorigenesis. 4-1BB is a co-stimulatory receptor expressed on activated CD8+ T cells, and regulates T cell immunity. Here, we investigated the role of B7-H3 in the growth and invasion of nasopharyngeal carcinoma (NPC) and the effect of its interaction with 4-1BB on tumor immunity. METHODS Short hairpin (sh) RNA was designed to knock down B7-H3 expression in NPC cells. NPC cells with stable knockdown of B7-H3 were established and injected into nude mice. The effects of B7-H3 on cell proliferation, apoptosis, and epithelial-to-mesenchymal transition (EMT) were detected by the CCK8 assay, flow cytometry, TUNEL assay, and western blot analysis. The migration and invasion abilities were determined using the Transwell assay and scratch assay. Co-immunoprecipitation (Co-IP) assays were performed to study the interaction between B7-H3 and 4-1BB. Anti-4-1BB antibody was used in a co-culture system and xenograft mice to study the effect of 4-1BB on NPC development. RESULTS NPC cells transfected with sh-B7-H3 showed a higher rate of apoptosis, slower growth rate, impaired migration, and less EMT in vitro. Xenograft mice with stable knockout of B7-H3 had lower tumor burdens, and the stripped tumors had lower rates of cell proliferation, higher rates of apoptosis, and less EMT in vivo. Additionally, decreased B7-H3 expression was positively correlated with interferon-γ, tumor necrosis factor-α, and 4-1BB+CD8+ tumor-infiltrating lymphocytes. Co-IP studies showed that B7-H3 interacts with 4-1BB. Also, the inhibitory effects of sh-B7-H3 on NPC tumor growth, invasion, and tumor immunity could be alleviated by the anti-4-1BB antibody both in vivo and in vitro. CONCLUSION Our findings suggest that B7-H3 may accelerate tumor growth, tumor cell invasion, and EMT, and interact with 4-1BB to produce CD8+ T cell exhaustion that inhibits tumor immunity. B7-H3 might serve as a novel target for treating NPC.
Collapse
Affiliation(s)
- Zhaoen Ma
- The First Affiliated Hospital of Jinan UniversityGuangzhouChina
- Department of OtolaryngologyThe Second Affiliated Hospital of Guangzhou Medical UniversityGuangzhouChina
| | - Gui Chen
- Department of Otolaryngology, Head and Neck SurgeryThe First Affiliated Hospital of Guangzhou Medical UniversityGuangzhouChina
| | - Hao Li
- Department of Otolaryngology, Head and Neck SurgeryThe First Affiliated Hospital of Guangzhou Medical UniversityGuangzhouChina
| | - Saixuan Yang
- Department of Otolaryngology, Head and Neck SurgeryThe First Affiliated Hospital of Guangzhou Medical UniversityGuangzhouChina
| | - Yali Xu
- Department of OtolaryngologyThe Second Affiliated Hospital of Guangzhou Medical UniversityGuangzhouChina
| | - Bolin Pan
- Guangzhou Medical UniversityGuangzhouChina
| | - Wuping Lai
- Guangzhou Medical UniversityGuangzhouChina
| | - Guangui Chen
- Department of OtolaryngologyThe Second Affiliated Hospital of Guangzhou Medical UniversityGuangzhouChina
| | - Wenjing Liao
- Department of Otolaryngology, Head and Neck SurgeryThe First Affiliated Hospital of Guangzhou Medical UniversityGuangzhouChina
| | - Xiaowen Zhang
- The First Affiliated Hospital of Jinan UniversityGuangzhouChina
- Department of Otolaryngology, Head and Neck SurgeryThe First Affiliated Hospital of Guangzhou Medical UniversityGuangzhouChina
| |
Collapse
|
39
|
Jin S, Wan S, Xiong R, Li Y, Dong T, Guan C. The role of regulatory T cells in vitiligo and therapeutic advances: a mini-review. Inflamm Res 2024; 73:1311-1332. [PMID: 38839628 DOI: 10.1007/s00011-024-01900-w] [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: 03/01/2024] [Revised: 05/21/2024] [Accepted: 05/29/2024] [Indexed: 06/07/2024] Open
Abstract
BACKGROUND Regulatory T cells (Tregs) play vital roles in controlling immune reactions and maintaining immune tolerance in the body. The targeted destruction of epidermal melanocytes by activated CD8+T cells is a key event in the development of vitiligo. However, Tregs may exert immunosuppressive effects on CD8+T cells, which could be beneficial in treating vitiligo. METHODS A comprehensive search of PubMed and Web of Science was conducted to gather information on Tregs and vitiligo. RESULTS In vitiligo, there is a decrease in Treg numbers and impaired Treg functions, along with potential damage to Treg-related signaling pathways. Increasing Treg numbers and enhancing Treg function could lead to immunosuppressive effects on CD8+T cells. Recent research progress on Tregs in vitiligo has been summarized, highlighting various Treg-related therapies being investigated for clinical use. The current status of Treg-related therapeutic strategies and potential future directions for vitiligo treatment are also discussed. CONCLUSIONS A deeper understanding of Tregs will be crucial for advancing Treg-related drug discovery and treatment development in vitiligo.
Collapse
Affiliation(s)
- Shiyu Jin
- Hangzhou Third Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, 310009, China
| | - Sheng Wan
- Hangzhou Third Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, 310009, China
- Department of Dermatology, Hangzhou Third People's Hospital, Hangzhou, 310009, China
| | - Renxue Xiong
- Hangzhou Third Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, 310009, China
- Department of Dermatology, Hangzhou Third People's Hospital, Hangzhou, 310009, China
| | - Yujie Li
- Hangzhou Third Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, 310009, China
| | - Tingru Dong
- Hangzhou Third Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, 310009, China
| | - Cuiping Guan
- Hangzhou Third Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, 310009, China.
- Department of Dermatology, Hangzhou Third People's Hospital, Hangzhou, 310009, China.
| |
Collapse
|
40
|
Segbefia SP, Asandem DA, Pobee A, Asare B, Prah AD, Baba‐Adam R, Amponsah JA, Kyei‐Baafour E, van der Puije W, Osei F, Teye‐Adjei D, Agyemang S, Brenko T, Bentum‐Ennin L, Tetteh JKA, Bonney KJH, Sakyi SA, Amoah LE, Kusi KA. Expression patterns of immune checkpoint proteins and Plasmodium falciparum-induced cytokines in chronic hepatitis B virus-infected and uninfected individuals: A cross-sectional study. Health Sci Rep 2024; 7:e2280. [PMID: 39086506 PMCID: PMC11286663 DOI: 10.1002/hsr2.2280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 06/19/2024] [Accepted: 07/15/2024] [Indexed: 08/02/2024] Open
Abstract
Background and Aim Chronic hepatitis B virus (CHB) infection remains a major public health problem. The American Association for the Study of Liver Diseases (AASLD) 2018 Hepatitis B Guidelines provide that CHB individuals not requiring antiviral therapy yet are monitored to determine the need for antiviral therapy in the future; however, these tests do not include measurement of cytokines and immune cell characterization. This case-control study compared the cytokine and immune checkpoint protein expression profiles between CHB individuals not yet on antiviral treatment and hepatitis B virus (HBV)-negative individuals. Methods CD4 and CD8 T cells from CHB and HBV-negative individuals were characterized for immune checkpoint proteins programmed cell death-1 (PD1), T cell Immunoglobulin domain and mucin domain-containing protein 3 (TIM-3), and cytotoxic T lymphocyte-associated antigen 4 (CTLA-4) (CD152), and a memory marker CXCR3 (CD183) using flow cytometry. Malaria-induced cytokine expression levels were determined by stimulating their blood cells with Plasmodium falciparum 3D7 strain antigens (CSP, AMA-1, and TRAP) in whole blood assays, and cytokine levels were measured using a 13-plex Luminex kit. Results HBV-negative and CHB individuals had comparable levels of CD4+ and CD8+ T cells. However, a proportion of the CD4+ and CD8+ populations from both groups, which were CXCR3+, expressed PD-1 and CD152. The ability to produce cytokines in response to malaria antigen stimulation was not significantly different between the groups. Conclusion These findings support excluding CHB individuals from antiviral therapy at this stage of infection. However, CHB individuals require regular monitoring to determine the need for later antiviral treatment.
Collapse
Affiliation(s)
- Selorm P. Segbefia
- Department of Immunology, NMIMR, College of Health SciencesUniversity of GhanaAccraGhana
- Department of Molecular Medicine, School of Medicine and DentistryCollege of Health Sciences, KNUSTKumasiGhana
| | - Diana A. Asandem
- Department of Biochemistry, Cell and Molecular Biology, West African Centre for Cell Biology of Infectious Pathogens, College of Basic and Applied SciencesUniversity of GhanaAccraGhana
- Department of Virology, NMIMR, College of Health SciencesUniversity of GhanaAccraGhana
| | - Abigail Pobee
- Department of Immunology, NMIMR, College of Health SciencesUniversity of GhanaAccraGhana
| | - Bright Asare
- Department of Immunology, NMIMR, College of Health SciencesUniversity of GhanaAccraGhana
- Department of Animal Biology and Conservation Science, College of Basic and Applied SciencesUniversity of GhanaAccraGhana
| | - Ahu Diana Prah
- Department of Biochemistry, Cell and Molecular Biology, West African Centre for Cell Biology of Infectious Pathogens, College of Basic and Applied SciencesUniversity of GhanaAccraGhana
| | - Rawdat Baba‐Adam
- Department of Immunology, NMIMR, College of Health SciencesUniversity of GhanaAccraGhana
- Department of Biochemistry, Cell and Molecular Biology, West African Centre for Cell Biology of Infectious Pathogens, College of Basic and Applied SciencesUniversity of GhanaAccraGhana
| | - Jones Amo Amponsah
- Department of Immunology, NMIMR, College of Health SciencesUniversity of GhanaAccraGhana
| | - Eric Kyei‐Baafour
- Department of Immunology, NMIMR, College of Health SciencesUniversity of GhanaAccraGhana
| | - William van der Puije
- Department of Immunology, NMIMR, College of Health SciencesUniversity of GhanaAccraGhana
| | - Frank Osei
- Department of Immunology, NMIMR, College of Health SciencesUniversity of GhanaAccraGhana
| | - Doreen Teye‐Adjei
- Department of Immunology, NMIMR, College of Health SciencesUniversity of GhanaAccraGhana
| | - Seth Agyemang
- Department of Biochemistry, Cell and Molecular Biology, West African Centre for Cell Biology of Infectious Pathogens, College of Basic and Applied SciencesUniversity of GhanaAccraGhana
| | - Theophilus Brenko
- Department of Immunology, NMIMR, College of Health SciencesUniversity of GhanaAccraGhana
| | - Lutterodt Bentum‐Ennin
- Department of Immunology, NMIMR, College of Health SciencesUniversity of GhanaAccraGhana
| | - John K. A. Tetteh
- Department of Immunology, NMIMR, College of Health SciencesUniversity of GhanaAccraGhana
| | - Kofi J. H. Bonney
- Department of Virology, NMIMR, College of Health SciencesUniversity of GhanaAccraGhana
| | - Samuel Asamoah Sakyi
- Department of Molecular Medicine, School of Medicine and DentistryCollege of Health Sciences, KNUSTKumasiGhana
| | - Linda E. Amoah
- Department of Immunology, NMIMR, College of Health SciencesUniversity of GhanaAccraGhana
- Department of Biochemistry, Cell and Molecular Biology, West African Centre for Cell Biology of Infectious Pathogens, College of Basic and Applied SciencesUniversity of GhanaAccraGhana
| | - Kwadwo A. Kusi
- Department of Immunology, NMIMR, College of Health SciencesUniversity of GhanaAccraGhana
- Department of Biochemistry, Cell and Molecular Biology, West African Centre for Cell Biology of Infectious Pathogens, College of Basic and Applied SciencesUniversity of GhanaAccraGhana
| |
Collapse
|
41
|
Wang J, Zheng P, Yu J, Yang X, Zhang J. Rational design of small-sized peptidomimetic inhibitors disrupting protein-protein interaction. RSC Med Chem 2024; 15:2212-2225. [PMID: 39026653 PMCID: PMC11253864 DOI: 10.1039/d4md00202d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 05/04/2024] [Indexed: 07/20/2024] Open
Abstract
Protein-protein interactions are fundamental to nearly all biological processes. Due to their structural flexibility, peptides have emerged as promising candidates for developing inhibitors targeting large and planar PPI interfaces. However, their limited drug-like properties pose challenges. Hence, rational modifications based on peptide structures are anticipated to expedite the innovation of peptide-based therapeutics. This review comprehensively examines the design strategies for developing small-sized peptidomimetic inhibitors targeting PPI interfaces, which predominantly encompass two primary categories: peptidomimetics with abbreviated sequences and low molecular weights and peptidomimetics mimicking secondary structural conformations. We have also meticulously detailed several instances of designing and optimizing small-sized peptidomimetics targeting PPIs, including MLL1-WDR5, PD-1/PD-L1, and Bak/Bcl-xL, among others, to elucidate the potential application prospects of these design strategies. Hopefully, this review will provide valuable insights and inspiration for the future development of PPI small-sized peptidomimetic inhibitors in pharmaceutical research endeavors.
Collapse
Affiliation(s)
- Junyuan Wang
- School of Pharmacy, Ningxia Medical University Yinchuan 750004 China
| | - Ping Zheng
- School of Pharmacy, Ningxia Medical University Yinchuan 750004 China
| | - Jianqiang Yu
- School of Pharmacy, Ningxia Medical University Yinchuan 750004 China
| | - Xiuyan Yang
- Medicinal Chemistry and Bioinformatics Center, School of Medicine, Shanghai Jiao Tong University Shanghai 200025 China
| | - Jian Zhang
- Key Laboratory of Protection, Development and Utilization of Medicinal Resources in Liupanshan Area, Ministry of Education, Peptide & Protein Drug Research Center, School of Pharmacy, Ningxia Medical University Yinchuan 750004 China
| |
Collapse
|
42
|
Katsuumi G, Shimizu I, Suda M, Yoshida Y, Furihata T, Joki Y, Hsiao CL, Jiaqi L, Fujiki S, Abe M, Sugimoto M, Soga T, Minamino T. SGLT2 inhibition eliminates senescent cells and alleviates pathological aging. NATURE AGING 2024; 4:926-938. [PMID: 38816549 PMCID: PMC11257941 DOI: 10.1038/s43587-024-00642-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 05/02/2024] [Indexed: 06/01/2024]
Abstract
It has been reported that accumulation of senescent cells in various tissues contributes to pathological aging and that elimination of senescent cells (senolysis) improves age-associated pathologies. Here, we demonstrate that inhibition of sodium-glucose co-transporter 2 (SGLT2) enhances clearance of senescent cells, thereby ameliorating age-associated phenotypic changes. In a mouse model of dietary obesity, short-term treatment with the SGLT2 inhibitor canagliflozin reduced the senescence load in visceral adipose tissue and improved adipose tissue inflammation and metabolic dysfunction, but normalization of plasma glucose by insulin treatment had no effect on senescent cells. Canagliflozin extended the lifespan of mice with premature aging even when treatment was started in middle age. Metabolomic analyses revealed that short-term treatment with canagliflozin upregulated 5-aminoimidazole-4-carboxamide-1-β-D-ribofuranoside, enhancing immune-mediated clearance of senescent cells by downregulating expression of programmed cell death-ligand 1. These findings suggest that inhibition of SGLT2 has an indirect senolytic effect by enhancing endogenous immunosurveillance of senescent cells.
Collapse
Affiliation(s)
- Goro Katsuumi
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
- Department of Cardiovascular Medicine, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Ippei Shimizu
- Department of Cardiovascular Aging, National Cerebral and Cardiovascular Center Research Institute, Osaka, Japan
- Department of Cardiovascular Medicine, National Cerebral and Cardiovascular Center, Osaka, Japan
| | - Masayoshi Suda
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - Yohko Yoshida
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
- Department of Advanced Senotherapeutics, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Takaaki Furihata
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Yusuke Joki
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Chieh-Lun Hsiao
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Liang Jiaqi
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Shinya Fujiki
- Department of Cardiovascular Medicine, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Manabu Abe
- Department of Animal Model Development, Brain Research Institute, Niigata University, Niigata, Japan
| | - Masataka Sugimoto
- Molecular and Cellular Aging, Tokyo Metropolitan Institute for Geriatrics and Gerontology, Tokyo, Japan
| | - Tomoyoshi Soga
- Institute for Advanced Biosciences, Keio University, Yamagata, Japan
- Human Biology-Microbiome-Quantum Research Center (WPI-Bio2Q), Keio University, Tokyo, Japan
| | - Tohru Minamino
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan.
- Japan Agency for Medical Research and Development-Core Research for Evolutionary Medical Science and Technology (AMED-CREST), Japan Agency for Medical Research and Development, Tokyo, Japan.
| |
Collapse
|
43
|
Papachristos AJ, Serrao-Brown H, Gill AJ, Clifton-Bligh R, Sidhu SB. Medullary Thyroid Cancer: Molecular Drivers and Immune Cellular Milieu of the Tumour Microenvironment-Implications for Systemic Treatment. Cancers (Basel) 2024; 16:2296. [PMID: 39001359 PMCID: PMC11240419 DOI: 10.3390/cancers16132296] [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: 05/12/2024] [Revised: 06/16/2024] [Accepted: 06/20/2024] [Indexed: 07/16/2024] Open
Abstract
In this review, we explore the underlying molecular biology of medullary thyroid carcinoma (MTC) and its interplay with the host immune system. MTC is consistently driven by a small number of specific pathogenic variants, beyond which few additional genetic events are required for tumorigenesis. This explains the exceedingly low tumour mutational burden seen in most MTC, in contrast to other cancers. However, because of the low tumour mutational burden (TMB), there is a correspondingly low level of tumour-associated neoantigens that are presented to the host immune system. This reduces tumour visibility and vigour of the anti-tumour immune response and suggests the efficacy of immunotherapy in MTC is likely to be poor, acknowledging this inference is largely based on the extrapolation of data from other tumour types. The dominance of specific RET (REarranged during Transfection) pathogenic variants in MTC tumorigenesis rationalizes the observed efficacy of the targeted RET-specific tyrosine kinase inhibitors (TKIs) in comparison to multi-kinase inhibitors (MKIs). Therapeutic durability of pathway inhibitors is an ongoing research focus. It may be limited by the selection pressure TKI treatment creates, promoting survival of resistant tumour cell clones that can escape pathway inhibition through binding-site mutations, activation of alternate pathways, and modulation of the cellular and cytokine milieu of the tumour microenvironment (TME).
Collapse
Affiliation(s)
- Alexander J Papachristos
- Northern Clinical School, Sydney Medical School, Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2006, Australia
- Endocrine Surgical Unit, Royal North Shore Hospital, Sydney, NSW 2065, Australia
| | - Hazel Serrao-Brown
- Endocrine Surgical Unit, Royal North Shore Hospital, Sydney, NSW 2065, Australia
| | - Anthony J Gill
- Northern Clinical School, Sydney Medical School, Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2006, Australia
- NSW Health Pathology, Department of Anatomical Pathology, Royal North Shore Hospital, Sydney, NSW 2065, Australia
- Cancer Diagnosis and Pathology Group, Kolling Institute of Medical Research, Royal North Shore Hospital, Sydney, NSW 2065, Australia
| | - Roderick Clifton-Bligh
- Cancer Diagnosis and Pathology Group, Kolling Institute of Medical Research, Royal North Shore Hospital, Sydney, NSW 2065, Australia
- Department of Endocrinology, Royal North Shore Hospital, Sydney, NSW 2065, Australia
| | - Stanley B Sidhu
- Northern Clinical School, Sydney Medical School, Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2006, Australia
- Endocrine Surgical Unit, Royal North Shore Hospital, Sydney, NSW 2065, Australia
| |
Collapse
|
44
|
Kuznetsova AV, Glukhova XA, Popova OP, Beletsky IP, Ivanov AA. Contemporary Approaches to Immunotherapy of Solid Tumors. Cancers (Basel) 2024; 16:2270. [PMID: 38927974 PMCID: PMC11201544 DOI: 10.3390/cancers16122270] [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: 05/28/2024] [Revised: 06/11/2024] [Accepted: 06/15/2024] [Indexed: 06/28/2024] Open
Abstract
In recent years, the arrival of the immunotherapy industry has introduced the possibility of providing transformative, durable, and potentially curative outcomes for various forms of malignancies. However, further research has shown that there are a number of issues that significantly reduce the effectiveness of immunotherapy, especially in solid tumors. First of all, these problems are related to the protective mechanisms of the tumor and its microenvironment. Currently, major efforts are focused on overcoming protective mechanisms by using different adoptive cell therapy variants and modifications of genetically engineered constructs. In addition, a complex workforce is required to develop and implement these treatments. To overcome these significant challenges, innovative strategies and approaches are necessary to engineer more powerful variations of immunotherapy with improved antitumor activity and decreased toxicity. In this review, we discuss recent innovations in immunotherapy aimed at improving clinical efficacy in solid tumors, as well as strategies to overcome the limitations of various immunotherapies.
Collapse
Affiliation(s)
- Alla V. Kuznetsova
- Laboratory of Molecular and Cellular Pathology, Russian University of Medicine (Formerly A.I. Evdokimov Moscow State University of Medicine and Dentistry), Ministry of Health of the Russian Federation, Bld 4, Dolgorukovskaya Str, 1127006 Moscow, Russia; (A.V.K.); (O.P.P.)
- Koltzov Institute of Developmental Biology, Russian Academy of Sciences, 26 Vavilov Street, 119334 Moscow, Russia
| | - Xenia A. Glukhova
- Onni Biotechnologies Ltd., Aalto University Campus, Metallimiehenkuja 10, 02150 Espoo, Finland; (X.A.G.); (I.P.B.)
| | - Olga P. Popova
- Laboratory of Molecular and Cellular Pathology, Russian University of Medicine (Formerly A.I. Evdokimov Moscow State University of Medicine and Dentistry), Ministry of Health of the Russian Federation, Bld 4, Dolgorukovskaya Str, 1127006 Moscow, Russia; (A.V.K.); (O.P.P.)
| | - Igor P. Beletsky
- Onni Biotechnologies Ltd., Aalto University Campus, Metallimiehenkuja 10, 02150 Espoo, Finland; (X.A.G.); (I.P.B.)
| | - Alexey A. Ivanov
- Laboratory of Molecular and Cellular Pathology, Russian University of Medicine (Formerly A.I. Evdokimov Moscow State University of Medicine and Dentistry), Ministry of Health of the Russian Federation, Bld 4, Dolgorukovskaya Str, 1127006 Moscow, Russia; (A.V.K.); (O.P.P.)
| |
Collapse
|
45
|
Zhu ZJ, Teng M, Liu Y, Chen FJ, Yao Y, Li EZ, Luo J. Immune escape of avian oncogenic Marek's disease herpesvirus and antagonistic host immune responses. NPJ Vaccines 2024; 9:109. [PMID: 38879650 PMCID: PMC11180173 DOI: 10.1038/s41541-024-00905-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 06/07/2024] [Indexed: 06/19/2024] Open
Abstract
Marek's disease virus (MDV) is a highly pathogenic and oncogenic alpha herpesvirus that causes Marek's disease (MD), which is one of the most important immunosuppressive and rapid-onset neoplastic diseases in poultry. The onset of MD lymphomas and other clinical diseases can be efficiently prevented by vaccination; these vaccines are heralded as the first demonstration of a successful vaccination strategy against a cancer. However, the persistent evolution of epidemic MDV strains towards greater virulence has recently resulted in frequent outbreaks of MD in vaccinated chicken flocks worldwide. Herein, we provide an overall review focusing on the discovery and identification of the strategies by which MDV evades host immunity and attacks the immune system. We have also highlighted the decrease in the immune efficacy of current MD vaccines. The prospects, strategies and new techniques for the development of efficient MD vaccines, together with the possibilities of antiviral therapy in MD, are also discussed.
Collapse
Affiliation(s)
- Zhi-Jian Zhu
- College of Biological and Food Engineering & Affiliated Central Hospital, Huanghuai University, Zhumadian, 463000, People's Republic of China
- Institute for Animal Health & UK-China Center of Excellence for Research on Avian Disease, Henan Academy of Agricultural Sciences, Zhengzhou, 450002, People's Republic of China
- Henan Provincial Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences, Zhengzhou, 450002, People's Republic of China
- Key Laboratory of Animal Immunology, Ministry of Agriculture and Rural Affairs of the People's Republic of China, Zhengzhou, 450002, People's Republic of China
| | - Man Teng
- Institute for Animal Health & UK-China Center of Excellence for Research on Avian Disease, Henan Academy of Agricultural Sciences, Zhengzhou, 450002, People's Republic of China
- Henan Provincial Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences, Zhengzhou, 450002, People's Republic of China
- Key Laboratory of Animal Immunology, Ministry of Agriculture and Rural Affairs of the People's Republic of China, Zhengzhou, 450002, People's Republic of China
| | - Yu Liu
- College of Biological and Food Engineering & Affiliated Central Hospital, Huanghuai University, Zhumadian, 463000, People's Republic of China
| | - Fu-Jia Chen
- College of Biological and Food Engineering & Affiliated Central Hospital, Huanghuai University, Zhumadian, 463000, People's Republic of China
| | - Yongxiu Yao
- The Pirbright Institute & UK-China Centre of Excellence for Research on Avian Diseases, Pirbright, Ash Road, Guildford, Surrey, GU24 0NF, UK
| | - En-Zhong Li
- College of Biological and Food Engineering & Affiliated Central Hospital, Huanghuai University, Zhumadian, 463000, People's Republic of China.
| | - Jun Luo
- Institute for Animal Health & UK-China Center of Excellence for Research on Avian Disease, Henan Academy of Agricultural Sciences, Zhengzhou, 450002, People's Republic of China.
- Henan Provincial Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences, Zhengzhou, 450002, People's Republic of China.
- Key Laboratory of Animal Immunology, Ministry of Agriculture and Rural Affairs of the People's Republic of China, Zhengzhou, 450002, People's Republic of China.
- Laboratory of Functional Microbiology and Animal Health, College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, 471003, People's Republic of China.
- Longhu Laboratory, Zhengzhou, 450046, People's Republic of China.
| |
Collapse
|
46
|
Hazra R, Chattopadhyay S, Mallick A, Gayen S, Roy S. Revealing the therapeutic properties of gut microbiota: transforming cancer immunotherapy from basic to clinical approaches. Med Oncol 2024; 41:175. [PMID: 38874788 DOI: 10.1007/s12032-024-02416-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 05/25/2024] [Indexed: 06/15/2024]
Abstract
The immune system plays a pivotal role in the battle against cancer, serving as a formidable guardian in the ongoing fight against malignant cells. To combat these malignant cells, immunotherapy has emerged as a prevalent approach leveraging antibodies and peptides such as anti-PD-1, anti-PD-L1, and anti-CTLA-4 to inhibit immune checkpoints and activate T lymphocytes. The optimization of gut microbiota plays a significant role in modulating the defense system in the body. This study explores the potential of certain gut-resident bacteria to amplify the impact of immunotherapy. Contemporary antibiotic treatments, which can impair gut flora, may diminish the efficacy of immune checkpoint blockers. Conversely, probiotics or fecal microbiota transplantation can help re-establish intestinal microflora equilibrium. Additionally, the gut microbiome has been implicated in various strategies to counteract immune resistance, thereby enhancing the success of cancer immunotherapy. This paper also acknowledges cutting-edge technologies such as nanotechnology, CAR-T therapy, ACT therapy, and oncolytic viruses in modulating gut microbiota. Thus, an exhaustive review of literature was performed to uncover the elusive link that could potentiate the gut microbiome's role in augmenting the success of cancer immunotherapy.
Collapse
Affiliation(s)
- Rudradeep Hazra
- Department of Pharmaceutical Technology, Kolkata-Group of Institutions, NSHM Knowledge Campus, 124, B. L. Saha Road, Tara Park, Behala, Kolkata, West Bengal, 700053, India
| | - Soumyadeep Chattopadhyay
- Department of Pharmaceutical Technology, Kolkata-Group of Institutions, NSHM Knowledge Campus, 124, B. L. Saha Road, Tara Park, Behala, Kolkata, West Bengal, 700053, India
| | - Arijit Mallick
- Department of Pharmaceutical Technology, Kolkata-Group of Institutions, NSHM Knowledge Campus, 124, B. L. Saha Road, Tara Park, Behala, Kolkata, West Bengal, 700053, India
| | - Sakuntala Gayen
- Department of Pharmaceutical Technology, Kolkata-Group of Institutions, NSHM Knowledge Campus, 124, B. L. Saha Road, Tara Park, Behala, Kolkata, West Bengal, 700053, India
| | - Souvik Roy
- Department of Pharmaceutical Technology, Kolkata-Group of Institutions, NSHM Knowledge Campus, 124, B. L. Saha Road, Tara Park, Behala, Kolkata, West Bengal, 700053, India.
| |
Collapse
|
47
|
Li Z, Xiong W, Liang Z, Wang J, Zeng Z, Kołat D, Li X, Zhou D, Xu X, Zhao L. Critical role of the gut microbiota in immune responses and cancer immunotherapy. J Hematol Oncol 2024; 17:33. [PMID: 38745196 PMCID: PMC11094969 DOI: 10.1186/s13045-024-01541-w] [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: 10/25/2023] [Accepted: 04/03/2024] [Indexed: 05/16/2024] Open
Abstract
The gut microbiota plays a critical role in the progression of human diseases, especially cancer. In recent decades, there has been accumulating evidence of the connections between the gut microbiota and cancer immunotherapy. Therefore, understanding the functional role of the gut microbiota in regulating immune responses to cancer immunotherapy is crucial for developing precision medicine. In this review, we extract insights from state-of-the-art research to decipher the complicated crosstalk among the gut microbiota, the systemic immune system, and immunotherapy in the context of cancer. Additionally, as the gut microbiota can account for immune-related adverse events, we discuss potential interventions to minimize these adverse effects and discuss the clinical application of five microbiota-targeted strategies that precisely increase the efficacy of cancer immunotherapy. Finally, as the gut microbiota holds promising potential as a target for precision cancer immunotherapeutics, we summarize current challenges and provide a general outlook on future directions in this field.
Collapse
Affiliation(s)
- Zehua Li
- Department of Plastic and Burn Surgery, West China Hospital, Sichuan University, Chengdu, China
- Chinese Academy of Medical Sciences (CAMS), CAMS Oxford Institute (COI), Nuffield Department of Medicine, University of Oxford, Oxford, England
| | - Weixi Xiong
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, China
- Institute of Brain Science and Brain-Inspired Technology of West China Hospital, Sichuan University, Chengdu, China
| | - Zhu Liang
- Chinese Academy of Medical Sciences (CAMS), CAMS Oxford Institute (COI), Nuffield Department of Medicine, University of Oxford, Oxford, England
- Target Discovery Institute, Center for Medicines Discovery, Nuffield Department of Medicine, University of Oxford, Oxford, England
| | - Jinyu Wang
- Departments of Obstetrics and Gynecology, West China Second University Hospital of Sichuan University, Chengdu, China
| | - Ziyi Zeng
- Department of Neonatology, West China Second University Hospital of Sichuan University, Chengdu, China
| | - Damian Kołat
- Department of Functional Genomics, Medical University of Lodz, Lodz, Poland
- Department of Biomedicine and Experimental Surgery, Medical University of Lodz, Lodz, Poland
| | - Xi Li
- Department of Urology, Churchill Hospital, Oxford University Hospitals NHS Foundation, Oxford, UK
| | - Dong Zhou
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, China
- Institute of Brain Science and Brain-Inspired Technology of West China Hospital, Sichuan University, Chengdu, China
| | - Xuewen Xu
- Department of Plastic and Burn Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Linyong Zhao
- Department of General Surgery and Gastric Cancer Center, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
48
|
Kashiwada T, Takano R, Ando F, Kuroda S, Miyabe Y, Owada R, Miyanaga A, Asatsuma-Okumura T, Hashiguchi M, Kanazawa Y, Yoshida H, Seike M, Gemma A, Iwai Y. Lysosomal degradation of PD-L1 is associated with immune-related adverse events during anti-PD-L1 immunotherapy in NSCLC patients. Front Pharmacol 2024; 15:1384733. [PMID: 38799168 PMCID: PMC11116720 DOI: 10.3389/fphar.2024.1384733] [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: 02/10/2024] [Accepted: 04/25/2024] [Indexed: 05/29/2024] Open
Abstract
Background: Immune checkpoint inhibitors (ICIs) can induce immune-related adverse events (irAEs). Liquid biomarkers to predict irAE occurrence are urgently needed. We previously developed an ELISA system to specifically detect soluble PD-L1 (sPD-L1) with PD-1-binding capacity (bsPD-L1). Here, we investigated the relationship between sPD-L1 and bsPD-L1 in gastric cancer (GC) and non-small cell lung cancer (NSCLC) treated with PD-1/PD-L1 blockade and their association with irAEs. Methods: We examined sPD-L1, bsPD-L1, matrix metalloproteinases (MMPs), and proinflammatory cytokine levels by ELISA in plasma samples from 117 GC patients prior to surgery and 72 NSCLC patients prior to and at 2 months after ICI treatment (anti-PD-1, n = 48; anti-PD-L1, n = 24). In mice treated with anti-PD-1/PD-L1 antibodies (Abs), sPD-L1 levels and localization of Abs were examined by ELISA and immunohistochemistry, respectively. Results:sPD-L1 was detected with higher frequency in GC patients than in NSCLC patients, whereas bsPD-L1 was detected with similar frequencies in GC and NSCLC patients. sPD-L1 levels were correlated with IL-1α, IL-1β, TNF-α, and IL-6 levels, while bsPD-L1 levels were correlated with MMP13, MMP3, and IFN-γ levels. In NSCLC patients, anti-PD-L1, but not anti-PD-1, treatment increased sPD-L1, which was associated with irAE development, but not with clinical outcomes. In mice, trafficking of anti-PD-L1 Abs to lysosomes in F4/80+ macrophages resulted in sPD-L1 production, which was suppressed by treatment with lysosomal degradation inhibitor chloroquine and macrophage depletion. Conclusion: Anti-PD-L1-mediated lysosomal degradation induces sPD-L1 production, which can serve as an indicator to predict irAE development during anti-PD-L1 treatment.
Collapse
Affiliation(s)
- Takeru Kashiwada
- Department of Pulmonary Medicine and Oncology, Nippon Medical School, Tokyo, Japan
| | - Ryotaro Takano
- Department of Cell Biology, Institute for Advanced Medical Sciences, Nippon Medical School, Tokyo, Japan
- Department of Gastrointestinal and Hepato-Biliary-Pancreatic Surgery, Nippon Medical School, Tokyo, Japan
| | - Fumihiko Ando
- Department of Cell Biology, Institute for Advanced Medical Sciences, Nippon Medical School, Tokyo, Japan
- Department of Gastrointestinal and Hepato-Biliary-Pancreatic Surgery, Nippon Medical School, Tokyo, Japan
| | - Shoko Kuroda
- Department of Cell Biology, Institute for Advanced Medical Sciences, Nippon Medical School, Tokyo, Japan
| | - Yoshishige Miyabe
- Department of Cell Biology, Institute for Advanced Medical Sciences, Nippon Medical School, Tokyo, Japan
- Department of Immunology and Parasitology, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Ryuji Owada
- Department of Cell Biology, Institute for Advanced Medical Sciences, Nippon Medical School, Tokyo, Japan
| | - Akihiko Miyanaga
- Department of Pulmonary Medicine and Oncology, Nippon Medical School, Tokyo, Japan
| | - Tomoko Asatsuma-Okumura
- Department of Cell Biology, Institute for Advanced Medical Sciences, Nippon Medical School, Tokyo, Japan
| | - Masaaki Hashiguchi
- Department of Cell Biology, Institute for Advanced Medical Sciences, Nippon Medical School, Tokyo, Japan
| | - Yoshikazu Kanazawa
- Department of Gastrointestinal and Hepato-Biliary-Pancreatic Surgery, Nippon Medical School, Tokyo, Japan
| | - Hiroshi Yoshida
- Department of Gastrointestinal and Hepato-Biliary-Pancreatic Surgery, Nippon Medical School, Tokyo, Japan
| | - Masahiro Seike
- Department of Pulmonary Medicine and Oncology, Nippon Medical School, Tokyo, Japan
| | - Akihiko Gemma
- Department of Pulmonary Medicine and Oncology, Nippon Medical School, Tokyo, Japan
| | - Yoshiko Iwai
- Department of Cell Biology, Institute for Advanced Medical Sciences, Nippon Medical School, Tokyo, Japan
| |
Collapse
|
49
|
Ando F, Kashiwada T, Kuroda S, Fujii T, Takano R, Miyabe Y, Kunugi S, Sakatani T, Miyanaga A, Asatsuma-Okumura T, Hashiguchi M, Kanazawa Y, Ohashi R, Yoshida H, Seike M, Gemma A, Iwai Y. Combination of plasma MMPs and PD-1-binding soluble PD-L1 predicts recurrence in gastric cancer and the efficacy of immune checkpoint inhibitors in non-small cell lung cancer. Front Pharmacol 2024; 15:1384731. [PMID: 38774209 PMCID: PMC11106465 DOI: 10.3389/fphar.2024.1384731] [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: 02/10/2024] [Accepted: 04/22/2024] [Indexed: 05/24/2024] Open
Abstract
Background The tumor microenvironment (TME) impacts the therapeutic efficacy of immune checkpoint inhibitors (ICIs). No liquid biomarkers are available to evaluate TME heterogeneity. Here, we investigated the clinical significance of PD-1-binding soluble PD-L1 (bsPD-L1) in gastric cancer (GC) patients and non-small cell lung cancer (NSCLC) patients treated with PD-1/PD-L1 blockade. Methods We examined bsPD-L1, matrix metalloproteinases (MMPs), and IFN-γ levels in plasma samples from GC patients (n = 117) prior to surgery and NSCLC patients (n = 72) prior to and 2 months after ICI treatment. We also examined extracellular matrix (ECM) integrity, PD-L1 expression, and T cell infiltration in tumor tissues from 25 GC patients by Elastica Masson-Goldner staining and immunohistochemical staining for PD-L1 and CD3, respectively. Results bsPD-L1 was detected in 17/117 GC patients and 16/72 NSCLC patients. bsPD-L1 showed strong or moderate correlations with plasma MMP13 or MMP3 levels, respectively, in both GC and NSCLC patients. bsPD-L1 expression in GC was associated with IFN-γ levels and intra-tumoral T cell infiltration, whereas MMP13 levels were associated with loss of ECM integrity, allowing tumor cells to access blood vessels. Plasma MMP3 and MMP13 levels were altered during ICI treatment. Combined bsPD-L1 and MMP status had higher predictive accuracy to identify two patient groups with favorable and poor prognosis than tumor PD-L1 expression: bsPD-L1+MMP13high in GC and bsPD-L1+(MMP3 and MMP13)increased in NSCLC were associated with poor prognosis, whereas bsPD-L1+MMP13low in GC and bsPD-L1+(MMP3 or MMP13)decreased in NSCLC were associated with favorable prognosis. Conclusion Plasma bsPD-L1 and MMP13 levels indicate T cell response and loss of ECM integrity, respectively, in the TME. The combination of bsPD-L1 and MMPs may represent a non-invasive tool to predict recurrence in GC and the efficacy of ICIs in NSCLC.
Collapse
Affiliation(s)
- Fumihiko Ando
- Department of Cell Biology, Institute for Advanced Medical Sciences, Nippon Medical School, Tokyo, Japan
- Department of Gastrointestinal and Hepato-Biliary-Pancreatic Surgery, Nippon Medical School, Tokyo, Japan
| | - Takeru Kashiwada
- Department of Pulmonary Medicine and Oncology, Nippon Medical School, Tokyo, Japan
| | - Shoko Kuroda
- Department of Cell Biology, Institute for Advanced Medical Sciences, Nippon Medical School, Tokyo, Japan
| | - Takenori Fujii
- Department of Integrated Diagnostic Pathology, Nippon Medical School, Tokyo, Japan
| | - Ryotaro Takano
- Department of Cell Biology, Institute for Advanced Medical Sciences, Nippon Medical School, Tokyo, Japan
- Department of Gastrointestinal and Hepato-Biliary-Pancreatic Surgery, Nippon Medical School, Tokyo, Japan
| | - Yoshishige Miyabe
- Department of Cell Biology, Institute for Advanced Medical Sciences, Nippon Medical School, Tokyo, Japan
- Department of Immunology and Parasitology, St. Marianna University School of Medicine, Kawasaki, Kanagawa, Japan
| | - Shinobu Kunugi
- Department of Analytic Human Pathology, Nippon Medical School, Tokyo, Japan
| | - Takashi Sakatani
- Department of Integrated Diagnostic Pathology, Nippon Medical School, Tokyo, Japan
| | - Akihiko Miyanaga
- Department of Pulmonary Medicine and Oncology, Nippon Medical School, Tokyo, Japan
| | - Tomoko Asatsuma-Okumura
- Department of Cell Biology, Institute for Advanced Medical Sciences, Nippon Medical School, Tokyo, Japan
| | - Masaaki Hashiguchi
- Department of Cell Biology, Institute for Advanced Medical Sciences, Nippon Medical School, Tokyo, Japan
| | - Yoshikazu Kanazawa
- Department of Gastrointestinal and Hepato-Biliary-Pancreatic Surgery, Nippon Medical School, Tokyo, Japan
| | - Ryuji Ohashi
- Department of Integrated Diagnostic Pathology, Nippon Medical School, Tokyo, Japan
| | - Hiroshi Yoshida
- Department of Gastrointestinal and Hepato-Biliary-Pancreatic Surgery, Nippon Medical School, Tokyo, Japan
| | - Masahiro Seike
- Department of Pulmonary Medicine and Oncology, Nippon Medical School, Tokyo, Japan
| | - Akihiko Gemma
- Department of Pulmonary Medicine and Oncology, Nippon Medical School, Tokyo, Japan
| | - Yoshiko Iwai
- Department of Cell Biology, Institute for Advanced Medical Sciences, Nippon Medical School, Tokyo, Japan
| |
Collapse
|
50
|
Stoeltje L, Luc JK, Haddad T, Schrankel CS. The roles of ABCB1/P-glycoprotein drug transporters in regulating gut microbes and inflammation: insights from animal models, old and new. Philos Trans R Soc Lond B Biol Sci 2024; 379:20230074. [PMID: 38497255 PMCID: PMC10945405 DOI: 10.1098/rstb.2023.0074] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 02/13/2024] [Indexed: 03/19/2024] Open
Abstract
Commensal enteric bacteria have evolved systems that enable growth in the ecologic niche of the host gastrointestinal tract. Animals evolved parallel mechanisms to survive the constant exposure to bacteria and their metabolic by-products. We propose that drug transporters encompass a crucial system to managing the gut microbiome. Drug transporters are present in the apical surface of gut epithelia. They detoxify cells from small molecules and toxins (xenobiotics) in the lumen. Here, we review what is known about commensal structure in the absence of the transporter ABCB1/P-glycoprotein in mammalian models. Knockout or low-activity alleles of ABCB1 lead to dysbiosis, Crohn's disease and ulcerative colitis in mammals. However, the exact function of ABCB1 in these contexts remain unclear. We highlight emerging models-the zebrafish Danio rerio and sea urchin Lytechinus pictus-that are poised to help dissect the fundamental mechanisms of ATP-binding cassette (ABC) transporters in the tolerance of commensal and pathogenic communities in the gut. We and others hypothesize that ABCB1 plays a direct role in exporting inflammatory bacterial products from host epithelia. Interdisciplinary work in this research area will lend novel insight to the transporter-mediated pathways that impact microbiome community structure and accelerate the pathogenesis of inflammatory bowel disease when perturbed. This article is part of the theme issue 'Sculpting the microbiome: how host factors determine and respond to microbial colonization'.
Collapse
Affiliation(s)
- Lauren Stoeltje
- Department of Biology, San Diego State University, 5500 Campanile Drive, Life Sciences North, Room 321, San Diego, CA 92182, USA
| | - Jenna K. Luc
- Department of Biology, San Diego State University, 5500 Campanile Drive, Life Sciences North, Room 321, San Diego, CA 92182, USA
| | - Timothaus Haddad
- Department of Biology, San Diego State University, 5500 Campanile Drive, Life Sciences North, Room 321, San Diego, CA 92182, USA
| | - Catherine S. Schrankel
- Department of Biology, San Diego State University, 5500 Campanile Drive, Life Sciences North, Room 321, San Diego, CA 92182, USA
| |
Collapse
|