51
|
Wang W, Lopez McDonald MC, Kim C, Ma M, Pan Z(T, Kaufmann C, Frank DA. The complementary roles of STAT3 and STAT1 in cancer biology: insights into tumor pathogenesis and therapeutic strategies. Front Immunol 2023; 14:1265818. [PMID: 38022653 PMCID: PMC10663227 DOI: 10.3389/fimmu.2023.1265818] [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: 07/23/2023] [Accepted: 10/23/2023] [Indexed: 12/01/2023] Open
Abstract
STATs are a family of transcription factors that regulate many critical cellular processes such as proliferation, apoptosis, and differentiation. Dysregulation of STATs is frequently observed in tumors and can directly drive cancer pathogenesis. STAT1 and STAT3 are generally viewed as mediating opposite roles in cancer development, with STAT1 suppressing tumorigenesis and STAT3 promoting oncogenesis. In this review, we investigate the specific roles of STAT1 and STAT3 in normal physiology and cancer biology, explore their interactions with each other, and offer insights into therapeutic strategies through modulating their transcriptional activity.
Collapse
Affiliation(s)
| | | | | | | | | | | | - David A. Frank
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, United States
| |
Collapse
|
52
|
Guo Y, Tong C, Wu Z, Lu Y, Wang Y, Han W. Reciprocal activation of antigen-presenting cells and CAR T cells triggers a widespread endogenous anti-tumor immune response through sustained high-level IFNγ production. Cancer Biol Med 2023; 20:j.issn.2095-3941.2023.0324. [PMID: 37929324 PMCID: PMC10690879 DOI: 10.20892/j.issn.2095-3941.2023.0324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 10/10/2023] [Indexed: 11/07/2023] Open
Affiliation(s)
- Yelei Guo
- Department of Bio-therapeutic, the First Medical Center, Chinese PLA General Hospital, Beijing 100853, China
| | - Chuan Tong
- Department of Bio-therapeutic, the First Medical Center, Chinese PLA General Hospital, Beijing 100853, China
| | - Zhiqiang Wu
- Department of Bio-therapeutic, the First Medical Center, Chinese PLA General Hospital, Beijing 100853, China
| | - Yuting Lu
- Department of Bio-therapeutic, the First Medical Center, Chinese PLA General Hospital, Beijing 100853, China
| | - Yao Wang
- Department of Bio-therapeutic, the First Medical Center, Chinese PLA General Hospital, Beijing 100853, China
| | - Weidong Han
- Department of Bio-therapeutic, the First Medical Center, Chinese PLA General Hospital, Beijing 100853, China
| |
Collapse
|
53
|
Suzuki K, Ohe R, Kabasawa T, Kitaoka T, Kawai M, Motoi F, Futakuchi M. Histological spatial analysis on the induction of PD-L1 + macrophages by CD8 + T cells at the marginal microenvironment of triple-negative breast cancer. Breast Cancer 2023; 30:1094-1104. [PMID: 37792212 PMCID: PMC10587303 DOI: 10.1007/s12282-023-01507-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 09/24/2023] [Indexed: 10/05/2023]
Abstract
BACKGROUND Programmed death-ligand 1 (PD-L1) plays important roles in the evasion of antitumor immunity. Because we observed the localization of PD-L1-positive (PD-L1+) cells in the marginal region of triple-negative breast cancer (TNBC) specimens, we hypothesized that the marginal microenvironment of TNBC would involve the induction of PD-L1+ cells. METHODS One hundred and one TNBC surgical specimens were examined. We performed immunohistochemical (IHC) studies of PD-L1, CD68, CD8, and pan-cytokeratin in these specimens. We analyzed the localization of IHC-positive cells and the distance between these cells by histological spatial analysis. RESULTS In 30.7% of TNBC specimens, PD-L1+ cells were located in the marginal region. Approximately three PD-L1+ cells accumulated around a single TNBC cell. Most PD-L1+ cells were located within 50 μm of TNBC cells. PD-L1+ cells were indicated to interact with TNBC cells in the marginal region. PD-L1+CD68+ cells were located in the marginal region, while CD68+ macrophages (MΦs) were observed either in the marginal region or the core region. PD-L1 expression in MΦs was induced in the marginal region. The colocalization of CD8+ T cells in the marginal region indicates that PD-L1 expression in MΦs would be induced by interaction with CD8+ T cells. Because CD8+ T cells are positive for CCL2, CCL2 may induce PD-L1 expression in MΦs. CONCLUSION At the marginal microenvironment of TNBC, PD-L1 expression would be induced in MΦs by interaction with CD8+ T cells through CCL2. The interaction between PD-L1+ MΦs and TNBC cells would facilitate the growth of TNBC under antitumor immunity. These interactions would be potential targets for restoring antitumor immunity and suppressing TNBC progression.
Collapse
Affiliation(s)
- Kazushi Suzuki
- Department of Pathology, Faculty of Medicine, Yamagata University, 2-2-2 Iida-Nishi, Yamagata, 990-9585, Japan.
| | - Rintaro Ohe
- Department of Pathology, Faculty of Medicine, Yamagata University, 2-2-2 Iida-Nishi, Yamagata, 990-9585, Japan
| | - Takanobu Kabasawa
- Department of Pathology, Faculty of Medicine, Yamagata University, 2-2-2 Iida-Nishi, Yamagata, 990-9585, Japan
| | - Takumi Kitaoka
- Department of Pathology, Faculty of Medicine, Yamagata University, 2-2-2 Iida-Nishi, Yamagata, 990-9585, Japan
| | - Masaaki Kawai
- Department of Surgery 1, Faculty of Medicine, Yamagata University, Yamagata, Japan
| | - Fuyuhiko Motoi
- Department of Surgery 1, Faculty of Medicine, Yamagata University, Yamagata, Japan
| | - Mitsuru Futakuchi
- Department of Pathology, Faculty of Medicine, Yamagata University, 2-2-2 Iida-Nishi, Yamagata, 990-9585, Japan
| |
Collapse
|
54
|
Wang S, Zhang G, Cui Q, Yang Y, Wang D, Liu A, Xia Y, Li W, Liu Y, Yu J. The DC-T cell axis is an effective target for the treatment of non-small cell lung cancer. Immun Inflamm Dis 2023; 11:e1099. [PMID: 38018578 PMCID: PMC10681037 DOI: 10.1002/iid3.1099] [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: 04/18/2023] [Revised: 11/06/2023] [Accepted: 11/09/2023] [Indexed: 11/30/2023] Open
Abstract
The dendritic cell (DC)-T cell axis is a bridge that connects innate and adaptive immunities. The initial immune response against tumors is mainly induced by mature antigen-presenting DCs. Enhancing the crosstalk between DCs and T cells may be an effective approach to improve the immune response to non-small cell lung cancer (NSCLC). In this article, a review was made of the interaction between DCs and T cells in the treatment of NSCLC and how this interaction affects the treatment outcome.
Collapse
Affiliation(s)
- Shuangcui Wang
- Department of OncologyFirst Teaching Hospital of Tianjin University of Traditional Chinese MedicineTianjinChina
- National Clinical Research Center for Chinese Medicine Acupuncture and MoxibustionTianjinChina
- Graduate SchoolTianjin University of Traditional Chinese MedicineTianjinChina
| | - Guan Zhang
- Department of OncologyFirst Teaching Hospital of Tianjin University of Traditional Chinese MedicineTianjinChina
- National Clinical Research Center for Chinese Medicine Acupuncture and MoxibustionTianjinChina
- Graduate SchoolTianjin University of Traditional Chinese MedicineTianjinChina
| | - Qian Cui
- Department of OncologyFirst Teaching Hospital of Tianjin University of Traditional Chinese MedicineTianjinChina
- National Clinical Research Center for Chinese Medicine Acupuncture and MoxibustionTianjinChina
- Graduate SchoolTianjin University of Traditional Chinese MedicineTianjinChina
| | - Yanjie Yang
- Department of OncologyFirst Teaching Hospital of Tianjin University of Traditional Chinese MedicineTianjinChina
- National Clinical Research Center for Chinese Medicine Acupuncture and MoxibustionTianjinChina
- Graduate SchoolTianjin University of Traditional Chinese MedicineTianjinChina
| | - Dong Wang
- Department of OncologyFirst Teaching Hospital of Tianjin University of Traditional Chinese MedicineTianjinChina
- National Clinical Research Center for Chinese Medicine Acupuncture and MoxibustionTianjinChina
- Graduate SchoolTianjin University of Traditional Chinese MedicineTianjinChina
| | - Aqing Liu
- Department of OncologyFirst Teaching Hospital of Tianjin University of Traditional Chinese MedicineTianjinChina
- National Clinical Research Center for Chinese Medicine Acupuncture and MoxibustionTianjinChina
- Graduate SchoolTianjin University of Traditional Chinese MedicineTianjinChina
| | - Ying Xia
- Department of OncologyFirst Teaching Hospital of Tianjin University of Traditional Chinese MedicineTianjinChina
- National Clinical Research Center for Chinese Medicine Acupuncture and MoxibustionTianjinChina
- Graduate SchoolTianjin University of Traditional Chinese MedicineTianjinChina
| | - Wentao Li
- Department of OncologyFirst Teaching Hospital of Tianjin University of Traditional Chinese MedicineTianjinChina
| | - Yunhe Liu
- Department of OncologyFirst Teaching Hospital of Tianjin University of Traditional Chinese MedicineTianjinChina
| | - Jianchun Yu
- Department of OncologyFirst Teaching Hospital of Tianjin University of Traditional Chinese MedicineTianjinChina
| |
Collapse
|
55
|
Monsrud AL, Avadhani V, Mosunjac MB, Flowers L, Krishnamurti U. Programmed Death Ligand-1 (PD-L1) Expression in Cervical Squamous Cell Carcinoma: Does it Correlate With Outcomes? Int J Gynecol Pathol 2023; 42:535-543. [PMID: 37562018 DOI: 10.1097/pgp.0000000000000975] [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: 08/12/2023]
Abstract
Cervical cancer is one of the most common gynecological malignancies. Upregulation of programmed death ligand-1 (PD-L1), an immunoregulatory protein, is associated with an adverse outcomes in several malignancies. Most studies evaluating PD-L1 expression in cervical squamous cell carcinoma (CSCC) lack data on outcomes. In this study, we correlate PD-L1 expression with clinicopathologic factors and clinical outcomes in invasive CSCC. Seventy-three cases of CSCC from 2010 to 2018 were immunostained for PD-L1. A combined positive score (CPS) of ≥1 and ≥10 was correlated with age, stage, and survival outcomes. Kaplan-Meier curves for progression-free survival and overall survival were plotted and compared using the log-rank test. Cox regression analysis was performed to identify significant prognostic factors (2-tailed P <0.05 was considered statistically significant). With CPS ≥1 or ≥10 as the cut-off, PD-L1 was positive in 52/73 (71.2%) and 23/73 (31.5%) of cases, respectively. PD-L1 positive patients present at a higher stage of disease, especially those with CPS ≥10. With CPS of ≥10 as the cut-off, the 5-yr progression-free survival and 5-yr overall survival were significantly lower ( P = 0.034 and 0.012, respectively). Only stage was statistically significant for worse overall survival on multivariate analysis. PD-L1 positive patients present at a higher stage of disease, and stage is an independent prognostic indicator for adverse outcomes in CSCC. This study highlights the potential of PD-L1 targeted therapy in patients with CSCC.
Collapse
|
56
|
Zhang X, Li R, Wang G. PDL1-Based Nomogram May Be of Potential Clinical Utility for Predicting Survival Outcome in Stage III Breast Cancer. BREAST CANCER (DOVE MEDICAL PRESS) 2023; 15:731-746. [PMID: 37905205 PMCID: PMC10613449 DOI: 10.2147/bctt.s435980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Accepted: 10/19/2023] [Indexed: 11/02/2023]
Abstract
Purpose Programmed cell death ligand 1 (PDL1) has the predictive and prognostic value in a great deal of cancers. This study aims to explore the expression of PDL1 in stage III breast cancer (BC) and its correlation with clinical outcome. Methods The protein expression of PDL1 in tumor tissues was determined by immunohistochemistry (IHC). The correlations between PDL1 and clinicopathological variables were performed by χ²-tests or Fisher's exact tests. The Cox proportional hazards model was used for univariate and multivariate analysis of the potential prognostic factors. Survival curves were estimated based on Kaplan-Meier analyses, and Log Rank test was used to contrast factors influencing the survival outcome. Results On the basis of the semiquantitative scoring method for PDL1 expression, the patients were divided into low PDL1 expression group (109 cases) and high PDL1 expression group (107 cases). PDL1 expression was correlated with positive lymph nodes, positive axillary lymph nodes, postoperative radiotherapy, and CK5/6 expression (P < 0.05). The PDL1 expression in tumor tissues was discovered to be a potential prognostic risk factor with the disease-free survival (DFS) and overall survival (OS) for stage III BC. Moreover, patients with high PDL1 expression showed longer lifetime (DFS and OS) compared to those with low PDL1 expression in total patient population (P < 0.05). Moreover, the nomogram showed that the prediction line is in good agreement with the reference line for postoperative 1-, 3-, and 5-year lifetime. The DCA curve showed that the 3- and 5-year lifetime by nomogram had so much better divination of the clinical application than only by PDL1. Conclusion PDL1 is a latent prognostic factor in stage III BC and is closely related to some clinicopathological features. PDL1 expression in tumor tissues is significantly associated with better lifetime rate in stage III BC.
Collapse
Affiliation(s)
- Xi Zhang
- Department of Anesthesiology, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin, Heilongjiang, People’s Republic of China
| | - Ruzhe Li
- Department of Anesthesiology, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin, Heilongjiang, People’s Republic of China
| | - Guonian Wang
- Department of Anesthesiology, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin, Heilongjiang, People’s Republic of China
| |
Collapse
|
57
|
Saberzadeh-Ardestani B, Graham RP, McMahon S, Ahanonu E, Shi Q, Williams C, Hubbard A, Zhang W, Muranyi A, Yan D, Jin Z, Shanmugam K, Sinicrope FA. Immune Marker Spatial Distribution and Clinical Outcome after PD-1 Blockade in Mismatch Repair-deficient, Advanced Colorectal Carcinomas. Clin Cancer Res 2023; 29:4268-4277. [PMID: 37566222 PMCID: PMC10592158 DOI: 10.1158/1078-0432.ccr-23-1109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 06/29/2023] [Accepted: 08/09/2023] [Indexed: 08/12/2023]
Abstract
PURPOSE Targeting the programmed cell death protein 1 (PD-1)/programmed cell death ligand 1 (PD-L1) interaction has led to durable responses in fewer than half of patients with mismatch repair-deficient (MMR-d) advanced colorectal cancers. Immune contexture, including spatial distribution of immune cells in the tumor microenvironment (TME), may predict immunotherapy outcome. EXPERIMENTAL DESIGN Immune contexture and spatial distribution, including cell-to-cell distance measurements, were analyzed by multiplex immunofluorescence (mIF) in primary colorectal cancers with d-MMR (N = 33) from patients treated with anti-PD-1 antibodies. By digital image analysis, density, ratio, intensity, and spatial distribution of PD-L1, PD-1, CD8, CD3, CD68, LAG3, TGFβR2, MHC-I, CD14, B2M, and pan-cytokeratin were computed. Feature selection was performed by regularized Cox regression with LASSO, and a proportional hazards model was fitted to predict progression-free survival (PFS). RESULTS For predicting survival among patients with MMR-d advanced colorectal cancer receiving PD-1 blockade, cell-to-cell distance measurements, but not cell densities or ratios, achieved statistical significance univariately. By multivariable feature selection, only mean number of PD-1+ cells within 10 μm of a PD-L1+ cell was significantly predictive of PFS. Dichotomization of this variable revealed that those with high versus low values had significantly prolonged PFS [median not reached (>83 months) vs. 8.5 months (95% confidence interval (95% CI), 4.7-NR)] with a median PFS of 28.4 months for all patients [adjusted HR (HRadj) = 0.14; 95% CI, 0.04-0.56; P = 0.005]. Expression of PD-1 was observed on CD8+ T cells; PD-L1 on CD3+ and CD8+ T lymphocytes, macrophages (CD68+), and tumor cells. CONCLUSIONS In d-MMR colorectal cancers, PD-1+ to PD-L1+ receptor to ligand proximity is a potential predictive biomarker for the effectiveness of PD-1 blockade.
Collapse
Affiliation(s)
- Bahar Saberzadeh-Ardestani
- Departments of Oncology and Medicine, Rochester, MN
- Gastrointestinal Research Unit, Mayo Clinic, Rochester, MN
| | - Rondell P. Graham
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN
| | - Sara McMahon
- Ventana Medical Systems, Inc./Roche Tissue Diagnostics, Tucson, AZ
| | - Eze Ahanonu
- Ventana Medical Systems, Inc./Roche Tissue Diagnostics, Tucson, AZ
| | - Qian Shi
- Division of Clinical Trials and Biostatistics, Mayo Clinic, Rochester, MN
| | - Crystal Williams
- Ventana Medical Systems, Inc./Roche Tissue Diagnostics, Tucson, AZ
| | - Antony Hubbard
- Ventana Medical Systems, Inc./Roche Tissue Diagnostics, Tucson, AZ
| | - Wenjun Zhang
- Ventana Medical Systems, Inc./Roche Tissue Diagnostics, Tucson, AZ
| | - Andrea Muranyi
- Ventana Medical Systems, Inc./Roche Tissue Diagnostics, Tucson, AZ
| | - Dongyao Yan
- Ventana Medical Systems, Inc./Roche Tissue Diagnostics, Tucson, AZ
| | - Zhaohui Jin
- Departments of Oncology and Medicine, Rochester, MN
| | | | - Frank A. Sinicrope
- Departments of Oncology and Medicine, Rochester, MN
- Gastrointestinal Research Unit, Mayo Clinic, Rochester, MN
- Mayo Clinic Comprehensive Cancer Center Rochester, MN
| |
Collapse
|
58
|
Lin W, Zhang Y, Yang Y, Lin B, Zhu M, Xu J, Chen Y, Wu W, Chen B, Chen X, Liu J, Wang H, Teng F, Yu X, Wang H, Lu J, Zhou Q, Teng L. Anti-PD-1/Her2 Bispecific Antibody IBI315 Enhances the Treatment Effect of Her2-Positive Gastric Cancer through Gasdermin B-Cleavage Induced Pyroptosis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2303908. [PMID: 37587833 PMCID: PMC10602533 DOI: 10.1002/advs.202303908] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Indexed: 08/18/2023]
Abstract
The majority of patients with human epidermal growth factor receptor 2 (Her2)-positive gastric cancer develop refractory to Her2-targeted therapy, where upregulation of immune checkpoints plays an essential role. Herein, a recombinant fully human IgG1 bispecific antibody IBI315 targeting both PD-1 and Her2 is developed and its antitumor efficacy as well as the underlying mechanism is investigated. IBI315 crosslinks the physical interaction between Her2-positive tumor cells and PD-1-positive T cells, resulting in significantly enhanced antitumor effects compared to each parent antibody or their combination, both in vitro and in vivo mouse tumor models reconstituted with human immune cells using patient-derived xenografts and organoids. Moreover, IBI315 treatment also induces the recruitment and activation of immune cells in tumors. Mechanistically, IBI315 triggers gasdermin B (GSDMB)-mediated pyroptosis in tumor cells, leading to the activation and recruiments of T cells. The activated T cells secret IFNγ, enhancing GSDMB expression and establishing a positive feedback loop of T cell activation and tumor cell killing. Notably, GSDMB is found to be elevated in Her2-positive gastric cancer cells, providing a rationale for IBI315's efficacy. IBI315 is supported here as a promising bispecific antibody-based immunotherapy approach for Her2-positive gastric cancer in preclinical studies, broadening the therapeutic landscape of this patient population.
Collapse
Affiliation(s)
- Wu Lin
- Department of Surgical OncologyThe First Affiliated HospitalZhejiang University School of MedicineHangzhouZhejiang310003China
- Department of Colorectal Surgery and Oncology (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences, Zhejiang Province, China)The Second Affiliated Hospital, Zhejiang University School of MedicineHangzhouZhejiang310009China
- Zhejiang Provincial Clinical Research Center for CANCERHangzhouZhejiang310009China
- Cancer Center of Zhejiang UniversityHangzhouZhejiang310009China
| | - Yingzi Zhang
- Department of Surgical OncologyThe First Affiliated HospitalZhejiang University School of MedicineHangzhouZhejiang310003China
| | - Yan Yang
- Department of Surgical OncologyThe First Affiliated HospitalZhejiang University School of MedicineHangzhouZhejiang310003China
| | - Ben Lin
- College of MedicineJiaxing UniversityJiaxingZhejiang314001China
| | - Mengjia Zhu
- Department of Drug DiscoveryInnovent Biologics (Suzhou) Co.SuzhouJiangsu215000China
| | - Jinling Xu
- Department of Drug DiscoveryInnovent Biologics (Suzhou) Co.SuzhouJiangsu215000China
| | - YiRan Chen
- Department of Surgical OncologyThe First Affiliated HospitalZhejiang University School of MedicineHangzhouZhejiang310003China
| | - Weiwei Wu
- Department of Drug DiscoveryInnovent Biologics (Suzhou) Co.SuzhouJiangsu215000China
| | - Bingliang Chen
- Department of Drug DiscoveryInnovent Biologics (Suzhou) Co.SuzhouJiangsu215000China
| | - Xiangliu Chen
- Department of Surgical OncologyThe First Affiliated HospitalZhejiang University School of MedicineHangzhouZhejiang310003China
| | - Jin Liu
- Department of Surgical OncologyThe First Affiliated HospitalZhejiang University School of MedicineHangzhouZhejiang310003China
| | - Haohao Wang
- Department of Surgical OncologyThe First Affiliated HospitalZhejiang University School of MedicineHangzhouZhejiang310003China
| | - Fei Teng
- Department of Surgical OncologyThe First Affiliated HospitalZhejiang University School of MedicineHangzhouZhejiang310003China
| | - Xiongfei Yu
- Department of Surgical OncologyThe First Affiliated HospitalZhejiang University School of MedicineHangzhouZhejiang310003China
| | - Haiyong Wang
- Department of Surgical OncologyThe First Affiliated HospitalZhejiang University School of MedicineHangzhouZhejiang310003China
| | - Jun Lu
- Department of Surgical OncologyThe First Affiliated HospitalZhejiang University School of MedicineHangzhouZhejiang310003China
| | - Quan Zhou
- Institute of Immunology, Department of Surgical Oncology of The First Affiliated HospitalZhejiang University School of MedicineHangzhouZhejiang310058China
| | - Lisong Teng
- Department of Surgical OncologyThe First Affiliated HospitalZhejiang University School of MedicineHangzhouZhejiang310003China
- Zhejiang Provincial Clinical Research Center for CANCERHangzhouZhejiang310009China
- Cancer Center of Zhejiang UniversityHangzhouZhejiang310009China
| |
Collapse
|
59
|
Ding B, Song Y, Liu S, Peng C, Zhang Y. Mechanisms underlying the changes in acetaldehyde dehydrogenase 1 in cholangiocarcinoma. J Cancer 2023; 14:3203-3213. [PMID: 37928420 PMCID: PMC10622993 DOI: 10.7150/jca.86967] [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: 06/09/2023] [Accepted: 09/08/2023] [Indexed: 11/07/2023] Open
Abstract
Cholangiocarcinoma (CCA) is the most recurrent malignant tumor found in the biliary system. It originates from the bile duct epithelial cells characterized by easy metastasis, high intermittent rate, and poor prognosis. Acetaldehyde dehydrogenase 1 (ALDH1), a marker of cancer stem cells, the levels of which are particularly elevated in various of malignant tumors. Additionally, the increased ALDH1 levels are closely related to the degree and prognosis of malignant tumors. This study reviewed the mechanisms underlying the changes in ALDH1 levels in CCA.
Collapse
Affiliation(s)
- Bai Ding
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Hunan Normal University, Changsha, 410005 Hunan Province, China
| | - Yinghui Song
- Central Laboratory of Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, 410015, China
| | - Sulai Liu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Hunan Normal University, Changsha, 410005 Hunan Province, China
- Central Laboratory of Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, 410015, China
| | - Chuang Peng
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Hunan Normal University, Changsha, 410005 Hunan Province, China
| | - Yujing Zhang
- Central Laboratory of Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, 410015, China
- Key Laboratory of Molecular Epidemiology of Hunan Province, School of Medicine, Hunan Normal University, Changsha, China
| |
Collapse
|
60
|
Spangenberg SH, Palermo A, Gazaniga NR, Martínez-Peña F, Guijas C, Chin EN, Rinschen MM, Sander PN, Webb B, Pereira LE, Jia Y, Meitz L, Siuzdak G, Lairson LL. Hydroxyproline metabolism enhances IFN-γ-induced PD-L1 expression and inhibits autophagic flux. Cell Chem Biol 2023; 30:1115-1134.e10. [PMID: 37467751 PMCID: PMC11426993 DOI: 10.1016/j.chembiol.2023.06.016] [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: 09/21/2022] [Revised: 04/20/2023] [Accepted: 06/16/2023] [Indexed: 07/21/2023]
Abstract
The immune checkpoint protein PD-L1 plays critical roles in both immune system homeostasis and tumor progression. Impaired PD-1/PD-L1 function promotes autoimmunity and PD-L1 expression within tumors promotes immune evasion. If and how changes in metabolism or defined metabolites regulate PD-L1 expression is not fully understood. Here, using a metabolomics activity screening-based approach, we have determined that hydroxyproline (Hyp) significantly and directly enhances adaptive (i.e., IFN-γ-induced) PD-L1 expression in multiple relevant myeloid and cancer cell types. Mechanistic studies reveal that Hyp acts as an inhibitor of autophagic flux, which allows it to regulate this negative feedback mechanism, thereby contributing to its overall effect on PD-L1 expression. Due to its prevalence in fibrotic tumors, these findings suggest that hydroxyproline could contribute to the establishment of an immunosuppressive tumor microenvironment and that Hyp metabolism could be targeted to pharmacologically control PD-L1 expression for the treatment of cancer or autoimmune diseases.
Collapse
Affiliation(s)
| | - Amelia Palermo
- Scripps Center for Metabolomics, the Scripps Research Institute, La Jolla, CA 92037, USA; Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Nathalia R Gazaniga
- Department of Chemistry, the Scripps Research Institute, La Jolla, CA 92037, USA
| | | | - Carlos Guijas
- Scripps Center for Metabolomics, the Scripps Research Institute, La Jolla, CA 92037, USA
| | - Emily N Chin
- Department of Chemistry, the Scripps Research Institute, La Jolla, CA 92037, USA
| | - Markus M Rinschen
- Scripps Center for Metabolomics, the Scripps Research Institute, La Jolla, CA 92037, USA
| | - Philipp N Sander
- Department of Chemistry, the Scripps Research Institute, La Jolla, CA 92037, USA
| | - Bill Webb
- Scripps Center for Metabolomics, the Scripps Research Institute, La Jolla, CA 92037, USA
| | - Laura E Pereira
- Department of Chemistry, the Scripps Research Institute, La Jolla, CA 92037, USA
| | - Ying Jia
- Department of Chemistry, the Scripps Research Institute, La Jolla, CA 92037, USA
| | - Lance Meitz
- Department of Chemistry, the Scripps Research Institute, La Jolla, CA 92037, USA
| | - Gary Siuzdak
- Scripps Center for Metabolomics, the Scripps Research Institute, La Jolla, CA 92037, USA; Department of Integrative Structural and Computational Biology, La Jolla, CA 92037, USA.
| | - Luke L Lairson
- Department of Chemistry, the Scripps Research Institute, La Jolla, CA 92037, USA.
| |
Collapse
|
61
|
Liu WL, Zhang YQ, Luo XJ, Zhu YY, Song L, Ming ZH, Zhang LX, Li MJ, Lv RC, Zhang GJ, Chen M. Novel Dual-Mode NIR-II/MRI Nanoprobe Targeting PD-L1 Accurately Evaluates the Efficacy of Immunotherapy for Triple-Negative Breast Cancer. Int J Nanomedicine 2023; 18:5141-5157. [PMID: 37705867 PMCID: PMC10497065 DOI: 10.2147/ijn.s417944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 08/25/2023] [Indexed: 09/15/2023] Open
Abstract
Background Durable responses to immune-checkpoint blocking therapy (ICT) targeting programmed cell death protein-1/ligand-1 (PD-1/PD-L1) have improved outcomes for patients with triple negative breast cancer (TNBC). Unfortunately, only 19-23% of patients benefit from ICT. Hence, non-invasive strategies evaluating responses to therapy and selecting patients who will benefit from ICT are critical issues for TNBC immunotherapy. Methods We developed a novel nanoparticle-Atezolizumab (NPs-Ate) consisting of indocyanine green (ICG), gadolinium-diethylenetriamine pentaacetic acid (Gd-DTPA), human serum albumin (HSA), and Atezolizumab. The efficiency of Gd-DTPA linking was verified using mass spectrometry, and the size of NPs-Ate was characterized using Nano-flow cytometry. The synthesized NPs-Ate were evaluated for fluorescence stability, penetration depth, and target specificity. TNBC cell lines and tumor-bearing mice models were used to identify the feasibility of this dual-modal second near-infrared/magnetic resonance imaging (NIR-II/MRI) system. Additionally, ICT combination with chemotherapy or radiotherapy in TNBC tumor-bearing mice models were used to assess dynamic changes of PD-L1 and predicted therapeutic responses with NPs-Ate. Results Atezolizumab, a monoclonal antibody, was successfully labeled with ICG and Gd-DTPA to generate NPs-Ate. This demonstrated strong fluorescence signals in our NIR-II imaging system, and relaxivity (γ1) of 9.77 mM-1 s-1. In tumor-bearing mice, the NIR-II imaging signal background ratio (SBR) reached its peak of 11.51 at 36 hours, while the MRI imaging SBR reached its highest as 1.95 after 12 hours of tracer injection. NPs-Ate specifically targets cells and tumors expressing PD-L1, enabling monitoring of PD-L1 status during immunotherapy. Combining therapies led to inhibited tumor growth, prolonged survival, and increased PD-L1 expression, effectively monitored using the non-invasive NPs-Ate imaging system. Conclusion The NIR-II/MRI NPs-Ate effectively reflected PD-L1 status during immunotherapy. Real-time and non-invasive immunotherapy and response/prognosis monitoring under NIR-II/MRI imaging guidance in TNBC is a promising and innovative technology with potential for extensive clinical applications in the future.
Collapse
Affiliation(s)
- Wan-Ling Liu
- Department of Breast-Thyroid-Surgery and Cancer Center, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, People’s Republic of China
- Fujian Key Laboratory of Precision Diagnosis and Treatment in Breast Cancer, Xiang’an Hospital of Xiamen University, Xiamen, People’s Republic of China
- Xiamen Key Laboratory of Endocrine-Related Cancer Precision Medicine, Xiamen, People’s Republic of China
- Xiamen Research Center of Clinical Medicine in Breast & Thyroid Cancers, Xiamen, People’s Republic of China
| | - Yong-Qu Zhang
- Department of Breast-Thyroid-Surgery and Cancer Center, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, People’s Republic of China
- Fujian Key Laboratory of Precision Diagnosis and Treatment in Breast Cancer, Xiang’an Hospital of Xiamen University, Xiamen, People’s Republic of China
- Xiamen Key Laboratory of Endocrine-Related Cancer Precision Medicine, Xiamen, People’s Republic of China
- Xiamen Research Center of Clinical Medicine in Breast & Thyroid Cancers, Xiamen, People’s Republic of China
- Department of Breast Center, Cancer Hospital of Shantou University Medical College, Shantou, People’s Republic of China
| | - Xiang-Jie Luo
- College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, People’s Republic of China
| | - Yuan-Yuan Zhu
- Department of Breast-Thyroid-Surgery and Cancer Center, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, People’s Republic of China
- Fujian Key Laboratory of Precision Diagnosis and Treatment in Breast Cancer, Xiang’an Hospital of Xiamen University, Xiamen, People’s Republic of China
- Xiamen Key Laboratory of Endocrine-Related Cancer Precision Medicine, Xiamen, People’s Republic of China
- Xiamen Research Center of Clinical Medicine in Breast & Thyroid Cancers, Xiamen, People’s Republic of China
| | - Liang Song
- Xiamen Institute of Rare Earth Materials, Haixi Institute, Chinese Academy of Sciences and Technology University, Xiamen, People’s Republic of China
| | - Zi-He Ming
- Department of Breast-Thyroid-Surgery and Cancer Center, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, People’s Republic of China
- Fujian Key Laboratory of Precision Diagnosis and Treatment in Breast Cancer, Xiang’an Hospital of Xiamen University, Xiamen, People’s Republic of China
- Xiamen Key Laboratory of Endocrine-Related Cancer Precision Medicine, Xiamen, People’s Republic of China
- Xiamen Research Center of Clinical Medicine in Breast & Thyroid Cancers, Xiamen, People’s Republic of China
| | - Li-Xin Zhang
- Department of Breast-Thyroid-Surgery and Cancer Center, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, People’s Republic of China
- Fujian Key Laboratory of Precision Diagnosis and Treatment in Breast Cancer, Xiang’an Hospital of Xiamen University, Xiamen, People’s Republic of China
- Xiamen Key Laboratory of Endocrine-Related Cancer Precision Medicine, Xiamen, People’s Republic of China
- Xiamen Research Center of Clinical Medicine in Breast & Thyroid Cancers, Xiamen, People’s Republic of China
| | - Meng-Jun Li
- Department of Breast-Thyroid-Surgery and Cancer Center, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, People’s Republic of China
- Fujian Key Laboratory of Precision Diagnosis and Treatment in Breast Cancer, Xiang’an Hospital of Xiamen University, Xiamen, People’s Republic of China
- Xiamen Key Laboratory of Endocrine-Related Cancer Precision Medicine, Xiamen, People’s Republic of China
- Xiamen Research Center of Clinical Medicine in Breast & Thyroid Cancers, Xiamen, People’s Republic of China
| | - Rui-Chan Lv
- Engineering Research Center of Molecular and Neuro Imaging, Ministry of Education, School of Life Science and Technology, Xidian University, Xi’an, Shanxi, People’s Republic of China
| | - Guo-Jun Zhang
- Department of Breast-Thyroid-Surgery and Cancer Center, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, People’s Republic of China
- Fujian Key Laboratory of Precision Diagnosis and Treatment in Breast Cancer, Xiang’an Hospital of Xiamen University, Xiamen, People’s Republic of China
- Xiamen Key Laboratory of Endocrine-Related Cancer Precision Medicine, Xiamen, People’s Republic of China
- Xiamen Research Center of Clinical Medicine in Breast & Thyroid Cancers, Xiamen, People’s Republic of China
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen, People’s Republic of China
| | - Min Chen
- Fujian Key Laboratory of Precision Diagnosis and Treatment in Breast Cancer, Xiang’an Hospital of Xiamen University, Xiamen, People’s Republic of China
- Xiamen Key Laboratory of Endocrine-Related Cancer Precision Medicine, Xiamen, People’s Republic of China
- Xiamen Research Center of Clinical Medicine in Breast & Thyroid Cancers, Xiamen, People’s Republic of China
| |
Collapse
|
62
|
Jia H, Wei P, Zhou S, Hu Y, Zhang C, Liang L, Li B, Gan Z, Xia Y, Jiang H, Shao M, Guo S, Yang Z, Zhong J, Ren F, Zhang H, Zhang Y, Zhao T. Attenuated Salmonella carrying siRNA-PD-L1 and radiation combinatorial therapy induces tumor regression on HCC through T cell-mediated immuno-enhancement. Cell Death Discov 2023; 9:318. [PMID: 37640735 PMCID: PMC10462685 DOI: 10.1038/s41420-023-01603-x] [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: 05/05/2023] [Revised: 07/20/2023] [Accepted: 08/11/2023] [Indexed: 08/31/2023] Open
Abstract
Hepatocellular carcinoma (HCC), the most prevalent type of aggressive liver cancer, accounts for the majority of liver cancer diagnoses and fatalities. Despite recent advancements in HCC treatment, it remains one of the deadliest cancers. Radiation therapy (RT) is among the locoregional therapy modalities employed to treat unresectable or medically inoperable HCC. However, radioresistance poses a significant challenge. It has been demonstrated that RT induced the upregulation of programmed death ligand 1 (PD-L1) on tumor cells, which may affect response to PD-1-based immunotherapy, providing a rationale for combining PD-1/PD-L1 inhibitors with radiation. Here, we utilized attenuated Salmonella as a carrier to explore whether attenuated Salmonella carrying siRNA-PD-L1 could effectively enhance the antitumor effect of radiotherapy on HCC-bearing mice. Our results showed that a combination of siRNA-PD-L1 and radiotherapy had a synergistic antitumor effect by inhibiting the expression of PD-L1 induced by radiation therapy. Mechanistic insights indicated that the combination treatment significantly suppressed tumor cell proliferation, promoted cell apoptosis, and stimulated immune cell infiltration and activation in tumor tissues. Additionally, the combination treatment increased the ratios of CD4+ T, CD8+ T, and NK cells from the spleen in tumor-bearing mice. This study presents a novel therapeutic strategy for HCC treatment, especially for patients with RT resistance.
Collapse
Affiliation(s)
- Huijie Jia
- Department of Oncology, The Third Affiliated Hospital of Xinxiang Medical University, 453000, Xinxiang, Henan, P. R. China
- Xinxiang Engineering Technology Research Center of Immune Checkpoint Drug for Liver-Intestinal Tumors, Xinxiang Medical University, 453000, Xinxiang, Henan, P. R. China
- Department of Pathology, School of Basic Medical Sciences, Xinxiang Medical University, 453000, Xinxiang, Henan, P. R. China
| | - Pengkun Wei
- Xinxiang Engineering Technology Research Center of Immune Checkpoint Drug for Liver-Intestinal Tumors, Xinxiang Medical University, 453000, Xinxiang, Henan, P. R. China
- Department of Immunology, School of Basic Medical Sciences, Xinxiang Medical University, 453000, Xinxiang, Henan, P. R. China
| | - Shijie Zhou
- Xinxiang Engineering Technology Research Center of Immune Checkpoint Drug for Liver-Intestinal Tumors, Xinxiang Medical University, 453000, Xinxiang, Henan, P. R. China
- Department of Immunology, School of Basic Medical Sciences, Xinxiang Medical University, 453000, Xinxiang, Henan, P. R. China
| | - Yuanyuan Hu
- Xinxiang Engineering Technology Research Center of Immune Checkpoint Drug for Liver-Intestinal Tumors, Xinxiang Medical University, 453000, Xinxiang, Henan, P. R. China
- Department of Immunology, School of Basic Medical Sciences, Xinxiang Medical University, 453000, Xinxiang, Henan, P. R. China
| | - Chunjing Zhang
- Xinxiang Engineering Technology Research Center of Immune Checkpoint Drug for Liver-Intestinal Tumors, Xinxiang Medical University, 453000, Xinxiang, Henan, P. R. China
- Department of Immunology, School of Basic Medical Sciences, Xinxiang Medical University, 453000, Xinxiang, Henan, P. R. China
| | - Lirui Liang
- Xinxiang Engineering Technology Research Center of Immune Checkpoint Drug for Liver-Intestinal Tumors, Xinxiang Medical University, 453000, Xinxiang, Henan, P. R. China
- Department of Immunology, School of Basic Medical Sciences, Xinxiang Medical University, 453000, Xinxiang, Henan, P. R. China
| | - Bingqing Li
- Xinxiang Engineering Technology Research Center of Immune Checkpoint Drug for Liver-Intestinal Tumors, Xinxiang Medical University, 453000, Xinxiang, Henan, P. R. China
- Department of Immunology, School of Basic Medical Sciences, Xinxiang Medical University, 453000, Xinxiang, Henan, P. R. China
| | - Zerui Gan
- Xinxiang Engineering Technology Research Center of Immune Checkpoint Drug for Liver-Intestinal Tumors, Xinxiang Medical University, 453000, Xinxiang, Henan, P. R. China
- Department of Immunology, School of Basic Medical Sciences, Xinxiang Medical University, 453000, Xinxiang, Henan, P. R. China
| | - Yuanling Xia
- Xinxiang Engineering Technology Research Center of Immune Checkpoint Drug for Liver-Intestinal Tumors, Xinxiang Medical University, 453000, Xinxiang, Henan, P. R. China
- Department of Immunology, School of Basic Medical Sciences, Xinxiang Medical University, 453000, Xinxiang, Henan, P. R. China
| | - Hanyu Jiang
- Xinxiang Engineering Technology Research Center of Immune Checkpoint Drug for Liver-Intestinal Tumors, Xinxiang Medical University, 453000, Xinxiang, Henan, P. R. China
- Department of Immunology, School of Basic Medical Sciences, Xinxiang Medical University, 453000, Xinxiang, Henan, P. R. China
| | - Mingguang Shao
- Xinxiang Engineering Technology Research Center of Immune Checkpoint Drug for Liver-Intestinal Tumors, Xinxiang Medical University, 453000, Xinxiang, Henan, P. R. China
- Department of Pathology, School of Basic Medical Sciences, Xinxiang Medical University, 453000, Xinxiang, Henan, P. R. China
| | - Sheng Guo
- Xinxiang Engineering Technology Research Center of Immune Checkpoint Drug for Liver-Intestinal Tumors, Xinxiang Medical University, 453000, Xinxiang, Henan, P. R. China
- Department of Immunology, School of Basic Medical Sciences, Xinxiang Medical University, 453000, Xinxiang, Henan, P. R. China
| | - Zishan Yang
- Xinxiang Engineering Technology Research Center of Immune Checkpoint Drug for Liver-Intestinal Tumors, Xinxiang Medical University, 453000, Xinxiang, Henan, P. R. China
- Department of Immunology, School of Basic Medical Sciences, Xinxiang Medical University, 453000, Xinxiang, Henan, P. R. China
| | - Jiateng Zhong
- Xinxiang Engineering Technology Research Center of Immune Checkpoint Drug for Liver-Intestinal Tumors, Xinxiang Medical University, 453000, Xinxiang, Henan, P. R. China
- Department of Pathology, School of Basic Medical Sciences, Xinxiang Medical University, 453000, Xinxiang, Henan, P. R. China
| | - Feng Ren
- Henan International Joint Laboratory of Immunity and Targeted Therapy for Liver-Intestinal Tumors, Xinxiang Medical University, 453000, Xinxiang, Henan, P. R. China
| | - Huiyong Zhang
- Synthetic Biology Engineering Lab of Henan Province, School of Life Science And Technology, Xinxiang Medical University, 453000, Xinxiang, Henan, P. R. China
| | - Yongxi Zhang
- Department of Oncology, The Third Affiliated Hospital of Xinxiang Medical University, 453000, Xinxiang, Henan, P. R. China.
| | - Tiesuo Zhao
- Department of Oncology, The Third Affiliated Hospital of Xinxiang Medical University, 453000, Xinxiang, Henan, P. R. China.
- Xinxiang Engineering Technology Research Center of Immune Checkpoint Drug for Liver-Intestinal Tumors, Xinxiang Medical University, 453000, Xinxiang, Henan, P. R. China.
- Department of Immunology, School of Basic Medical Sciences, Xinxiang Medical University, 453000, Xinxiang, Henan, P. R. China.
| |
Collapse
|
63
|
Miyashita H, Kurzrock R, Bevins NJ, Thangathurai K, Lee S, Pabla S, Nesline M, Glenn ST, Conroy JM, DePietro P, Rubin E, Sicklick JK, Kato S. T-cell priming transcriptomic markers: implications of immunome heterogeneity for precision immunotherapy. NPJ Genom Med 2023; 8:19. [PMID: 37553332 PMCID: PMC10409760 DOI: 10.1038/s41525-023-00359-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Accepted: 06/14/2023] [Indexed: 08/10/2023] Open
Abstract
Immune checkpoint blockade is effective for only a subset of cancers. Targeting T-cell priming markers (TPMs) may enhance activity, but proper application of these agents in the clinic is challenging due to immune complexity and heterogeneity. We interrogated transcriptomics of 15 TPMs (CD137, CD27, CD28, CD80, CD86, CD40, CD40LG, GITR, ICOS, ICOSLG, OX40, OX40LG, GZMB, IFNG, and TBX21) in a pan-cancer cohort (N = 514 patients, 30 types of cancer). TPM expression was analyzed for correlation with histological type, microsatellite instability high (MSI-H), tumor mutational burden (TMB), and programmed death-ligand 1 (PD-L1) expression. Among 514 patients, the most common histological types were colorectal (27%), pancreatic (11%), and breast cancer (10%). No statistically significant association between histological type and TPM expression was seen. In contrast, expression of GZMB (granzyme B, a serine protease stored in activated T and NK cells that induces cancer cell apoptosis) and IFNG (activates cytotoxic T cells) were significantly higher in tumors with MSI-H, TMB ≥ 10 mutations/mb and PD-L1 ≥ 1%. PD-L1 ≥ 1% was also associated with significantly higher CD137, GITR, and ICOS expression. Patients' tumors were classified into "Hot", "Mixed", or "Cold" clusters based on TPM expression using hierarchical clustering. The cold cluster showed a significantly lower proportion of tumors with PD-L1 ≥ 1%. Overall, 502 patients (98%) had individually distinct patterns of TPM expression. Diverse expression patterns of TPMs independent of histological type but correlating with other immunotherapy biomarkers (PD-L1 ≥ 1%, MSI-H and TMB ≥ 10 mutations/mb) were observed. Individualized selection of patients based on TPM immunomic profiles may potentially help with immunotherapy optimization.
Collapse
Affiliation(s)
- Hirotaka Miyashita
- Department of Hematology and Oncology, Dartmouth Cancer Center, Lebanon, NH, USA.
| | - Razelle Kurzrock
- Worldwide Innovative Network (WIN) for Personalized Cancer Therapy, Paris, France
- Division of Hematology and Oncology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Nicholas J Bevins
- Department of Pathology, University of California San Diego, La Jolla, CA, USA
| | - Kartheeswaran Thangathurai
- The Shraga Segal Department for Microbiology, Immunology and Genetics, Ben-Gurion University of the Negev, Beer Sheva, Israel
- Department of Physical Science, University of Vavuniya, Vavuniya, Sri Lanka
| | - Suzanna Lee
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, Department of Medicine, UC, San Diego Moores Cancer Center, La Jolla, CA, USA
| | | | | | - Sean T Glenn
- Roswell Park Comprehensive Cancer Center, Center for Personalized Medicine, Buffalo, NY, USA
| | | | | | - Eitan Rubin
- The Shraga Segal Department for Microbiology, Immunology and Genetics, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Jason K Sicklick
- Division of Surgical Oncology, Department of Surgery, and Center for Personalized Cancer Therapy, University of California, San Diego, La Jolla, CA, USA
| | - Shumei Kato
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, Department of Medicine, UC, San Diego Moores Cancer Center, La Jolla, CA, USA.
| |
Collapse
|
64
|
Martin SD, Bhuiyan I, Soleimani M, Wang G. Biomarkers for Immune Checkpoint Inhibitors in Renal Cell Carcinoma. J Clin Med 2023; 12:4987. [PMID: 37568390 PMCID: PMC10419620 DOI: 10.3390/jcm12154987] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 07/25/2023] [Accepted: 07/26/2023] [Indexed: 08/13/2023] Open
Abstract
Immune checkpoint inhibitor (ICI) therapy has revolutionized renal cell carcinoma treatment. Patients previously thought to be palliative now occasionally achieve complete cures from ICI. However, since immunotherapies stimulate the immune system to induce anti-tumor immunity, they often lead to adverse autoimmunity. Furthermore, some patients receive no benefit from ICI, thereby unnecessarily risking adverse events. In many tumor types, PD-L1 expression levels, immune infiltration, and tumor mutation burden predict the response to ICI and help inform clinical decision making to better target ICI to patients most likely to experience benefits. Unfortunately, renal cell carcinoma is an outlier, as these biomarkers fail to discriminate between positive and negative responses to ICI therapy. Emerging biomarkers such as gene expression profiles and the loss of pro-angiogenic proteins VHL and PBRM-1 show promise for identifying renal cell carcinoma cases likely to respond to ICI. This review provides an overview of the mechanistic underpinnings of different biomarkers and describes the theoretical rationale for their use. We discuss the effectiveness of each biomarker in renal cell carcinoma and other cancer types, and we introduce novel biomarkers that have demonstrated some promise in clinical trials.
Collapse
Affiliation(s)
- Spencer D. Martin
- Department of Pathology and Laboratory Medicine, Faculty of Medicine, University of British Columbia, Vancouver, BC V5Z 1M9, Canada;
| | - Ishmam Bhuiyan
- Faculty of Medicine, University of British Columbia, Vancouver, BC V6T 1Z3, Canada;
| | - Maryam Soleimani
- Division of Medical Oncology, Faculty of Medicine, University of British Columbia, Vancouver, BC V6T 1Z3, Canada;
- British Columbia Cancer Vancouver Centre, Vancouver, BC V5Z 4E6, Canada
| | - Gang Wang
- Department of Pathology and Laboratory Medicine, Faculty of Medicine, University of British Columbia, Vancouver, BC V5Z 1M9, Canada;
- British Columbia Cancer Vancouver Centre, Vancouver, BC V5Z 4E6, Canada
| |
Collapse
|
65
|
Rudjord-Levann AM, Ye Z, Hafkenscheid L, Horn S, Wiegertjes R, Nielsen MA, Song M, Mathiesen CB, Stoop J, Stowell S, Straten PT, Leffler H, Vakhrushev SY, Dabelsteen S, Olsen JV, Wandall HH. Galectin-1 induces a tumor-associated macrophage phenotype and upregulates indoleamine 2,3-dioxygenase-1. iScience 2023; 26:106984. [PMID: 37534161 PMCID: PMC10391608 DOI: 10.1016/j.isci.2023.106984] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 04/18/2023] [Accepted: 05/24/2023] [Indexed: 08/04/2023] Open
Abstract
Galectins are a group of carbohydrate-binding proteins with a presumed immunomodulatory role and an elusive function on antigen-presenting cells. Here we analyzed the expression of galectin-1 and found upregulation of galectin-1 in the extracellular matrix across multiple tumors. Performing an in-depth and dynamic proteomic and phosphoproteomic analysis of human macrophages stimulated with galectin-1, we show that galectin-1 induces a tumor-associated macrophage phenotype with increased expression of key immune checkpoint protein programmed cell death 1 ligand 1 (PD-L1/CD274) and immunomodulator indoleamine 2,3-dioxygenase-1 (IDO1). Galectin-1 induced IDO1 and its active metabolite kynurenine in a dose-dependent manner through JAK/STAT signaling. In a 3D organotypic tissue model system equipped with genetically engineered tumorigenic epithelial cells, we analyzed the cellular source of galectin-1 in the extracellular matrix and found that galectin-1 is derived from epithelial and stromal cells. Our results highlight the potential of targeting galectin-1 in immunotherapeutic treatment of human cancers.
Collapse
Affiliation(s)
- Asha M. Rudjord-Levann
- Copenhagen Center for Glycomics, Departments of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Zilu Ye
- Copenhagen Center for Glycomics, Departments of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Lise Hafkenscheid
- Copenhagen Center for Glycomics, Departments of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Sabrina Horn
- Copenhagen Center for Glycomics, Departments of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Renske Wiegertjes
- Copenhagen Center for Glycomics, Departments of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Mathias A.I. Nielsen
- Copenhagen Center for Glycomics, Departments of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Ming Song
- Copenhagen Center for Glycomics, Departments of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Caroline B.K. Mathiesen
- Copenhagen Center for Glycomics, Departments of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jesse Stoop
- Copenhagen Center for Glycomics, Departments of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Sean Stowell
- Department of Pathology, Brigham and Women’s Hospital, Boston, MA 02115, USA
| | - Per Thor Straten
- Center for Cancer Immune Therapy, Copenhagen University Hospital, Herlev, Denmark
| | - Hakon Leffler
- Division of Microbiology, Immunology and Glycobiology, BMC C1228b, Klinikgatan 28, Lund, Sweden
| | - Sergey Y. Vakhrushev
- Copenhagen Center for Glycomics, Departments of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Sally Dabelsteen
- Department of Oral Medicine and Pathology, School of Dentistry, University of Copenhagen, Copenhagen, Denmark
| | - Jesper V. Olsen
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Hans H. Wandall
- Copenhagen Center for Glycomics, Departments of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
66
|
Lin Q, Wang X, Hu Y. The opportunities and challenges in immunotherapy: Insights from the regulation of PD-L1 in cancer cells. Cancer Lett 2023:216318. [PMID: 37454966 DOI: 10.1016/j.canlet.2023.216318] [Citation(s) in RCA: 26] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 07/07/2023] [Accepted: 07/13/2023] [Indexed: 07/18/2023]
Abstract
The immunosuppressive molecule programmed death-ligand 1 (PD-L1) is frequently upregulated in human cancers. Binding of PD-L1 to its receptor, programmed death-1 (PD-1), on activated T cells facilitates cancer cells to evade the host immune system. Antibody-based PD-1/PD-L1 inhibitors can inhibit PD-1/PD-L1 interaction allowing reactivate cytotoxic T cells to eradicate advanced cancer cells. However, the majority of cancer patients fail to respond to anti-PD-1/PD-L1 therapies and the molecular mechanisms for this remain poorly understood. Recent studies show that PD-L1 expression level on tumor cells affect the clinical efficacy of immune checkpoint therapies. Thus, furthering our understanding of the regulatory mechanisms of PD-L1 expression in cancer cells will be critical to improve clinical response rates and the efficacy of PD-1/PD-L1 immune therapies. Here we review recent studies, primarily focusing on the mechanisms that regulate PD-L1 expression at the transcriptional, post-transcriptional and protein level, with the purpose to drive the development of more targeted and effective anti-PD-1/PD-L1 cancer therapies.
Collapse
Affiliation(s)
- Qingyu Lin
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang Province, 150001, China; Key Laboratory of Science and Engineering for the Multi-modal Prevention and Control of Major Chronic Diseases, Ministry of Industry and Information Technology, China
| | - Xingwen Wang
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang Province, 150001, China; Key Laboratory of Science and Engineering for the Multi-modal Prevention and Control of Major Chronic Diseases, Ministry of Industry and Information Technology, China
| | - Ying Hu
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang Province, 150001, China; Key Laboratory of Science and Engineering for the Multi-modal Prevention and Control of Major Chronic Diseases, Ministry of Industry and Information Technology, China.
| |
Collapse
|
67
|
Hong TH, Bang YH, Joe CY, Hwang S, Lee B, Lee N, Park S, Jung HA, Sun JM, Ahn JS, Ahn MJ, Choi YL, Lee SH. Programmed Death-Ligand 1 Copy Number Alteration as an Adjunct Biomarker of Response to Immunotherapy in Advanced NSCLC. J Thorac Oncol 2023; 18:896-906. [PMID: 37028596 DOI: 10.1016/j.jtho.2023.03.024] [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: 11/17/2022] [Revised: 03/08/2023] [Accepted: 03/29/2023] [Indexed: 04/08/2023]
Abstract
INTRODUCTION This study aimed to evaluate the value of programmed death-ligand 1 (PD-L1) copy number (CN) alteration as an additional biomarker to standard immunohistochemistry (IHC) in predicting response to immune checkpoint inhibitor (ICI) therapy in advanced NSCLC. METHODS Before ICI monotherapy, tumor PD-L1 CN alteration (gain, neutral, or loss) was called using whole-exome sequencing data and compared with IHC results (tumor proportion score ≥50, 1-49, or 0). Progression-free survival (PFS) and overall survival were correlated with both biomarkers. In addition, the impact of CN alteration was further evaluated in two independent cohorts using next-generation sequencing panel. RESULTS A total of 291 patients with advanced-stage NSCLC met the study inclusion criteria. Although the IHC classification distinguished the best responsive group (tumor proportion score ≥ 50), the CN-based classification distinguished the worst responsive group (CN loss) from the others (PFS, p = 0.020; overall survival, p = 0.004). After adjusting for IHC results, CN loss was an independent risk factor for progression (adjusted hazard ratio = 1.32, 95% confidence interval: 1.00-1.73, p = 0.049) and death (adjusted hazard ratio = 1.39, 95% confidence interval: 1.05-1.85, p = 0.022). A risk classification system was developed on the basis of IHC and CN profiles, which outperformed the conventional IHC system. In the validation cohorts, CN loss determined by next-generation sequencing panel was independently associated with worse PFS after ICI treatment, revealing its practical value. CONCLUSIONS This is the first study to directly compare CN alterations with IHC results and survival outcomes after anti-PD-(L)1 therapy. Tumor PD-L1 CN loss can serve as an adjunct biomarker to predict the lack of response. Prospective studies are required to further validate this biomarker.
Collapse
Affiliation(s)
- Tae Hee Hong
- Department of Thoracic and Cardiovascular Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea; Department of Digital Health, Samsung Advanced Institute for Health Science & Technology (SAIHST), Sungkyunkwan University, Seoul, Republic of Korea
| | - Yeong Hak Bang
- Department of Digital Health, Samsung Advanced Institute for Health Science & Technology (SAIHST), Sungkyunkwan University, Seoul, Republic of Korea; Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Cheol Yong Joe
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea; Department of Health Sciences and Technology, Samsung Advanced Institute of Health Sciences and Technology, Sungkyunkwan University, Seoul, Republic of Korea
| | - Soohyun Hwang
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Boram Lee
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Naeun Lee
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea; Department of Health Sciences and Technology, Samsung Advanced Institute of Health Sciences and Technology, Sungkyunkwan University, Seoul, Republic of Korea
| | - Sehhoon Park
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Hyun-Ae Jung
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Jong-Mu Sun
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Jin Seok Ahn
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Myung-Ju Ahn
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Yoon-La Choi
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Se-Hoon Lee
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea; Department of Health Sciences and Technology, Samsung Advanced Institute of Health Sciences and Technology, Sungkyunkwan University, Seoul, Republic of Korea.
| |
Collapse
|
68
|
Zhang Y, Dou Y, Liu Y, Di M, Bian H, Sun X, Yang Q. Advances in Therapeutic Applications of Extracellular Vesicles. Int J Nanomedicine 2023; 18:3285-3307. [PMID: 37346366 PMCID: PMC10281276 DOI: 10.2147/ijn.s409588] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 06/08/2023] [Indexed: 06/23/2023] Open
Abstract
Extracellular vesicles (EVs) are nanoscale bilayer phospholipid membrane vesicles released by cells. Contained large molecules such as nucleic acid, protein, and lipid, EVs are an integral part of cell communication. The contents of EVs vary based on the cell source and play an important role in both pathological and physiological conditions. EVs can be used as drugs or targets in disease treatment, and changes in the contents of EVs can indicate the progression of diseases. In recent years, with the continuous exploration of the structure, characteristics, and functions of EVs, the potential of engineered EVs for drug delivery and therapy being constantly explored. This review provides a brief overview of the structure, characteristics and functions of EVs, summarizes the advanced application of EVs and outlook on the prospect of it. It is our hope that this review will increase understanding of the current development of medical applications of EVs and help us overcome future challenges.
Collapse
Affiliation(s)
- Yiming Zhang
- Department of Spine Surgery, Tianjin Hospital, Tianjin University, Tianjin, People’s Republic of China
- Clinical School of Orthopedics, Tianjin Medical University, Tianjin, People’s Republic of China
| | - Yiming Dou
- Department of Spine Surgery, Tianjin Hospital, Tianjin University, Tianjin, People’s Republic of China
- Clinical School of Orthopedics, Tianjin Medical University, Tianjin, People’s Republic of China
| | - Yang Liu
- Department of Spine Surgery, Tianjin Hospital, Tianjin University, Tianjin, People’s Republic of China
- Clinical School of Orthopedics, Tianjin Medical University, Tianjin, People’s Republic of China
| | - Mingyuan Di
- Department of Spine Surgery, Tianjin Hospital, Tianjin University, Tianjin, People’s Republic of China
- Clinical School of Orthopedics, Tianjin Medical University, Tianjin, People’s Republic of China
| | - Hanming Bian
- Department of Spine Surgery, Tianjin Hospital, Tianjin University, Tianjin, People’s Republic of China
- Clinical School of Orthopedics, Tianjin Medical University, Tianjin, People’s Republic of China
| | - Xun Sun
- Department of Spine Surgery, Tianjin Hospital, Tianjin University, Tianjin, People’s Republic of China
- Clinical School of Orthopedics, Tianjin Medical University, Tianjin, People’s Republic of China
| | - Qiang Yang
- Department of Spine Surgery, Tianjin Hospital, Tianjin University, Tianjin, People’s Republic of China
- Clinical School of Orthopedics, Tianjin Medical University, Tianjin, People’s Republic of China
| |
Collapse
|
69
|
Sammarco E, Manfredi F, Nuzzo A, Ferrari M, Bonato A, Salfi A, Serafin D, Zatteri L, Antonuzzo A, Galli L. Immune Checkpoint Inhibitor Rechallenge in Renal Cell Carcinoma: Current Evidence and Future Directions. Cancers (Basel) 2023; 15:3172. [PMID: 37370782 DOI: 10.3390/cancers15123172] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 06/11/2023] [Accepted: 06/12/2023] [Indexed: 06/29/2023] Open
Abstract
Immune checkpoint inhibitor-based therapies represent the current standard of care in the first-line treatment of advanced renal cell carcinoma. Despite a clear benefit in survival outcomes, a considerable proportion of patients experience disease progression; prospective data about second-line therapy after first-line treatment with immune checkpoint inhibitors are limited to small phase II studies. As with other solid tumors (such as melanoma and non-small cell lung cancer), preliminary data about the clinical efficacy of rechallenge of immunotherapy (alone or in combination with other drugs) in renal cell carcinoma are beginning to emerge. Nevertheless, the role of rechallenge in immunotherapy in this setting of disease remains unclear and cannot be considered a standard of care; currently some randomized trials are exploring this approach in patients with metastatic renal cell carcinoma. The aim of our review is to summarize main evidence available in the literature concerning immunotherapy rechallenge in renal carcinoma, especially focusing on biological rationale of resistance to immune checkpoint inhibitors, on the published data of clinical efficacy and on future perspectives.
Collapse
Affiliation(s)
- Enrico Sammarco
- Unit of Medical Oncology 2, Azienda Ospedaliero-Universitaria Pisana, Santa Chiara Hospital, 56126 Pisa, Italy
| | - Fiorella Manfredi
- Unit of Medical Oncology 2, Azienda Ospedaliero-Universitaria Pisana, Santa Chiara Hospital, 56126 Pisa, Italy
| | - Amedeo Nuzzo
- Unit of Medical Oncology 2, Azienda Ospedaliero-Universitaria Pisana, Santa Chiara Hospital, 56126 Pisa, Italy
| | - Marco Ferrari
- Unit of Medical Oncology 2, Azienda Ospedaliero-Universitaria Pisana, Santa Chiara Hospital, 56126 Pisa, Italy
| | - Adele Bonato
- Unit of Medical Oncology 2, Azienda Ospedaliero-Universitaria Pisana, Santa Chiara Hospital, 56126 Pisa, Italy
| | - Alessia Salfi
- Unit of Medical Oncology 2, Azienda Ospedaliero-Universitaria Pisana, Santa Chiara Hospital, 56126 Pisa, Italy
| | - Debora Serafin
- Unit of Medical Oncology 2, Azienda Ospedaliero-Universitaria Pisana, Santa Chiara Hospital, 56126 Pisa, Italy
| | - Luca Zatteri
- Unit of Medical Oncology 2, Azienda Ospedaliero-Universitaria Pisana, Santa Chiara Hospital, 56126 Pisa, Italy
| | - Andrea Antonuzzo
- Unit of Medical Oncology 1, Azienda Ospedaliero-Universitaria Pisana, Santa Chiara Hospital, 56126 Pisa, Italy
| | - Luca Galli
- Unit of Medical Oncology 2, Azienda Ospedaliero-Universitaria Pisana, Santa Chiara Hospital, 56126 Pisa, Italy
| |
Collapse
|
70
|
Cazzato G, Lettini T, Colagrande A, Trilli I, Ambrogio F, Laface C, Parente P, Maiorano E, Ingravallo G. Immunohistochemical Expression of Programmed Cell Death Ligand 1 (PD-L1) in Human Cutaneous Malignant Melanoma: A Narrative Review with Historical Perspectives. Genes (Basel) 2023; 14:1252. [PMID: 37372432 DOI: 10.3390/genes14061252] [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: 05/23/2023] [Revised: 06/09/2023] [Accepted: 06/10/2023] [Indexed: 06/29/2023] Open
Abstract
Programmed death-ligand 1 (PD-L1) is the primary ligand of the receptor programmed death-1 (PD-1) which is constitutively expressed or activated in myeloid, lymphoid (T, B and NK), normal epithelial cells, and cancer. The PD-1/PD-L1 interaction is crucial for the physiological development of immunological tolerance but also in the development of the cancer. Among these, malignant melanoma represents a tumour in which the immunohistochemical expression of PD-L1 is important to guide future therapeutic choices based on the presence/absence of expression. Various clones have been used over time for immunohistochemical determination, and different results and heterogeneity remain among the various studies in the literature. We perform a narrative review of the present studies in order to discuss and take stock of what certain achievements have been made in this field, what challenges remain, and what possible solutions can be found.
Collapse
Affiliation(s)
- Gerardo Cazzato
- Section of Molecular Pathology, Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe-J), University of Bari, 70124 Bari, Italy
| | - Teresa Lettini
- Section of Molecular Pathology, Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe-J), University of Bari, 70124 Bari, Italy
| | - Anna Colagrande
- Section of Molecular Pathology, Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe-J), University of Bari, 70124 Bari, Italy
| | - Irma Trilli
- Odontomatostologic Clinic, Department of Innovative Technologies in Medicine and Dentistry, University of Chieti, 66100 Chieti, Italy
| | - Francesca Ambrogio
- Section of Dermatology and Venereology, Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe-J), University of Bari, 70124 Bari, Italy
| | - Carmelo Laface
- Medical Oncology, Dario Camberlingo Hospital, 72021 Francavilla Fontana, BR, Italy
| | - Paola Parente
- Pathology Unit, Fondazione IRCCS Ospedale Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, FG, Italy
| | - Eugenio Maiorano
- Section of Molecular Pathology, Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe-J), University of Bari, 70124 Bari, Italy
| | - Giuseppe Ingravallo
- Section of Molecular Pathology, Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe-J), University of Bari, 70124 Bari, Italy
| |
Collapse
|
71
|
Campelo SN, Huang PH, Buie CR, Davalos RV. Recent Advancements in Electroporation Technologies: From Bench to Clinic. Annu Rev Biomed Eng 2023; 25:77-100. [PMID: 36854260 DOI: 10.1146/annurev-bioeng-110220-023800] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/02/2023]
Abstract
Over the past decade, the increased adoption of electroporation-based technologies has led to an expansion of clinical research initiatives. Electroporation has been utilized in molecular biology for mammalian and bacterial transfection; for food sanitation; and in therapeutic settings to increase drug uptake, for gene therapy, and to eliminate cancerous tissues. We begin this article by discussing the biophysics required for understanding the concepts behind the cell permeation phenomenon that is electroporation. We then review nano- and microscale single-cell electroporation technologies before scaling up to emerging in vivo applications.
Collapse
Affiliation(s)
- Sabrina N Campelo
- Department of Biomedical Engineering and Mechanics, Virginia Tech-Wake Forest School of Biomedical Engineering and Sciences, Virginia Tech, Blacksburg, Virginia, USA;
| | - Po-Hsun Huang
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Cullen R Buie
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Rafael V Davalos
- Department of Biomedical Engineering and Mechanics, Virginia Tech-Wake Forest School of Biomedical Engineering and Sciences, Virginia Tech, Blacksburg, Virginia, USA;
| |
Collapse
|
72
|
Nie JZ, Wang MT, Nie D. Regulations of Tumor Microenvironment by Prostaglandins. Cancers (Basel) 2023; 15:3090. [PMID: 37370700 PMCID: PMC10296267 DOI: 10.3390/cancers15123090] [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: 04/04/2023] [Revised: 05/31/2023] [Accepted: 06/06/2023] [Indexed: 06/29/2023] Open
Abstract
Prostaglandins, the bioactive lipids generated from the metabolism of arachidonic acid through cyclooxygenases, have potent effects on many constituents of tumor microenvironments. In this review, we will describe the formation and activities of prostaglandins in the context of the tumor microenvironment. We will discuss the regulation of cancer-associated fibroblasts and immune constituents by prostaglandins and their roles in immune escapes during tumor progression. The review concludes with future perspectives on improving the efficacy of immunotherapy through repurposing non-steroid anti-inflammatory drugs and other prostaglandin modulators.
Collapse
Affiliation(s)
- Jeffrey Z. Nie
- Department of Medical Microbiology, Immunology and Cell Biology, School of Medicine, Simmons Cancer Institute, Southern Illinois University, Springfield, IL 62702, USA
| | - Man-Tzu Wang
- Hillman Cancer Center, University of Pittsburg School of Medicine, Pittsburg, PA 15232, USA
| | - Daotai Nie
- Department of Medical Microbiology, Immunology and Cell Biology, School of Medicine, Simmons Cancer Institute, Southern Illinois University, Springfield, IL 62702, USA
| |
Collapse
|
73
|
Giacomelli M, Monti M, Pezzola DC, Lonardi S, Bugatti M, Missale F, Cioncada R, Melocchi L, Giustini V, Villanacci V, Baronchelli C, Manenti S, Imberti L, Giurisato E, Vermi W. Immuno-Contexture and Immune Checkpoint Molecule Expression in Mismatch Repair Proficient Colorectal Carcinoma. Cancers (Basel) 2023; 15:3097. [PMID: 37370706 DOI: 10.3390/cancers15123097] [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: 05/05/2023] [Revised: 06/01/2023] [Accepted: 06/01/2023] [Indexed: 06/29/2023] Open
Abstract
Colorectal carcinoma (CRC) represents a lethal disease with heterogeneous outcomes. Only patients with mismatch repair (MMR) deficient CRC showing microsatellite instability and hyper-mutated tumors can obtain clinical benefits from current immune checkpoint blockades; on the other hand, immune- or target-based therapeutic strategies are very limited for subjects with mismatch repair proficient CRC (CRCpMMR). Here, we report a comprehensive typing of immune infiltrating cells in CRCpMMR. We also tested the expression and interferon-γ-modulation of PD-L1/CD274. Relevant findings were subsequently validated by immunohistochemistry on fixed materials. CRCpMMR contain a significantly increased fraction of CD163+ macrophages (TAMs) expressing TREM2 and CD66+ neutrophils (TANs) together with decrease in CD4-CD8-CD3+ double negative T lymphocytes (DNTs); no differences were revealed by the analysis of conventional and plasmacytoid dendritic cell populations. A fraction of tumor-infiltrating T-cells displays an exhausted phenotype, co-expressing PD-1 and TIM-3. Remarkably, expression of PD-L1 on fresh tumor cells and TAMs was undetectable even after in vitro stimulation with interferon-γ. These findings confirm the immune suppressive microenvironment of CRCpMMR characterized by dense infiltration of TAMs, occurrence of TANs, lack of DNTs, T-cell exhaustion, and interferon-γ unresponsiveness by host and tumor cells. Appropriate bypass strategies should consider these combinations of immune escape mechanisms in CRCpMMR.
Collapse
Affiliation(s)
- Mauro Giacomelli
- Department of Pathology, ASST Spedali Civili di Brescia, 25123 Brescia, Italy
| | - Matilde Monti
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
| | - Diego Cesare Pezzola
- Department of Surgery, Surgery Division II, ASST Spedali Civili di Brescia, 25123 Brescia, Italy
| | - Silvia Lonardi
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
| | - Mattia Bugatti
- Department of Pathology, ASST Spedali Civili di Brescia, 25123 Brescia, Italy
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
| | - Francesco Missale
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
- Department of Head & Neck Oncology & Surgery Otorhinolaryngology, Antoni Van Leeuwenhoek-Nederlands Kanker Instituut, 1066 CX Amsterdam, The Netherlands
| | - Rossella Cioncada
- Department of Pathology, ASST Spedali Civili di Brescia, 25123 Brescia, Italy
| | - Laura Melocchi
- Department of Pathology, Fondazione Poliambulanza, 25124 Brescia, Italy
| | - Viviana Giustini
- CREA Laboratory, AIL Center for Hemato-Oncologic Research, Diagnostic Department, ASST Spedali Civili di Brescia, 25123 Brescia, Italy
| | - Vincenzo Villanacci
- Department of Pathology, ASST Spedali Civili di Brescia, 25123 Brescia, Italy
| | - Carla Baronchelli
- Department of Pathology, ASST Spedali Civili di Brescia, 25123 Brescia, Italy
| | - Stefania Manenti
- Department of Pathology, ASST Spedali Civili di Brescia, 25123 Brescia, Italy
| | - Luisa Imberti
- Section of Microbiology, University of Brescia, 25123 Brescia, Italy
| | - Emanuele Giurisato
- Department of Biotechnology Chemistry and Pharmacy, University of Siena, 53100 Siena, Italy
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PL, UK
| | - William Vermi
- Department of Pathology, ASST Spedali Civili di Brescia, 25123 Brescia, Italy
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
- Department of Pathology and Immunology, School of Medicine, Washington University, Saint Louis, MO 63130, USA
| |
Collapse
|
74
|
Becker MW, Peters LD, Myint T, Smurlick D, Powell A, Brusko TM, Phelps EA. Immune engineered extracellular vesicles to modulate T cell activation in the context of type 1 diabetes. SCIENCE ADVANCES 2023; 9:eadg1082. [PMID: 37267353 PMCID: PMC10765990 DOI: 10.1126/sciadv.adg1082] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 04/27/2023] [Indexed: 06/04/2023]
Abstract
Extracellular vesicles (EVs) can affect immune responses through antigen presentation and costimulation or coinhibition. We generated designer EVs to modulate T cells in the context of type 1 diabetes, a T cell-mediated autoimmune disease, by engineering a lymphoblast cell line, K562, to express HLA-A*02 (HLA-A2) alongside costimulatory CD80 and/or coinhibitory programmed death ligand 1 (PD-L1). EVs presenting HLA-A2 and CD80 activated CD8+ T cells in a dose, antigen, and HLA-specific manner. Adding PD-L1 to these EVs produced an immunoregulatory response, reducing CD8+ T cell activation and cytotoxicity in vitro. EVs alone could not stimulate T cells without antigen-presenting cells. EVs lacking CD80 were ineffective at modulating CD8+ T cell activation, suggesting that both peptide-HLA complex and costimulation are required for EV-mediated immune modulation. These results provide mechanistic insight into the rational design of EVs as a cell-free approach to immunotherapy that can be tailored to promote inflammatory or tolerogenic immune responses.
Collapse
Affiliation(s)
- Matthew W. Becker
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL
| | - Leeana D. Peters
- Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL, USA
- University of Florida Diabetes Institute, University of Florida, Gainesville, FL, USA
| | - Thinzar Myint
- Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL, USA
- University of Florida Diabetes Institute, University of Florida, Gainesville, FL, USA
| | - Dylan Smurlick
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL
| | - Andrece Powell
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL
| | - Todd M. Brusko
- Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL, USA
- University of Florida Diabetes Institute, University of Florida, Gainesville, FL, USA
- Department of Pediatrics, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Edward A. Phelps
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL
- Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL, USA
- University of Florida Diabetes Institute, University of Florida, Gainesville, FL, USA
| |
Collapse
|
75
|
Blangero F, Robert M, Andraud T, Dumontet C, Vidal H, Eljaafari A. Contribution of Mesenchymal Stem Cells from Obese Adipose Tissue to PD-L1 Over-Expression and Breast Cancer Progression through Pathogenic Th17 Cell Activation. Cancers (Basel) 2023; 15:cancers15112963. [PMID: 37296927 DOI: 10.3390/cancers15112963] [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: 03/30/2023] [Revised: 05/09/2023] [Accepted: 05/24/2023] [Indexed: 06/12/2023] Open
Abstract
BACKGROUND Obesity is a well-known risk factor for cancer. We have previously reported the role of adipose-tissue-derived mesenchymal stem cells from obese individuals (ob-ASC) in the promotion of pathogenic Th17 cells and immune check point (ICP) upregulation. Thus, we postulated herein that this mechanism could contribute to breast cancer (BC) aggressiveness. METHODS Conditioning medium (CM) from mitogen-activated ob-ASC and immune cell co-cultures were added to two human breast cancer cell line (BCCL) cultures. Expressions of pro-inflammatory cytokines, angiogenesis markers, metalloproteinases, and PD-L1 (a major ICP) were measured at the mRNA and/or protein levels. BCCL migration was explored in wound healing assays. Anti-cytokine neutralizing antibodies (Ab) were added to co-cultures. RESULTS CM from ob-ASC/MNC co-cultures increased IL-1β, IL-8, IL-6, VEGF-A, MMP-9, and PD-L1 expressions in both BCCLs and accelerated their migration. The use of Abs demonstrated differential effects for IL-17A and IFNγ on BCCL pro-inflammatory cytokine over-expression or PD-L1 upregulation, respectively, but potentiating effects on BCCL migration. Finally, co-cultures with ob-ASC, but not lean ASC, enhanced PD-L1 expression. CONCLUSIONS Our results demonstrate increased inflammation and ICP markers and accelerated BCCL migration following the activation of pathogenic Th17 cells by ob-ASC, which could represent a new mechanism linking obesity with BC progression.
Collapse
Affiliation(s)
- Ferdinand Blangero
- CarMeN Laboratory, INSERM U1060, INRAE U1397, University Claude Bernard Lyon 1, Hospices Civils de Lyon, Centre Hospitalier Lyon Sud, 69310 Pïerre Bénite, France
| | - Maud Robert
- CarMeN Laboratory, INSERM U1060, INRAE U1397, University Claude Bernard Lyon 1, Hospices Civils de Lyon, Centre Hospitalier Lyon Sud, 69310 Pïerre Bénite, France
- Bariatric Surgery Department, Edouard Herriot Hospital, 69003 Lyon, France
| | - Thomas Andraud
- CarMeN Laboratory, INSERM U1060, INRAE U1397, University Claude Bernard Lyon 1, Hospices Civils de Lyon, Centre Hospitalier Lyon Sud, 69310 Pïerre Bénite, France
| | - Charles Dumontet
- Center of Research in Cancerology of Lyon, INSERM U1052, CNRS 5286, University Claude Bernard Lyon 1, Centre Léon Berard, 69008 Lyon, France
| | - Hubert Vidal
- CarMeN Laboratory, INSERM U1060, INRAE U1397, University Claude Bernard Lyon 1, Hospices Civils de Lyon, Centre Hospitalier Lyon Sud, 69310 Pïerre Bénite, France
| | - Assia Eljaafari
- CarMeN Laboratory, INSERM U1060, INRAE U1397, University Claude Bernard Lyon 1, Hospices Civils de Lyon, Centre Hospitalier Lyon Sud, 69310 Pïerre Bénite, France
- Research Department, Hospices Civils de Lyon, 69002 Lyon, France
| |
Collapse
|
76
|
Budi HS, Farhood B. Targeting oral tumor microenvironment for effective therapy. Cancer Cell Int 2023; 23:101. [PMID: 37221555 DOI: 10.1186/s12935-023-02943-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Accepted: 05/11/2023] [Indexed: 05/25/2023] Open
Abstract
Oral cancers are among the common head and neck malignancies. Different anticancer therapy modalities such as chemotherapy, immunotherapy, radiation therapy, and also targeted molecular therapy may be prescribed for targeting oral malignancies. Traditionally, it has been assumed that targeting malignant cells alone by anticancer modalities such as chemotherapy and radiotherapy suppresses tumor growth. In the last decade, a large number of experiments have confirmed the pivotal role of other cells and secreted molecules in the tumor microenvironment (TME) on tumor progression. Extracellular matrix and immunosuppressive cells such as tumor-associated macrophages, myeloid-derived suppressor cells (MDSCs), cancer-associated fibroblasts (CAFs), and regulatory T cells (Tregs) play key roles in the progression of tumors like oral cancers and resistance to therapy. On the other hand, infiltrated CD4 + and CD8 + T lymphocytes, and natural killer (NK) cells are key anti-tumor cells that suppress the proliferation of malignant cells. Modulation of extracellular matrix and immunosuppressive cells, and also stimulation of anticancer immunity have been suggested to treat oral malignancies more effectively. Furthermore, the administration of some adjuvants or combination therapy modalities may suppress oral malignancies more effectively. In this review, we discuss various interactions between oral cancer cells and TME. Furthermore, we also review the basic mechanisms within oral TME that may cause resistance to therapy. Potential targets and approaches for overcoming the resistance of oral cancers to various anticancer modalities will also be reviewed. The findings for targeting cells and potential therapeutic targets in clinical studies will also be reviewed.
Collapse
Affiliation(s)
- Hendrik Setia Budi
- Department of Oral Biology, Dental Pharmacology, Faculty of Dental Medicine, Universitas Airlangga, Surabaya, Indonesia.
| | - Bagher Farhood
- Department of Medical Physics and Radiology, Faculty of Paramedical Sciences, Kashan University of Medical Sciences, Kashan, Iran.
| |
Collapse
|
77
|
Li Y, Tan R, Li R, Tian R, Liu Z, Wang X, Chen E, Pan T, Qu H. PKM2/STAT1-mediated PD-L1 upregulation on neutrophils during sepsis promotes neutrophil organ accumulation by serving an anti-apoptotic role. J Inflamm (Lond) 2023; 20:16. [PMID: 37131151 PMCID: PMC10155438 DOI: 10.1186/s12950-023-00341-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 04/25/2023] [Indexed: 05/04/2023] Open
Abstract
BACKGROUND Delayed neutrophil apoptosis during sepsis may impact neutrophil organ accumulation and tissue immune homeostasis. Elucidating the mechanisms underlying neutrophil apoptosis may help identify potential therapeutic targets. Glycolysis is critical to neutrophil activities during sepsis. However, the precise mechanisms through which glycolysis regulates neutrophil physiology remain under-explored, especially those involving the non-metabolic functions of glycolytic enzymes. In the present study, the impact of programmed death ligand-1 (PD-L1) on neutrophil apoptosis was explored. The regulatory effect of the glycolytic enzyme, pyruvate kinase M2 (PKM2), whose role in septic neutrophils remains unaddressed, on neutrophil PD-L1 expression was also explored. METHODS Peripheral blood neutrophils were isolated from patients with sepsis and healthy controls. PD-L1 and PKM2 levels were determined by flow cytometry and Western blotting, respectively. Dimethyl sulfoxide (DMSO)-differentiated HL-60 cells were stimulated with lipopolysaccharide (LPS) as an in vitro simulation of septic neutrophils. Cell apoptosis was assessed by annexin V/propidium iodide (annexin V/PI) staining, as well as determination of protein levels of cleaved caspase-3 and myeloid cell leukemia-1 (Mcl-1) by Western blotting. An in vivo model of sepsis was constructed by intraperitoneal injection of LPS (5 mg/kg) for 16 h. Pulmonary and hepatic neutrophil infiltration was assessed by flow cytometry or immunohistochemistry. RESULTS PD-L1 level was elevated on neutrophils under septic conditions. Administration of neutralizing antibodies against PD-L1 partially reversed the inhibitory effect of LPS on neutrophil apoptosis. Neutrophil infiltration into the lung and liver was also reduced in PD-L1-/- mice 16 h after sepsis induction. PKM2 was upregulated in septic neutrophils and promoted neutrophil PD-L1 expression both in vitro and in vivo. In addition, PKM2 nuclear translocation was increased after LPS stimulation, which promoted PD-L1 expression by directly interacting with and activating signal transducer and activator of transcription 1 (STAT1). Inhibition of PKM2 activity or STAT1 activation also led to increased neutrophil apoptosis. CONCLUSION In this study, a PKM2/STAT1-mediated upregulation of PD-L1 on neutrophils and the anti-apoptotic effect of upregulated PD-L1 on neutrophils during sepsis were identified, which may result in increased pulmonary and hepatic neutrophil accumulation. These findings suggest that PKM2 and PD-L1 could serve as potential therapeutic targets.
Collapse
Affiliation(s)
- Yinjiaozhi Li
- Department of Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Er Road, Shanghai, 200025, China
| | - Ruoming Tan
- Department of Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Er Road, Shanghai, 200025, China
| | - Ranran Li
- Department of Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Er Road, Shanghai, 200025, China
| | - Rui Tian
- Department of Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Er Road, Shanghai, 200025, China
| | - Zhaojun Liu
- Department of Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Er Road, Shanghai, 200025, China
| | - Xiaoli Wang
- Department of Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Er Road, Shanghai, 200025, China
| | - Erzhen Chen
- Department of Emergency Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Er Road, Shanghai, 200025, China
| | - Tingting Pan
- Department of Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Er Road, Shanghai, 200025, China.
| | - Hongping Qu
- Department of Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Er Road, Shanghai, 200025, China.
| |
Collapse
|
78
|
Galsky MD, Bajorin DF, Witjes JA, Gschwend JE, Tomita Y, Nasroulah F, Li J, Collette S, Valderrama BP, Grimm MO, Appleman L, Gravis G, Necchi A, Ye D, Stenner F, Wind-Rotolo M, Zhang J, Ünsal-Kaçmaz K. Disease-free Survival Analysis for Patients with High-risk Muscle-invasive Urothelial Carcinoma from the Randomized CheckMate 274 Trial by PD-L1 Combined Positive Score and Tumor Cell Score. Eur Urol 2023; 83:432-440. [PMID: 36868932 PMCID: PMC10520464 DOI: 10.1016/j.eururo.2023.01.016] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 12/09/2022] [Accepted: 01/03/2023] [Indexed: 03/05/2023]
Abstract
BACKGROUND The CheckMate 274 trial demonstrated improved disease-free survival (DFS) with adjuvant nivolumab versus placebo in patients with muscle-invasive urothelial carcinoma at high risk of recurrence after radical surgery in both the intent-to-treat population and the subset with tumor programmed death ligand 1 (PD-L1) expression ≥1%. OBJECTIVE To analyze DFS by combined positive score (CPS), which is based on PD-L1 expression in both tumor and immune cells. DESIGN, SETTING, AND PARTICIPANTS We randomized a total of 709 patients 1:1 to nivolumab 240 mg or placebo every 2 wk intravenously for ≤1 yr of adjuvant treatment. INTERVENTION Nivolumab 240 mg. OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS Primary endpoints were DFS in the intent-to-treat population and patients with tumor PD-L1 expression ≥1% using the tumor cell (TC) score. CPS was determined retrospectively from previously stained slides. Tumor samples with both quantifiable CPS and TC were analyzed. RESULTS AND LIMITATIONS Of 629 patients evaluable for CPS and TC, 557 (89%) had CPS ≥1, 72 (11%) had CPS <1, 249 (40%) had TC ≥1%, and 380 (60%) had TC <1%. Among patients with TC <1%, 81% (n = 309) had CPS ≥1. DFS was improved with nivolumab versus placebo for patients with TC ≥1% (hazard ratio [HR] 0.50, 95% confidence interval [CI] 0.35-0.71), those with CPS ≥1 (HR 0.62, 95% CI 0.49-0.78), and patients with both TC <1% and CPS ≥1 (HR 0.73, 95% CI 0.54-0.99). CONCLUSION More patients had CPS ≥1 than TC ≥1%, and most patients who had TC <1% had CPS ≥1. In addition, patients with CPS ≥1 experienced improved DFS with nivolumab. These results may, in part, explain the mechanisms underlying a benefit with adjuvant nivolumab even in patients who had both TC <1% and CPS ≥1. PATIENT SUMMARY We studied survival time without cancer recurrence (disease-free survival; DFS) for patients treated with nivolumab versus placebo after surgery to remove the bladder or components of the urinary tract for bladder cancer in the CheckMate 274 trial. We assessed the impact of levels of the protein PD-L1 expressed either on tumor cells (tumor cell score; TC) or on both tumor cells and immune cells surrounding the tumor (combined positive score; CPS). DFS was impoved with nivolumab versus placebo for patients with TC ≥1%, CPS ≥1, and for patients with both TC <1% and CPS ≥1. This analysis may help physicians understand which patients would benefit most from treatment with nivolumab.
Collapse
Affiliation(s)
| | - Dean F Bajorin
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | | | - Yoshihiko Tomita
- Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | | | - Jun Li
- Bristol Myers Squibb, Princeton, NJ, USA
| | | | | | | | - Leonard Appleman
- University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh, PA, USA
| | - Gwenaelle Gravis
- Institut Paoli-Calmettes Aix-Marseille Université, Marseille, France
| | - Andrea Necchi
- Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Dingwei Ye
- Fudan University Shanghai Cancer Center, Shanghai, China
| | | | | | | | | |
Collapse
|
79
|
Villarroel-Espindola F, Ejsmentewicz T, Gonzalez-Stegmaier R, Jorquera RA, Salinas E. Intersections between innate immune response and gastric cancer development. World J Gastroenterol 2023; 29:2222-2240. [PMID: 37124883 PMCID: PMC10134417 DOI: 10.3748/wjg.v29.i15.2222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 11/07/2022] [Accepted: 03/13/2023] [Indexed: 04/14/2023] Open
Abstract
Worldwide, gastric cancer (GC) is the fifth most commonly diagnosed malignancy. It has a reduced prevalence but has maintained its poor prognosis being the fourth leading cause of deaths related to cancer. The highest mortality rates occur in Asian and Latin American countries, where cases are usually diagnosed at advanced stages. Overall, GC is viewed as the consequence of a multifactorial process, involving the virulence of the Helicobacter pylori (H. pylori) strains, as well as some environmental factors, dietary habits, and host intrinsic factors. The tumor microenvironment in GC appears to be chronically inflamed which promotes tumor progression and reduces the therapeutic opportunities. It has been suggested that inflammation assessment needs to be measured qualitatively and quantitatively, considering cell-infiltration types, availability of receptors to detect damage and pathogens, and presence or absence of aggressive H. pylori strains. Gastrointestinal epithelial cells express several Toll-like receptors and determine the first defensive line against pathogens, and have been also described as mediators of tumorigenesis. However, other molecules, such as cytokines related to inflammation and innate immunity, including immune checkpoint molecules, interferon-gamma pathway and NETosis have been associated with an increased risk of GC. Therefore, this review will explore innate immune activation in the context of premalignant lesions of the gastric epithelium and established gastric tumors.
Collapse
Affiliation(s)
- Franz Villarroel-Espindola
- Translational Medicine Unit, Instituto Oncologico Fundacion Arturo Lopez Perez, Santiago 7500000, Metropolitan region, Chile
| | - Troy Ejsmentewicz
- Translational Medicine Unit, Instituto Oncologico Fundacion Arturo Lopez Perez, Santiago 7500000, Metropolitan region, Chile
| | - Roxana Gonzalez-Stegmaier
- Translational Medicine Unit, Instituto Oncologico Fundacion Arturo Lopez Perez, Santiago 7500000, Metropolitan region, Chile
| | - Roddy A Jorquera
- Translational Medicine Unit, Instituto Oncologico Fundacion Arturo Lopez Perez, Santiago 7500000, Metropolitan region, Chile
| | - Esteban Salinas
- Translational Medicine Unit, Instituto Oncologico Fundacion Arturo Lopez Perez, Santiago 7500000, Metropolitan region, Chile
| |
Collapse
|
80
|
Mayoh C, Gifford AJ, Terry R, Lau LMS, Wong M, Rao P, Shai-Hee T, Saletta F, Khuong-Quang DA, Qin V, Mateos MK, Meyran D, Miller KE, Yuksel A, Mould EVA, Bowen-James R, Govender D, Senapati A, Zhukova N, Omer N, Dholaria H, Alvaro F, Tapp H, Diamond Y, Pozza LD, Moore AS, Nicholls W, Gottardo NG, McCowage G, Hansford JR, Khaw SL, Wood PJ, Catchpoole D, Cottrell CE, Mardis ER, Marshall GM, Tyrrell V, Haber M, Ziegler DS, Vittorio O, Trapani JA, Cowley MJ, Neeson PJ, Ekert PG. A novel transcriptional signature identifies T-cell infiltration in high-risk paediatric cancer. Genome Med 2023; 15:20. [PMID: 37013636 PMCID: PMC10071693 DOI: 10.1186/s13073-023-01170-x] [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: 10/13/2022] [Accepted: 03/08/2023] [Indexed: 04/05/2023] Open
Abstract
BACKGROUND Molecular profiling of the tumour immune microenvironment (TIME) has enabled the rational choice of immunotherapies in some adult cancers. In contrast, the TIME of paediatric cancers is relatively unexplored. We speculated that a more refined appreciation of the TIME in childhood cancers, rather than a reliance on commonly used biomarkers such as tumour mutation burden (TMB), neoantigen load and PD-L1 expression, is an essential prerequisite for improved immunotherapies in childhood solid cancers. METHODS We combined immunohistochemistry (IHC) with RNA sequencing and whole-genome sequencing across a diverse spectrum of high-risk paediatric cancers to develop an alternative, expression-based signature associated with CD8+ T-cell infiltration of the TIME. Furthermore, we explored transcriptional features of immune archetypes and T-cell receptor sequencing diversity, assessed the relationship between CD8+ and CD4+ abundance by IHC and deconvolution predictions and assessed the common adult biomarkers such as neoantigen load and TMB. RESULTS A novel 15-gene immune signature, Immune Paediatric Signature Score (IPASS), was identified. Using this signature, we estimate up to 31% of high-risk cancers harbour infiltrating T-cells. In addition, we showed that PD-L1 protein expression is poorly correlated with PD-L1 RNA expression and TMB and neoantigen load are not predictive of T-cell infiltration in paediatrics. Furthermore, deconvolution algorithms are only weakly correlated with IHC measurements of T-cells. CONCLUSIONS Our data provides new insights into the variable immune-suppressive mechanisms dampening responses in paediatric solid cancers. Effective immune-based interventions in high-risk paediatric cancer will require individualised analysis of the TIME.
Collapse
Affiliation(s)
- Chelsea Mayoh
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW, Kensington, NSW, Australia
- School of Clinical Medicine, UNSW Medicine & Health, UNSW Sydney, Kensington, NSW, Australia
- University of New South Wales Centre for Childhood Cancer Research, UNSW, Kensington, NSW, Australia
| | - Andrew J Gifford
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW, Kensington, NSW, Australia
- School of Clinical Medicine, UNSW Medicine & Health, UNSW Sydney, Kensington, NSW, Australia
- Anatomical Pathology, NSW Health Pathology, Prince of Wales Hospital, Randwick, NSW, Australia
| | - Rachael Terry
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW, Kensington, NSW, Australia
- School of Clinical Medicine, UNSW Medicine & Health, UNSW Sydney, Kensington, NSW, Australia
| | - Loretta M S Lau
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW, Kensington, NSW, Australia
- School of Clinical Medicine, UNSW Medicine & Health, UNSW Sydney, Kensington, NSW, Australia
- Kids Cancer Centre, Sydney Children's Hospital, Randwick, NSW, Australia
| | - Marie Wong
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW, Kensington, NSW, Australia
- School of Clinical Medicine, UNSW Medicine & Health, UNSW Sydney, Kensington, NSW, Australia
| | - Padmashree Rao
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW, Kensington, NSW, Australia
| | - Tyler Shai-Hee
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW, Kensington, NSW, Australia
| | - Federica Saletta
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW, Kensington, NSW, Australia
| | - Dong-Anh Khuong-Quang
- Children's Cancer Centre, Royal Children's Hospital, Parkville, VIC, Australia
- Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, VIC, Australia
| | - Vicky Qin
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Parkville, VIC, Australia
- The Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC, Australia
| | - Marion K Mateos
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW, Kensington, NSW, Australia
- School of Clinical Medicine, UNSW Medicine & Health, UNSW Sydney, Kensington, NSW, Australia
- Kids Cancer Centre, Sydney Children's Hospital, Randwick, NSW, Australia
| | - Deborah Meyran
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Parkville, VIC, Australia
- The Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC, Australia
| | - Katherine E Miller
- The Steve and Cindy Rasmussen Institute for Genomic Medicine, Nationwide Children's Hospital, Columbus, OH, USA
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Aysen Yuksel
- Tumour Bank, Children's Hospital Westmead, Westmead, NSW, Australia
| | - Emily V A Mould
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW, Kensington, NSW, Australia
| | - Rachel Bowen-James
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW, Kensington, NSW, Australia
- School of Computer Science and Engineering, UNSW Sydney, Kensington, NSW, Australia
- School of Biomedical Engineering, UNSW Sydney, Kensington, NSW, Australia
| | - Dinisha Govender
- Cancer Centre for Children, Children's Hospital Westmead, Westmead, NSW, Australia
| | - Akanksha Senapati
- Kids Cancer Centre, Sydney Children's Hospital, Randwick, NSW, Australia
| | - Nataliya Zhukova
- Monash Children's Hospital, Melbourne, VIC, Australia
- Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Paediatrics, School of Clinical Sciences at Monash Health, Monash University, Clayton, VIC, Australia
| | - Natacha Omer
- Oncology Service, Children's Health Queensland Hospital & Health Service, Brisbane, QLD, Australia
- The University of Queensland Frazer Institute, Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia
| | - Hetal Dholaria
- Department of Paediatric and Adolescent Oncology and Haematology, Perth Children's Hospital, Nedlands, WA, Australia
- Brain Tumour Research Program, Telethon Kids Institute, Nedlands, WA, Australia
| | - Frank Alvaro
- John Hunter Children's Hospital, New Lambton Heights, NSW, Australia
| | - Heather Tapp
- Michael Rice Cancer Centre, Women's and Children's Hospital, South Australia Health and Medical Research Institute, Adelaide, SA, Australia
| | - Yonatan Diamond
- Kids Cancer Centre, Sydney Children's Hospital, Randwick, NSW, Australia
- Children's Cancer Centre, Royal Children's Hospital, Parkville, VIC, Australia
| | - Luciano Dalla Pozza
- Cancer Centre for Children, Children's Hospital Westmead, Westmead, NSW, Australia
| | - Andrew S Moore
- Oncology Service, Children's Health Queensland Hospital & Health Service, Brisbane, QLD, Australia
- Child Health Research Centre, The University of Queensland, Brisbane, QLD, Australia
| | - Wayne Nicholls
- Oncology Service, Children's Health Queensland Hospital & Health Service, Brisbane, QLD, Australia
- School of Medicine, The University of Queensland, Brisbane, QLD, Australia
| | - Nicholas G Gottardo
- Department of Paediatric and Adolescent Oncology and Haematology, Perth Children's Hospital, Nedlands, WA, Australia
- Brain Tumour Research Program, Telethon Kids Institute, Nedlands, WA, Australia
| | - Geoffrey McCowage
- Cancer Centre for Children, Children's Hospital Westmead, Westmead, NSW, Australia
| | - Jordan R Hansford
- Michael Rice Cancer Centre, Women's and Children's Hospital, South Australia Health and Medical Research Institute, Adelaide, SA, Australia
- South Australia ImmunoGENomics Cancer Institute, University of Adelaide, Adelaide, SA, Australia
| | - Seong-Lin Khaw
- Children's Cancer Centre, Royal Children's Hospital, Parkville, VIC, Australia
- Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, VIC, Australia
| | - Paul J Wood
- Monash Children's Hospital, Melbourne, VIC, Australia
- Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Paediatrics, School of Clinical Sciences at Monash Health, Monash University, Clayton, VIC, Australia
| | | | - Catherine E Cottrell
- The Steve and Cindy Rasmussen Institute for Genomic Medicine, Nationwide Children's Hospital, Columbus, OH, USA
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH, USA
- Department of Pathology, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Elaine R Mardis
- The Steve and Cindy Rasmussen Institute for Genomic Medicine, Nationwide Children's Hospital, Columbus, OH, USA
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH, USA
- Department of Neurosurgery, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Glenn M Marshall
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW, Kensington, NSW, Australia
- School of Clinical Medicine, UNSW Medicine & Health, UNSW Sydney, Kensington, NSW, Australia
- Kids Cancer Centre, Sydney Children's Hospital, Randwick, NSW, Australia
| | - Vanessa Tyrrell
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW, Kensington, NSW, Australia
- School of Clinical Medicine, UNSW Medicine & Health, UNSW Sydney, Kensington, NSW, Australia
| | - Michelle Haber
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW, Kensington, NSW, Australia
- School of Clinical Medicine, UNSW Medicine & Health, UNSW Sydney, Kensington, NSW, Australia
| | - David S Ziegler
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW, Kensington, NSW, Australia
- School of Clinical Medicine, UNSW Medicine & Health, UNSW Sydney, Kensington, NSW, Australia
- Kids Cancer Centre, Sydney Children's Hospital, Randwick, NSW, Australia
| | - Orazio Vittorio
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW, Kensington, NSW, Australia
- School of Clinical Medicine, UNSW Medicine & Health, UNSW Sydney, Kensington, NSW, Australia
| | - Joseph A Trapani
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Parkville, VIC, Australia
- The Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC, Australia
| | - Mark J Cowley
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW, Kensington, NSW, Australia
- School of Clinical Medicine, UNSW Medicine & Health, UNSW Sydney, Kensington, NSW, Australia
| | - Paul J Neeson
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Parkville, VIC, Australia
- The Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC, Australia
| | - Paul G Ekert
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW, Kensington, NSW, Australia.
- School of Clinical Medicine, UNSW Medicine & Health, UNSW Sydney, Kensington, NSW, Australia.
- Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, VIC, Australia.
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Parkville, VIC, Australia.
- The Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC, Australia.
| |
Collapse
|
81
|
Elomaa H, Ahtiainen M, Väyrynen SA, Ogino S, Nowak JA, Lau MC, Helminen O, Wirta EV, Seppälä TT, Böhm J, Mecklin JP, Kuopio T, Väyrynen JP. Spatially resolved multimarker evaluation of CD274 (PD-L1)/PDCD1 (PD-1) immune checkpoint expression and macrophage polarisation in colorectal cancer. Br J Cancer 2023; 128:2104-2115. [DOI: 10.1038/s41416-023-02238-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 03/14/2023] [Accepted: 03/15/2023] [Indexed: 04/03/2023] Open
Abstract
Abstract
Background
The CD274 (PD-L1)/PDCD1 (PD-1) immune checkpoint interaction may promote cancer progression, but the expression patterns and prognostic significance of PD-L1 and PD-1 in the colorectal cancer microenvironment are inadequately characterised.
Methods
We used a custom 9-plex immunohistochemistry assay to quantify the expression patterns of PD-L1 and PD-1 in macrophages, T cells, and tumour cells in 910 colorectal cancer patients. We evaluated cancer-specific mortality according to immune cell subset densities using multivariable Cox regression models.
Results
Compared to PD-L1– macrophages, PD-L1+ macrophages were more likely M1-polarised than M2-polarised and located closer to tumour cells. PD-L1+ macrophage density in the invasive margin associated with longer cancer-specific survival [Ptrend = 0.0004, HR for the highest vs. lowest quartile, 0.52; 95% CI: 0.34–0.78]. T cell densities associated with longer cancer-specific survival regardless of PD-1 expression (Ptrend < 0.005 for both PD-1+ and PD-1– subsets). Higher densities of PD-1+ T cell/PD-L1+ macrophage clusters associated with longer cancer-specific survival (Ptrend < 0.005).
Conclusions
PD-L1+ macrophages show distinct polarisation profiles (more M1-like), spatial features (greater co-localisation with tumour cells and PD-1+ T cells), and associations with favourable clinical outcome. Our comprehensive multimarker assessment could enhance the understanding of immune checkpoints in the tumour microenvironment and promote the development of improved immunotherapies.
Collapse
|
82
|
Shi MW, Huang J, Sun Y. Neoadjuvant Immunotherapy for Head and Neck Squamous Cell Carcinoma: Expecting Its Application in Temporal Bone Squamous Cell Carcinoma. Curr Med Sci 2023; 43:213-222. [PMID: 36971976 DOI: 10.1007/s11596-023-2700-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 09/14/2022] [Indexed: 03/29/2023]
Abstract
Temporal bone malignant tumors are characterized by atypical clinical symptoms, and easy recurrence and metastasis. They account for 0.2% of head and neck tumors, and the most common pathological type is squamous cell carcinoma. Patients with squamous cell carcinoma of the temporal bone are often at advanced stages when diagnosed, and lose the chance for surgery. Neoadjuvant immunotherapy has recently been approved as the first-line treatment for refractory recurrent/metastatic squamous cell carcinoma of the head and neck. However, it remains to be determined whether neoadjuvant immunotherapy can be used as the first-line treatment for temporal bone squamous cell carcinoma to reduce the tumor stage before surgery, or as a palliative treatment for patients with unresectable advanced stage carcinoma. The present study reviews the development of immunotherapy and its clinical application in head and neck squamous cell carcinoma, summarizes the treatment of temporal bone squamous cell carcinoma, and prospects the neoadjuvant immunotherapy as the first-line treatment for temporal bone squamous cell carcinoma.
Collapse
Affiliation(s)
- Meng-Wen Shi
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Jing Huang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| | - Yu Sun
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
83
|
Chen K, Wang Q, Liu X, Tian X, Dong A, Yang Y. Immune profiling and prognostic model of pancreatic cancer using quantitative pathology and single-cell RNA sequencing. J Transl Med 2023; 21:210. [PMID: 36944944 PMCID: PMC10031915 DOI: 10.1186/s12967-023-04051-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 03/08/2023] [Indexed: 03/23/2023] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) has a complex tumor immune microenvironment (TIME), the clinical value of which remains elusive. This study aimed to delineate the immune landscape of PDAC and determine the clinical value of immune features in TIME. METHODS Univariable and multivariable Cox regression analyses were performed to evaluate the clinical value of immune features and establish a new prognostic model. We also conducted single-cell RNA sequencing (scRNA-seq) to further characterize the immune profiles of PDAC and explore cell-to-cell interactions. RESULTS There was a significant difference in the immune profiles between PDAC and adjacent noncancerous tissues. Several novel immune features were captured by quantitative pathological analysis on multiplex immunohistochemistry (mIHC), some of which were significantly correlated with the prognosis of patients with PDAC. A risk score-based prognostic model was established based on these immune features. We also constructed a user-friendly nomogram plot to predict the overall survival (OS) of patients by combining the risk score and clinicopathological features. Both mIHC and scRNA-seq analysis revealed PD-L1 expression was low in PDAC. We found that PD1 + cells were distributed in different T cell subpopulations, and were not enriched in a specific subpopulation. In addition, there were other conserved receptor-ligand pairs (CCL5-SDC1/4) besides the PD1-PD-L1 interaction between PD1 + T cells and PD-L1 + tumor cells. CONCLUSION Our findings reveal the immune landscape of PDAC and highlight the significant value of the combined application of mIHC and scRNA-seq for uncovering TIME, which might provide new clues for developing immunotherapy combination strategies.
Collapse
Affiliation(s)
- Kai Chen
- Department of General Surgery, Peking University First Hospital, 8th Xishiku Street, Beijing, 100034, China
| | - Qi Wang
- Department of General Surgery, Peking University First Hospital, 8th Xishiku Street, Beijing, 100034, China
| | - Xinxin Liu
- Department of General Surgery, Peking University First Hospital, 8th Xishiku Street, Beijing, 100034, China
| | - Xiaodong Tian
- Department of General Surgery, Peking University First Hospital, 8th Xishiku Street, Beijing, 100034, China.
| | - Aimei Dong
- Department of Endocrinology, Peking University First Hospital, 8th Xishiku Street, Beijing, 100034, China.
| | - Yinmo Yang
- Department of General Surgery, Peking University First Hospital, 8th Xishiku Street, Beijing, 100034, China.
| |
Collapse
|
84
|
Zhang H, Liu L, Liu J, Dang P, Hu S, Yuan W, Sun Z, Liu Y, Wang C. Roles of tumor-associated macrophages in anti-PD-1/PD-L1 immunotherapy for solid cancers. Mol Cancer 2023; 22:58. [PMID: 36941614 PMCID: PMC10029244 DOI: 10.1186/s12943-023-01725-x] [Citation(s) in RCA: 90] [Impact Index Per Article: 90.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Accepted: 01/16/2023] [Indexed: 03/23/2023] Open
Abstract
In recent years, tumor immunotherapy has made significant progress. However, tumor immunotherapy, particularly immune checkpoint inhibitors (e.g., PD-1/PD-L1 inhibitors), benefits only a tiny proportion of patients in solid cancers. The tumor microenvironment (TME) acts a significant role in tumor immunotherapy. Studies reported that tumor-associated macrophages (TAMs), as one of the main components of TME, seriously affected the therapeutic effect of PD-1/PD-L1 inhibitors. In this review, we analyzed TAMs from epigenetic and single-cell perspectives and introduced the role and mechanisms of TAMs in anti-programmed death protein 1(anti-PD-1) therapy. In addition, we summarized combination regimens that enhance the efficacy of tumor PD-1/PD-L1 inhibitors and elaborated on the role of the TAMs in different solid cancers. Eventually, the clinical value of TAMs by influencing the therapeutic effect of tumor PD-1/PD-L1 inhibitors was discussed. These above are beneficial to elucidate poor therapeutic effect of PD-1/PD-L1 inhibitors in solid tumors from the point of view of TAMs and explore the strategies to improve its objective remission rate of solid cancers.
Collapse
Affiliation(s)
- Hao Zhang
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450001, China
| | - Lin Liu
- Henan Institute of Interconnected Intelligent Health Management, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
- Department of Ultrasound, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Jinbo Liu
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450001, China
| | - Pengyuan Dang
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450001, China
| | - Shengyun Hu
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450001, China
| | - Weitang Yuan
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450001, China
| | - Zhenqiang Sun
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450001, China.
- Henan Institute of Interconnected Intelligent Health Management, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China.
| | - Yang Liu
- Department of Radiotherapy, Henan Cancer Hospital, Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, 450001, China.
| | - Chengzeng Wang
- Henan Institute of Interconnected Intelligent Health Management, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China.
- Department of Ultrasound, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China.
| |
Collapse
|
85
|
Missiaen R, Lesner NP, Simon MC. HIF: a master regulator of nutrient availability and metabolic cross-talk in the tumor microenvironment. EMBO J 2023; 42:e112067. [PMID: 36808622 PMCID: PMC10015374 DOI: 10.15252/embj.2022112067] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 12/13/2022] [Accepted: 12/16/2022] [Indexed: 02/22/2023] Open
Abstract
A role for hypoxia-inducible factors (HIFs) in hypoxia-dependent regulation of tumor cell metabolism has been thoroughly investigated and covered in reviews. However, there is limited information available regarding HIF-dependent regulation of nutrient fates in tumor and stromal cells. Tumor and stromal cells may generate nutrients necessary for function (metabolic symbiosis) or deplete nutrients resulting in possible competition between tumor cells and immune cells, a result of altered nutrient fates. HIF and nutrients in the tumor microenvironment (TME) affect stromal and immune cell metabolism in addition to intrinsic tumor cell metabolism. HIF-dependent metabolic regulation will inevitably result in the accumulation or depletion of essential metabolites in the TME. In response, various cell types in the TME will respond to these hypoxia-dependent alterations by activating HIF-dependent transcription to alter nutrient import, export, and utilization. In recent years, the concept of metabolic competition has been proposed for critical substrates, including glucose, lactate, glutamine, arginine, and tryptophan. In this review, we discuss how HIF-mediated mechanisms control nutrient sensing and availability in the TME, the competition for nutrients, and the metabolic cross-talk between tumor and stromal cells.
Collapse
Affiliation(s)
- Rindert Missiaen
- Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA, USA
| | - Nicholas P Lesner
- Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA, USA
| | - M Celeste Simon
- Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
86
|
Zhang T, Yu-Jing L, Ma T. Role of regulation of PD-1 and PD-L1 expression in sepsis. Front Immunol 2023; 14:1029438. [PMID: 36969168 PMCID: PMC10035551 DOI: 10.3389/fimmu.2023.1029438] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Accepted: 02/27/2023] [Indexed: 03/11/2023] Open
Abstract
Long term immunosuppression is problematic during sepsis. The PD-1 and PD-L1 immune checkpoint proteins have potent immunosuppressive functions. Recent studies have revealed several features of PD-1 and PD-L1 and their roles in sepsis. Here, we summarize the overall findings of PD-1 and PD-L1 by first reviewing the biological features of PD-1 and PD-L1 and then discussing the mechanisms that control the expression of PD-1 and PD-L1. We then review the functions of PD-1 and PD-L1 in physiological settings and further discuss PD-1 and PD-L1 in sepsis, including their involvement in several sepsis-related processes and their potential therapeutic relevance in sepsis. In general, PD-1 and PD-L1 have critical roles in sepsis, indicating that their regulation may be a potential therapeutic target for sepsis.
Collapse
Affiliation(s)
- Teng Zhang
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Li Yu-Jing
- Department of Neurology, Tianjin Medical University General Hospital, Tianjin, China
| | - Tao Ma
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China
- *Correspondence: Tao Ma,
| |
Collapse
|
87
|
Robbins Y, Friedman J, Redman J, Sievers C, Lassoued W, Gulley JL, Allen CT. Tumor cell HLA class I expression and pathologic response following neoadjuvant immunotherapy for newly diagnosed head and neck cancer. Oral Oncol 2023; 138:106309. [PMID: 36682187 PMCID: PMC9974754 DOI: 10.1016/j.oraloncology.2023.106309] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 12/29/2022] [Accepted: 01/13/2023] [Indexed: 01/21/2023]
Abstract
OBJECTIVES Biomarkers are needed to identify patients likely to respond to neoadjuvant immunotherapy (NIT) prior to receiving definitive treatment. MATERIALS AND METHODS We hypothesized that expression of tumor cell HLA class I would correlate with pathologic response (PR) following NIT for primary untreated head and neck cancer. Multispectral immunofluorescence of pre- and post-treatment biopsy specimens from a neoadjuvant study of bintrafusp alfa, a dual TGF-β and PD-L1 inhibitor, was performed. RESULTS Discordant expression of tumor cell HLA class I and PD-L1 measured by multispectral immunofluorescence was observed with most positive tumor cells expressing HLA class I or PD-L1 but not both. Spatial analysis revealed colocalization between tumor parenchyma T cells and HLA class I positive tumors cells, but no clear colocalization between T cells and PD-L1 positive tumor cells. Greater pre-treatment tumor cell HLA class I expression, but not PD-L1 expression or tumor T cell infiltration, correlated with the development of a PR. Additionally, increased tumor cell HLA class I expression after NIT compared to before NIT correlated with development of a PR, whereas inconsistent changes in PD-L1 and T cell infiltration were observed after treatment in all patients. CONCLUSIONS These data provide the rationale for the study of tumor cell HLA class I expression in larger prospective studies powered to determine the performance of biomarkers of PR in newly diagnosed HNSCC patients receiving NIT.
Collapse
Affiliation(s)
- Yvette Robbins
- Section on Translational Tumor Immunology, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD, United States
| | - Jay Friedman
- Section on Translational Tumor Immunology, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD, United States
| | - Jason Redman
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Cem Sievers
- Section on Translational Tumor Immunology, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD, United States
| | - Wiem Lassoued
- Tumor Immune Microenvironment Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - James L Gulley
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States; Tumor Immune Microenvironment Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Clint T Allen
- Section on Translational Tumor Immunology, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD, United States; Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States.
| |
Collapse
|
88
|
Cheng K, Feng X, Chai Z, Wang Z, Liu Z, Yan Z, Wang Y, Zhang S. 4-1BB-Based CAR T Cells Effectively Reverse Exhaustion and Enhance the Anti-Tumor Immune Response through Autocrine PD-L1 scFv Antibody. Int J Mol Sci 2023; 24:ijms24044197. [PMID: 36835603 PMCID: PMC9961031 DOI: 10.3390/ijms24044197] [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: 11/30/2022] [Revised: 02/03/2023] [Accepted: 02/08/2023] [Indexed: 02/22/2023] Open
Abstract
Exhaustion of chimeric antigen receptor (CAR) T cells is one of the limitations for CAR T efficacy in solid tumors and for tumor recurrence after initial CAR T treatment. Tumor treatment with a combination of programmed cell death receptor-1 (PD-1)/programmed cell death ligand-1 (PD-L1) blockage and CD28-based CAR T cells has been intensively studied. However, it remains largely unclear whether autocrine single-chain variable fragments (scFv) PD-L1 antibody can improve 4-1BB-based CAR T cell anti-tumor activity and revert CAR T cell exhaustion. Here, we studied T cells engineered with autocrine PD-L1 scFv and 4-1BB-containing CAR. The antitumor activity and exhaustion of CAR T cells were investigated in vitro and in a xenograft cancer model using NCG mice. CAR T cells with autocrine PD-L1 scFv antibody demonstrate enhanced anti-tumor activity in solid tumors and hematologic malignancies by blocking the PD-1/PD-L1 signaling. Importantly, we found that CAR T exhaustion was largely diminished by autocrine PD-L1 scFv antibody in vivo. As such, 4-1BB CAR T with autocrine PD-L1 scFv antibody combined the power of CAR T cells and the immune checkpoint inhibitor, thereby increasing the anti-tumor immune function and CAR T persistence, providing a cell therapy solution for a better clinical outcome.
Collapse
Affiliation(s)
- Kang Cheng
- Laboratory of Biotechnology Drugs, School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Xiangming Feng
- Laboratory of Biotechnology Drugs, School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Zhirong Chai
- Laboratory of Epigenetics and Translational Medicine, School of Life Sciences, Henan University, Kaifeng 475004, China
| | - Zhenzhen Wang
- Laboratory of Biotechnology Drugs, School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Zheng Liu
- Laboratory of Epigenetics and Translational Medicine, School of Life Sciences, Henan University, Kaifeng 475004, China
| | - Zhanchao Yan
- Laboratory of Epigenetics and Translational Medicine, School of Life Sciences, Henan University, Kaifeng 475004, China
| | - Yanming Wang
- Laboratory of Epigenetics and Translational Medicine, School of Life Sciences, Henan University, Kaifeng 475004, China
- Correspondence: (Y.W.); (S.Z.)
| | - Shoutao Zhang
- Laboratory of Biotechnology Drugs, School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China
- Longhu Laboratory of Advanced Immunology, Zhengzhou 450001, China
- Correspondence: (Y.W.); (S.Z.)
| |
Collapse
|
89
|
Yang M, Gong C, Song K, Huang N, Chen H, Gong H, Yang Y, Guo S, Xiao H. APPL1 Is a Prognostic Biomarker and Correlated with Treg Cell Infiltration via Oxygen-Consuming Metabolism in Renal Clear Cell Carcinoma. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2023; 2023:5885203. [PMID: 36846720 PMCID: PMC9957629 DOI: 10.1155/2023/5885203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Revised: 11/07/2022] [Accepted: 11/24/2022] [Indexed: 02/19/2023]
Abstract
Kidney renal clear cell carcinoma (KIRC) is one of the most hazardous tumors in the urinary system. The regulation of oxygen consumption in renal clear cell carcinoma is a consequence of adaptive reprogramming of oxidative metabolism in tumor cells. APPL1 is a signaling adaptor involved in cell survival, oxidative stress, inflammation, and energy metabolism. However, the correlation of APPL1 with regulatory T cell (Treg) infiltration and prognostic value in KIRC remain unclear. In this study, we comprehensively predicted the potential function and prognostic value of APPL1 in KIRC. For KIRC patients, relatively low expression of APPL1 was associated with high degree of metastasis, pathological stage, and shorter overall time or poor prognosis. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses suggested that low expression of APPL1 may be adapted to the malignant progression of tumors via affecting oxygen-consuming metabolism. In addition, the expression level of APPL1 was negatively correlated with Treg cell infiltration and chemotherapy sensitivity, which indicated that APPL1 may regulate the tumor immune infiltration and chemotherapy resistance by decrease oxygen-consuming metabolic process in KIRC. Therefore, APPL1 may become one of the important prognostic factors, and it may serve as a candidate prognostic biomarker in KIRC.
Collapse
Affiliation(s)
- Ming Yang
- The Lab of Aging Research, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Chuhui Gong
- The Lab of Aging Research, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Kangping Song
- Rehabilitation Medicine Center, West China Hospital, Sichuan University, Chengdu, China
| | - Ning Huang
- The Lab of Aging Research, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Honghan Chen
- The Lab of Aging Research, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Hui Gong
- The Lab of Aging Research, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Yu Yang
- The Lab of Aging Research, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Shujing Guo
- Department of Health Management & Institute of Health Management, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Hengyi Xiao
- The Lab of Aging Research, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
90
|
Ye Y, Xie Y, Pei L, Jiang Z, Wu C, Liu S. Platycodin D induces neutrophil apoptosis by downregulating PD-L1 expression to inhibit breast cancer pulmonary metastasis. Int Immunopharmacol 2023; 115:109733. [PMID: 37724959 DOI: 10.1016/j.intimp.2023.109733] [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: 11/25/2022] [Revised: 01/07/2023] [Accepted: 01/11/2023] [Indexed: 01/21/2023]
Abstract
During breast cancer development, programmed cell death 1 ligand 1 (PD-L1) overexpression in neutrophils leads to delayed apoptosis and promotes neutrophil hyperproliferation in the lung to form a premetastatic niche, which is beneficial for pulmonary metastasis. Platycodin D (PlaD), a triterpenoid saponin with known anti-inflammatory and antitumor effects, has been reported to downregulate PD-L1 expression. This study aimed to investigate the inhibitory effect of PlaD on neutrophil PD-L1 in 4 T1 tumor-bearing mice and the potential mechanism of breast cancer pulmonary metastasis. In this study, the orthotopic 4 T1 murine mammary carcinoma model was administered 10 and 20 mg/kg PlaD by gavage. PlaD reduced the excess neutrophils and decreased their high migratory capacity in bone marrow, peripheral blood and lung tissue in the premetastatic period, thereby effectively inhibiting tumor growth and pulmonary metastasis. Moreover, PlaD inhibited the phosphatidylinositol-3-kinase (PI3K)/Akt pathway by decreasing the expression of PD-L1 in neutrophils and promoted neutrophil apoptosis. In vitro, PlaD treatment decreased the viability and inhibited migration of neutrophil-like dHL-60 in a dose-dependent manner. Similarly, PlaD inhibited the increase in PD-L1 induced by IFN-γ stimulation and subsequently induced apoptosis in dHL-60 cells. In conclusion, the administration of PlaD inhibited the PI3K/Akt signaling pathway by reducing the expression of PD-L1 in neutrophils. PlaD promoted neutrophil apoptosis, thereby inhibiting the establishment of a premetastatic niche and ultimately blocking the development of pulmonary metastasis.
Collapse
Affiliation(s)
- Yiyi Ye
- Institute of Chinese Traditional Surgery, LongHua Hospital, Shanghai University of Traditional Chinese Medicine, 725 Wanpingnan Road, Shanghai 200032, China.
| | - Ying Xie
- Institute of Chinese Traditional Surgery, LongHua Hospital, Shanghai University of Traditional Chinese Medicine, 725 Wanpingnan Road, Shanghai 200032, China
| | - Lixia Pei
- Institute of Chinese Traditional Surgery, LongHua Hospital, Shanghai University of Traditional Chinese Medicine, 725 Wanpingnan Road, Shanghai 200032, China
| | - Ziwei Jiang
- Institute of Chinese Traditional Surgery, LongHua Hospital, Shanghai University of Traditional Chinese Medicine, 725 Wanpingnan Road, Shanghai 200032, China
| | - Chunyu Wu
- Department of Breast Surgery, LongHua Hospital, Shanghai University of Traditional Chinese Medicine, 725 Wanpingnan Road, Shanghai 200032, China
| | - Sheng Liu
- Institute of Chinese Traditional Surgery, LongHua Hospital, Shanghai University of Traditional Chinese Medicine, 725 Wanpingnan Road, Shanghai 200032, China; Department of Breast Surgery, LongHua Hospital, Shanghai University of Traditional Chinese Medicine, 725 Wanpingnan Road, Shanghai 200032, China.
| |
Collapse
|
91
|
Torres-Ruiz J, Absalón-Aguilar A, Reyes-Islas JA, Cassiano-Quezada F, Mejía-Domínguez NR, Pérez-Fragoso A, Maravillas-Montero JL, Núñez-Álvarez C, Juárez-Vega G, Culebro-Bermejo A, Gómez-Martín D. Peripheral expansion of myeloid-derived suppressor cells is related to disease activity and damage accrual in inflammatory myopathies. Rheumatology (Oxford) 2023; 62:775-784. [PMID: 35766810 DOI: 10.1093/rheumatology/keac374] [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: 03/29/2022] [Revised: 05/26/2022] [Accepted: 06/18/2022] [Indexed: 02/04/2023] Open
Abstract
OBJECTIVE To assess the proportion of myeloid-derived suppressor cells (MDSCs), their expression of arginase-1 and programmed cell death ligand 1 (PD-L1) and their relationship with the clinical phenotype of patients with idiopathic inflammatory myopathies (IIMs). METHODS We recruited 37 IIM adult patients and 10 healthy donors in Mexico City. We evaluated their clinical features, the proportion of MDSCs and their expression of PD-L1 and arginase-1 by flow cytometry. Polymorphonuclear (PMN)-MDSCs were defined as CD33dim, CD11b+ and CD66b+ while monocytic (M)-MDSCs were CD33+, CD11b+, HLA-DR- and CD14+. Serum cytokines were analysed with a multiplex assay. We compared the quantitative variables with the Kruskal-Wallis and Mann-Whitney U tests and assessed correlations with Spearman's ρ. RESULTS Most patients had dermatomyositis [n = 30 (81.0%)]. IIM patients had a peripheral expansion of PMN-MDSCs and M-MDSCs with an enhanced expression of arginase-1 and PD-L1. Patients with active disease had a decreased percentage {median 1.75% [interquartile range (IQR) 0.31-5.50 vs 10.71 [3.16-15.58], P = 0.011} of M-MDSCs and a higher absolute number of PD-L1+ M-MDSCs [median 23.21 cells/mm3 (IQR 11.16-148.9) vs 5.95 (4.66-102.7), P = 0.046] with increased expression of PD-L1 [median 3136 arbitrary units (IQR 2258-4992) vs 1961 (1885-2335), P = 0.038]. PD-L1 expression in PMN-MDSCs correlated with the visual analogue scale of pulmonary disease activity (r = 0.34, P = 0.040) and damage (r = 0.36, P = 0.031), serum IL-5 (r = 0.55, P = 0.003), IL-6 (r = 0.46, P = 0.003), IL-8 (r = 0.53, P = 0.018), IL-10 (r = 0.48, P = 0.005) and GM-CSF (r = 0.48, P = 0.012). M-MDSCs negatively correlated with the skeletal Myositis Intention to Treat Index (r = -0.34, P = 0.038) and positively with IL-6 (r = 0.40, P = 0.045). CONCLUSION MDSCs expressing arginase-1 and PD-L1 are expanded in IIM and correlate with disease activity, damage accrual and serum cytokines.
Collapse
Affiliation(s)
- Jiram Torres-Ruiz
- Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán
| | - Abdiel Absalón-Aguilar
- Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán
| | - Juan Alberto Reyes-Islas
- Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán
| | - Fabiola Cassiano-Quezada
- Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán
| | - Nancy R Mejía-Domínguez
- Red de Apoyo a la Investigación, Coordinacion de Investigación Científica, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Alfredo Pérez-Fragoso
- Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán
| | - José Luis Maravillas-Montero
- Red de Apoyo a la Investigación, Coordinacion de Investigación Científica, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Carlos Núñez-Álvarez
- Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán
| | - Guillermo Juárez-Vega
- Red de Apoyo a la Investigación, Coordinacion de Investigación Científica, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Alejandro Culebro-Bermejo
- Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán
| | - Diana Gómez-Martín
- Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán
| |
Collapse
|
92
|
Ye S, Li S, Qin L, Zheng W, Liu B, Li X, Ren Z, Zhao H, Hu X, Ye N, Li G. GBP2 promotes clear cell renal cell carcinoma progression through immune infiltration and regulation of PD‑L1 expression via STAT1 signaling. Oncol Rep 2023; 49:49. [PMID: 36660930 PMCID: PMC9887463 DOI: 10.3892/or.2023.8486] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Accepted: 01/05/2023] [Indexed: 01/21/2023] Open
Abstract
Guanylate‑binding protein 2 (GBP2) has been widely studied in cancer, however, its potential role in clear cell renal cell carcinoma (ccRCC) is not fully elucidated. The present study aimed to explore the effect of GBP2 on tumor progression and its possible underlying molecular mechanisms in ccRCC. The Cancer Genome Atlas, Gene Expression Omnibus, Cancer Cell Line Encyclopedia databases, and several bioinformatics analysis tools, such as Gene Expression Profiling Interactive Analysis 2, Kaplan‑Meier plotter, UALCAN, LinkedOmics, Metascape, GeneMANIA and Tumor Immune Estimation Resource, were used to characterize the functional relationship between GBP2 and ccRCC. Focusing on the association between GBP2 and programmed death ligand 1 (PD‑L1) in vitro, the regulatory mechanism was investigated by knockdown and overexpression of GBP2 in Caki‑1 and 786‑O cells using reverse transcription‑quantitative PCR, western blotting and co‑immunoprecipitation techniques. The results indicated that GBP2 was commonly upregulated in ccRCC, correlating with worse prognosis. In addition, GBP2 expression levels were positively associated with different patterns of immune cell infiltration, suggesting that the GBP2 gene regulates PD‑L1 expression via the signal transducer and activator of transcription 1 (STAT1) pathway. The present study suggested that GBP2 regulates tumor immune infiltration and promotes tumor immune escape through PD‑L1 expression, revealing a potential immunotherapeutic target for ccRCC.
Collapse
Affiliation(s)
- Shujiang Ye
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230012, P.R. China,Anhui Public Health Clinical Center, Hefei, Anhui 230012, P.R. China
| | - Siyu Li
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230012, P.R. China,Anhui Public Health Clinical Center, Hefei, Anhui 230012, P.R. China
| | - Lei Qin
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230012, P.R. China,Anhui Public Health Clinical Center, Hefei, Anhui 230012, P.R. China
| | - Wei Zheng
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230012, P.R. China,Anhui Public Health Clinical Center, Hefei, Anhui 230012, P.R. China
| | - Bin Liu
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230012, P.R. China,Anhui Public Health Clinical Center, Hefei, Anhui 230012, P.R. China
| | - Xiaohui Li
- Department of Anatomy, School of Basic Medicine, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Zhenhua Ren
- Department of Anatomy, School of Basic Medicine, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Huaiming Zhao
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230012, P.R. China,Anhui Public Health Clinical Center, Hefei, Anhui 230012, P.R. China
| | - Xudong Hu
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230012, P.R. China,Anhui Public Health Clinical Center, Hefei, Anhui 230012, P.R. China
| | - Nan Ye
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230012, P.R. China,Anhui Public Health Clinical Center, Hefei, Anhui 230012, P.R. China
| | - Guangyuan Li
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230012, P.R. China,Anhui Public Health Clinical Center, Hefei, Anhui 230012, P.R. China,The Lu'an Hospital Affiliated to Anhui Medical University, Lu'an, Anhui 237005, P.R. China,The Lu'an People's Hospital, Lu'an, Anhui 237005, P.R. China,Correspondence to: Dr Guangyuan Li, Department of Urology, The First Affiliated Hospital of Anhui Medical University, 100 Huaihai Avenue, Hefei, Anhui 230012, P.R. China, E-mail:
| |
Collapse
|
93
|
Xin H, Zhou C, Wang G, Liu Y, Zhang J, Liu Y, Li B, Zhang J, Su M, Li Z, Wang G. Heterogeneity of PD-L1 expression and CD8 lymphocyte infiltration in metastatic colorectal cancer and their prognostic significance. Heliyon 2023; 9:e13048. [PMID: 36814622 PMCID: PMC9939551 DOI: 10.1016/j.heliyon.2023.e13048] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 01/13/2023] [Accepted: 01/13/2023] [Indexed: 01/18/2023] Open
Abstract
Purpose In recent years, immune checkpoint inhibitors have become a major therapeutic method for the treatment of metastatic colorectal cancer (mCRC). Growing evidence indicates that tumour-infiltrating lymphocytes (TILs) in the tumour microenvironment are a prerequisite for the effectiveness of PD-1/PD-L1 blockade therapy. In this study, we aimed to compare PD-L1 expression and cluster of differentiation 4 (CD4) and CD8 TIL infiltration in primary tumours and paired metastases. Patients and methods Altogether, 111 patients with mCRC who underwent surgery at our hospital were included. PD-L1, CD4, and CD8 expression were detected by immunohistochemistry in a tissue microarray. PD-L1 expression was assessed using the combined positivity score (CPS), and a score ≥1 was judged as positive. The area proportion of TILs with positive staining ≥10% was classified as "high", while <10% was classified as "low". Results We observed the discordance of PD-L1 expression between primary tumours and paired metastases in 35/111 (31.5%) patients (κ = 0.137, P = 0.142). This heterogeneity was significantly correlated with discordance of CD8 TIL infiltration between primary tumours and paired metastases (P = 0.003). Compared with corresponding colorectal cancer tumours, lung metastases showed more CD8 TIL infiltration (P = 0.022, median: 8.5% vs. 5.0%), whereas liver metastases exhibited less CD8 TIL infiltration (P = 0.028, median: 3.0% vs. 5.0%). Area proportion of CD4+ and CD8+ TIL infiltration in lung metastases were all higher than those in liver metastases (P = 0.005, median: 15.0% vs. 9.0%; P = 0.001, median: 8.5% vs. 3.0%). Compared with p MMR (MSI-L/MS-S) subgroup, area proportion of CD8 TIL infiltration in primary tumours and CD4, CD8 TIL infiltration in paired metastases were all higher in d MMR (MSI-H) group (P = 0.026, median: 15.0% vs 5.0%; P = 0.039, median: 15.0% vs 9.0%; P = 0.015, median: 15.0% vs 5.0%). Preoperative chemo/radiotherapy may increase CD8 TIL infiltration in primary tumours (P = 0.045, median: 10.0% vs. 5.0%). CD8 TIL infiltration in primary tumours was an independent predictive factor for overall survival (HR 0.28, 95% CI 0.09-0.93, P = 0.038). Conclusion Heterogeneity in PD-L1 expression and CD8 TIL infiltration was found between primary tumours and paired metastases in mCRC. CD8 TIL infiltration in primary tumours could independently forecast the overall survival of patients with mCRC.
Collapse
Key Words
- CD8 tumour infiltrating lymphocytes (TILs)
- CD8, cluster of differentiation 8
- CPS, combined positivity score
- Heterogeneity
- MS-S, microsatellite stability
- MSI-H, microsatellite instability-high
- MSI-L, microsatellite instability-low
- Metastatic colorectal cancer (mCRC)
- PD-L1, programmed death-ligand 1
- Prognosis
- Programmed death-ligand 1 (PD-L1)
- TILs, tumour infiltrating lymphocytes
- dMMR, deficient mismatch repair
- mCRC, metastatic colorectal cancer
- pMMR, proficient mismatch repair
Collapse
Affiliation(s)
- Haisong Xin
- Department of General Surgery, Hebei Medical University Fourth Affiliated Hospital, Shijiazhuang, Hebei, People’s Republic of China
| | - Chaoxi Zhou
- Department of General Surgery, Hebei Medical University Fourth Affiliated Hospital, Shijiazhuang, Hebei, People’s Republic of China
| | - Guanglin Wang
- Department of General Surgery, Hebei Medical University Fourth Affiliated Hospital, Shijiazhuang, Hebei, People’s Republic of China
| | - Yan Liu
- Department of Endocrinology, Hebei Medical University Third Affiliated Hospital, Shijiazhuang, Hebei, People’s Republic of China
| | - Juan Zhang
- Department of General Surgery, Hebei Medical University Fourth Affiliated Hospital, Shijiazhuang, Hebei, People’s Republic of China
| | - Youqiang Liu
- Department of General Surgery, Hebei Medical University Fourth Affiliated Hospital, Shijiazhuang, Hebei, People’s Republic of China
| | - Baokun Li
- Department of General Surgery, Hebei Medical University Fourth Affiliated Hospital, Shijiazhuang, Hebei, People’s Republic of China
| | - Jianfeng Zhang
- Department of General Surgery, Hebei Medical University Fourth Affiliated Hospital, Shijiazhuang, Hebei, People’s Republic of China
| | - Mingming Su
- Department of General Surgery, Hebei Medical University Fourth Affiliated Hospital, Shijiazhuang, Hebei, People’s Republic of China
| | - Zhihan Li
- Department of General Surgery, Hebei Medical University Fourth Affiliated Hospital, Shijiazhuang, Hebei, People’s Republic of China
| | - Guiying Wang
- Department of General Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, People’s Republic of China,Department of General Surgery, Hebei Medical University Fourth Affiliated Hospital, Shijiazhuang, Hebei, People’s Republic of China,Corresponding author. Department of General Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang City, Hebei Province, 050051, People’s Republic of China.
| |
Collapse
|
94
|
Mondal P, Patel NS, Bailey K, Aravind S, Cartwright SB, Hollingsworth MA, Lazenby AJ, Carlson MA. Induction of pancreatic neoplasia in the KRAS/TP53 Oncopig. Dis Model Mech 2023; 16:286617. [PMID: 36579622 PMCID: PMC9884120 DOI: 10.1242/dmm.049699] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 12/13/2022] [Indexed: 12/30/2022] Open
Abstract
The 5-year survival of pancreatic cancer (PC) remains low. Murine models may not adequately mimic human PC and can be too small for medical device development. A large-animal PC model could address these issues. We induced and characterized pancreatic tumors in Oncopigs (transgenic swine containing KRASG12D and TP53R167H). The oncopigs underwent injection of adenovirus expressing Cre recombinase (AdCre) into one of the main pancreatic ducts. Resultant tumors were characterized by histology, cytokine expression, exome sequencing and transcriptome analysis. Ten of 14 Oncopigs (71%) had gross tumor within 3 weeks. At necropsy, all of these subjects had gastric outlet obstruction secondary to pancreatic tumor and phlegmon. Oncopigs with injections without Cre recombinase and wild-type pigs with AdCre injection did not show notable effect. Exome and transcriptome analysis of the porcine pancreatic tumors revealed similarity to the molecular signatures and pathways of human PC. Although further optimization and validation of this porcine PC model would be beneficial, it is anticipated that this model will be useful for focused research and development of diagnostic and therapeutic technologies for PC. This article has an associated First Person interview with the joint first authors of the paper.
Collapse
Affiliation(s)
- Pinaki Mondal
- Department of Surgery, University of Nebraska Medical Center, Omaha, NE 68198, USA,Department of Surgery and VA Research Service, Nebraska-Western Iowa Health Care System, Omaha, NE 68105, USA
| | - Neesha S. Patel
- Department of Surgery, University of Nebraska Medical Center, Omaha, NE 68198, USA,Department of Surgery and VA Research Service, Nebraska-Western Iowa Health Care System, Omaha, NE 68105, USA
| | - Katie Bailey
- Department of Surgery, University of Nebraska Medical Center, Omaha, NE 68198, USA,Department of Surgery and VA Research Service, Nebraska-Western Iowa Health Care System, Omaha, NE 68105, USA
| | - Shruthishree Aravind
- Department of Surgery, University of Nebraska Medical Center, Omaha, NE 68198, USA,Department of Surgery and VA Research Service, Nebraska-Western Iowa Health Care System, Omaha, NE 68105, USA
| | - Sara B. Cartwright
- Department of Surgery, University of Nebraska Medical Center, Omaha, NE 68198, USA,Department of Surgery and VA Research Service, Nebraska-Western Iowa Health Care System, Omaha, NE 68105, USA
| | - Michael A. Hollingsworth
- Eppley Institute for Research in Cancer and Allied Diseases, Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Audrey J. Lazenby
- Department of Pathology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Mark A. Carlson
- Department of Surgery, University of Nebraska Medical Center, Omaha, NE 68198, USA,Department of Surgery and VA Research Service, Nebraska-Western Iowa Health Care System, Omaha, NE 68105, USA,Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE 68198, USA,Author for correspondence ()
| |
Collapse
|
95
|
Zhang Y, Wu J, Zhao C, Zhang S, Zhu J. Recent Advancement of PD-L1 Detection Technologies and Clinical Applications in the Era of Precision Cancer Therapy. J Cancer 2023; 14:850-873. [PMID: 37056391 PMCID: PMC10088895 DOI: 10.7150/jca.81899] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 03/14/2023] [Indexed: 04/15/2023] Open
Abstract
Programmed death-1 is a protein found on the surface of immune cells that can interact with its ligand, programmed death-ligand 1 (PD-L1), which is expressed on the plasma membrane, the surface of secreted cellular exosomes, in cell nuclei, or as a circulating soluble protein. This interaction can lead to immune escape in cancer patients. In clinical settings, PD-L1 plays an important role in tumor disease diagnosis, determining therapeutic effectiveness, and predicting patient prognosis. PD-L1 inhibitors are also essential components of tumor immunotherapy. Thus, the detection of PD-L1 levels is crucial, especially in the era of precision cancer therapy. In recent years, innovations have been made in traditional immunoassay methods and the development of new immunoassays for PD-L1 detection. This review aims to summarize recent research progress in tumor PD-L1 detection technology and highlight the clinical applications of PD-L1.
Collapse
Affiliation(s)
- Yuanfeng Zhang
- Binzhou Medical University, Yantai, Shandong, 264003, China
| | - Juanjuan Wu
- Binzhou People's Hospital Affiliated to Shandong First Medical University, Binzhou, Shandong, 256600, China
| | - Chaobin Zhao
- Binzhou Medical University, Yantai, Shandong, 264003, China
| | - Shuyuan Zhang
- Binzhou Medical University, Yantai, Shandong, 264003, China
| | - Jianbo Zhu
- Binzhou People's Hospital Affiliated to Shandong First Medical University, Binzhou, Shandong, 256600, China
- ✉ Corresponding author: Pro. Jianbo Zhu, Binzhou People's Hospital Affiliated to Shandong First Medical University, 515 Yellow River Seven Road, Binzhou, Shandong, 256600, China; ,
| |
Collapse
|
96
|
Zhou S, Yang H. Immunotherapy resistance in non-small-cell lung cancer: From mechanism to clinical strategies. Front Immunol 2023; 14:1129465. [PMID: 37090727 PMCID: PMC10115980 DOI: 10.3389/fimmu.2023.1129465] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 03/28/2023] [Indexed: 04/25/2023] Open
Abstract
The high primary resistance incidence and unavoidable secondary resistance are the major clinical obstacle to lasting long-term benefits in Non-small-cell lung cancer (NSCLC) patients treated with immunotherapy. The mechanisms of immunotherapy resistance in NSCLC are complex, mainly involving tumor cells and tumor microenvironment (TME) infiltrating immune cells, including TAMs, B cells, NK cells, and T cells. The selection of clinical strategies for NSCLC progression after immunotherapy resistance should depend on the progressive mode. The progression pattern of NSCLC patients after immunotherapy resistance can be divided into oligo-progression and systemic/multiple progression, which should be considered for further treatment selection. In the future, it needs to explore how to optimize the combined therapy and explore strategies to reprogram infiltrating immune cells under various genetic backgrounds of tumor cells and timely reshape TME during antitumor treatments.
Collapse
Affiliation(s)
- Suna Zhou
- Key Laboratory of Radiation Oncology of Taizhou, Radiation Oncology Institute of Enze Medical Health Academy, Department of Radiation Oncology, Taizhou Hospital Affiliated to Wenzhou Medical University, Taizhou, Zhejiang, China
- Department of Radiation Oncology, Xi’an No. 3 Hospital, The Affiliated Hospital of Northwest University, Xi’an, Shaanxi, China
| | - Haihua Yang
- Key Laboratory of Radiation Oncology of Taizhou, Radiation Oncology Institute of Enze Medical Health Academy, Department of Radiation Oncology, Taizhou Hospital Affiliated to Wenzhou Medical University, Taizhou, Zhejiang, China
- *Correspondence: Haihua Yang,
| |
Collapse
|
97
|
PD-1/PD-L1 inhibitor plus chemotherapy versus standard of care in the first-line treatment for recurrent or metastatic head and neck squamous cell carcinoma. Eur Arch Otorhinolaryngol 2023; 280:1-9. [PMID: 35907001 DOI: 10.1007/s00405-022-07571-9] [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: 07/01/2022] [Accepted: 07/22/2022] [Indexed: 01/07/2023]
Abstract
OBJECTIVE This study aimed to evaluate the efficacy and safety of programmed cell death-1/programmed cell death-ligand 1 (PD-1/PD-L1) inhibitor plus chemotherapy vs standard of care (SoC) treatment in the first-line treatment for recurrent or metastatic head and neck squamous cell carcinoma (R/M-SCCHN). METHODS Randomized controlled trials (RCTs) that investigated PD-1/PD-L1 inhibitor plus chemotherapy vs SoC as first-line treatment for R/M-SCCHN were searched from electronic databases (PubMed, Embase and Cochrane Library). The primary outcomes were overall survival (OS), progression-free survival (PFS), objective response rate (ORR), and adverse events (AEs). RESULTS In total, three phase 3 RCTs (KEYNOTE-048, CAPTAIN-1st, and JUPITER-02; n = 1120) with three PD-1 inhibitors (pembrolizumab, camrelizumab and toripalimab) were included in the analysis. Compared with SoC, PD-1 inhibitor plus chemotherapy significantly prolonged PFS (hazard ratio [HR] 0.66, 95% CI 0.40-0.93, p < 0.001) and OS (HR 0.73, 95% CI 0.60-0.86, p < 0.001) of patients. There was no statistical differences in ORR (odds ratio [OR] 1.26; 95% CI 0.97-1.64, p = 0.086), grade 3 or higher AEs (OR 0.77, 95% CI 0.50-1.17, p = 0.221), and treatment-related deaths (OR 1.34, 95% CI 0.60-2.98, p = 0.470) between the two groups. CONCLUSION PD-1 inhibitor plus chemotherapy showed more survival benefit than SoC in the first-line treatment for R/M-SCCHN, with a similar safety profile.
Collapse
|
98
|
Zhao L, Zhang W, Luan F, Chen X, Wu H, He Q, Weng Q, Ding L, Yang B. Butein suppresses PD-L1 expression via downregulating STAT1 in non-small cell lung cancer. Biomed Pharmacother 2023; 157:114030. [PMID: 36455456 DOI: 10.1016/j.biopha.2022.114030] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 11/13/2022] [Accepted: 11/17/2022] [Indexed: 11/29/2022] Open
Abstract
PD-L1 (programmed cell death ligand 1) is frequently up-regulated in tumors and is critical in tumor immune escape. In addition to antibodies that block the interaction between PD-L1 and PD-1 (programmed cell death protein 1), small-molecule compounds that suppress PD-L1 expression also exhibit significant anti-tumor effects, emerging as a new strategy targeting PD-L1. By using a cell-based screening model, we found that butein, a natural chalcone compound, significantly reduced the cytoplasm and cell surface expression of PD-L1. This effect was further validated in various non-small cell lung cancer (NSCLC) cell lines and primary cells derived from clinical NSCLC tissues. Butein inhibited PD-L1 transcription, but not the half-life of PD-L1 protein. Butein reduced STAT1 level and butein-induced PD-L1 suppression was eliminated by the absence of STAT1. By co-culture system, butein improved tumor elimination by increasing the killing ability of CD8+ T cells. By in vivo study, we further confirmed that butein downregulated PD-L1 expression and improved infiltration of CD8+ T cells in tumor tissues. Taken together, our study suggested that butein could suppress the transcription of PD-L1 via downregulating STAT1, providing a theoretical basis for the application of butein in anti-tumor therapy.
Collapse
Affiliation(s)
- Lin Zhao
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Wenxin Zhang
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Fengming Luan
- Department of Gastrointestinal Surgery, Second Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou 310009, China
| | - Xi Chen
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Honghai Wu
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Qiaojun He
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China; The Innovation Institute for Artificial Intelligence in Medicine, Zhejiang University, Hangzhou 310018, China; Cancer Center of Zhejiang University, Hangzhou 310058, China
| | - Qinjie Weng
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China; Center of Drug Safety Evaluation and Research, Zhejiang University, Hangzhou 310058, China
| | - Ling Ding
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China.
| | - Bo Yang
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China; The Innovation Institute for Artificial Intelligence in Medicine, Zhejiang University, Hangzhou 310018, China.
| |
Collapse
|
99
|
Pang K, Shi ZD, Wei LY, Dong Y, Ma YY, Wang W, Wang GY, Cao MY, Dong JJ, Chen YA, Zhang P, Hao L, Xu H, Pan D, Chen ZS, Han CH. Research progress of therapeutic effects and drug resistance of immunotherapy based on PD-1/PD-L1 blockade. Drug Resist Updat 2023; 66:100907. [PMID: 36527888 DOI: 10.1016/j.drup.2022.100907] [Citation(s) in RCA: 63] [Impact Index Per Article: 63.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Revised: 11/12/2022] [Accepted: 11/27/2022] [Indexed: 12/05/2022]
Abstract
The binding of programmed death-1 (PD-1) on the surface of T cells and PD-1 ligand 1 (PD-L1) on tumor cells can prevent the immune-killing effect of T cells on tumor cells and promote the immune escape of tumor cells. Therefore, immune checkpoint blockade targeting PD-1/PD-L1 is a reliable tumor therapy with remarkable efficacy. However, the main challenges of this therapy are low response rate and acquired resistance, so that the outcomes of this therapy are usually unsatisfactory. This review begins with the description of biological structure of the PD-1/PD-L1 immune checkpoint and its role in a variety of cells. Subsequently, the therapeutic effects of immune checkpoint blockers (PD-1 / PD-L1 inhibitors) in various tumors were introduced and analyzed, and the reasons affecting the function of PD-1/PD-L1 were systematically analyzed. Then, we focused on analyzing, sorting out and introducing the possible underlying mechanisms of primary and acquired resistance to PD-1/PD-L1 blockade including abnormal expression of PD-1/PD-L1 and some factors, immune-related pathways, tumor immune microenvironment, and T cell dysfunction and others. Finally, promising therapeutic strategies to sensitize the resistant patients with PD-1/PD-L1 blockade treatment were described. This review is aimed at providing guidance for the treatment of various tumors, and highlighting the drug resistance mechanisms to offer directions for future tumor treatment and improvement of patient prognosis.
Collapse
Affiliation(s)
- Kun Pang
- Department of Urology, Xuzhou Clinical School of Xuzhou Medical University, Xuzhou Central Hospital, 199 Jiefang South Road, Xuzhou, Jiangsu, China; School of Life Sciences, Jiangsu Normal University, Jiangsu, China
| | - Zhen-Duo Shi
- Department of Urology, Xuzhou Clinical School of Xuzhou Medical University, Xuzhou Central Hospital, 199 Jiefang South Road, Xuzhou, Jiangsu, China; School of Life Sciences, Jiangsu Normal University, Jiangsu, China; Department of Urology, Heilongjiang Provincial Hospital, Heilongjiang, China
| | - Liu-Ya Wei
- School of Pharmacy, Weifang Medical University, Weifang, Shandong 261053, China
| | - Yang Dong
- Department of Urology, Xuzhou Clinical School of Xuzhou Medical University, Xuzhou Central Hospital, 199 Jiefang South Road, Xuzhou, Jiangsu, China
| | - Yu-Yang Ma
- Graduate School, Bengbu Medical College, Building 1, Administration Building, 2600 Donghai Avenue, Bengbu, Anhui, China
| | - Wei Wang
- Department of Medical College, Southeast University, 87 DingjiaQiao, Nanjing, China
| | - Guang-Yue Wang
- Graduate School, Bengbu Medical College, Building 1, Administration Building, 2600 Donghai Avenue, Bengbu, Anhui, China
| | - Ming-Yang Cao
- Department of Urology, Xuzhou Clinical School of Xuzhou Medical University, Xuzhou Central Hospital, 199 Jiefang South Road, Xuzhou, Jiangsu, China
| | - Jia-Jun Dong
- School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang 212013, Jiangsu Province, China
| | - Yu-Ang Chen
- Department of Urology, Xuzhou Clinical School of Xuzhou Medical University, Xuzhou Central Hospital, 199 Jiefang South Road, Xuzhou, Jiangsu, China
| | - Peng Zhang
- Graduate School, Bengbu Medical College, Building 1, Administration Building, 2600 Donghai Avenue, Bengbu, Anhui, China
| | - Lin Hao
- Department of Urology, Xuzhou Clinical School of Xuzhou Medical University, Xuzhou Central Hospital, 199 Jiefang South Road, Xuzhou, Jiangsu, China
| | - Hao Xu
- Graduate School, Bengbu Medical College, Building 1, Administration Building, 2600 Donghai Avenue, Bengbu, Anhui, China
| | - Deng Pan
- Graduate School, Bengbu Medical College, Building 1, Administration Building, 2600 Donghai Avenue, Bengbu, Anhui, China
| | - Zhe-Sheng Chen
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, USA.
| | - Cong-Hui Han
- Department of Urology, Xuzhou Clinical School of Xuzhou Medical University, Xuzhou Central Hospital, 199 Jiefang South Road, Xuzhou, Jiangsu, China; School of Life Sciences, Jiangsu Normal University, Jiangsu, China; Department of Urology, Heilongjiang Provincial Hospital, Heilongjiang, China.
| |
Collapse
|
100
|
Xiu W, Pang J, Hu Y, Shi H. Immune-related mechanisms and immunotherapy in extragonadal germ cell tumors. Front Immunol 2023; 14:1145788. [PMID: 37138865 PMCID: PMC10149945 DOI: 10.3389/fimmu.2023.1145788] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 04/03/2023] [Indexed: 05/05/2023] Open
Abstract
Purpose of review Extragonadal germ cell tumors (EGCTs) are relatively rare tumors, accounting for 1%-5% of all GCTs. In this review, we summarize the current research progress regarding the pathogenesis, diagnosis, and treatment of EGCTs from an immunology perspective. Recent findings The histological origin of EGCTs is related to a gonadal origin, but they are located outside the gonad. They show great variation in morphology and can occur in the cranium, mediastinum, sacrococcygeal bone, and other areas. The pathogenesis of EGCTs is poorly understood, and their differential diagnosis is extensive and challenging. EGCT behavior varies greatly according to patient age, histological subtype, and clinical stage. Summary This review provides ideas for the future application of immunology in the fight against such diseases, which is a hot topic currently.
Collapse
Affiliation(s)
- Weigang Xiu
- 1Division of Thoracic Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Jiyun Pang
- 1Division of Thoracic Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
- West China School of Medicine, Sichuan University, Chengdu, China
| | - Yang Hu
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, China
- *Correspondence: Yang Hu, ; Huashan Shi,
| | - Huashan Shi
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
- *Correspondence: Yang Hu, ; Huashan Shi,
| |
Collapse
|