1
|
Li S, Liu Y, Zhang X, Liu Y, Si L, Jiang S, Wang A, Che X, Chen J, Hu J. Multi-pathway oxidative stress amplification via controllably targeted nanomaterials for photoimmunotherapy of tumors. J Nanobiotechnology 2025; 23:33. [PMID: 39844145 PMCID: PMC11753039 DOI: 10.1186/s12951-025-03116-4] [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: 09/27/2024] [Accepted: 01/13/2025] [Indexed: 01/24/2025] Open
Abstract
Photoimmunotherapy, which combines phototherapy with immunotherapy, exhibits significantly improved therapeutic effects compared with mono-treatment regimens. However, its use is associated with drawbacks, such as insufficient reactive oxygen species (ROS) production and uneven photosensitizer distribution. To address these issues, we developed a controllable, targeted nanosystem that enhances oxidative stress through multiple pathways, achieving synergistic photothermal, photodynamic, and immunotherapy effects for tumor treatment. These nanoparticles (D/I@HST NPs) accurately target overexpressed transferrin receptors (TfRs) on the surface of tumor cells through surface-modified transferrin (Tf). After endocytosis, D/I@HST NPs generate ROS under 808-nm laser irradiation, breaking the ROS-responsive crosslinking agent and increasing drug release and utilization. Tf also carries Fe3+, which is reduced to Fe2+ by iron reductase in the acidic tumor microenvironment (TME). Consequently, the endoperoxide bridge structure in dihydroartemisinin is cleaved, causing additional ROS generation. Furthermore, the released IR-780 exerts both photodynamic and photothermal effects, enhancing tumor cell death. This multi-pathway oxidative stress amplification and photothermal effect can trigger immunogenic cell death in tumors, promoting the release of relevant antigens and damage-associated molecular patterns, thereby increasing dendritic cell maturation and sensitivity of tumor cells to immunotherapy. Mature dendritic cells transmit signals to T cells, increasing T cells infiltration and activation, facilitating tumor growth inhibition and the suppression of lung metastasis. Furthermore, the myeloid-derived suppressor cells in the tumor decreases significantly after treatment. In summary, this multi-pathway oxidative stress-amplified targeted nanosystem effectively inhibits tumors, reverses the immunosuppressive tumor microenvironment, and provides new insights into tumor immunotherapy combined with phototherapy.
Collapse
Affiliation(s)
- Song Li
- School of Pharmacy, Binzhou Medical University, Yantai, 264003, China
| | - Yunheng Liu
- School of Pharmacy, Binzhou Medical University, Yantai, 264003, China
| | - Xiaokang Zhang
- School of Pharmacy, Binzhou Medical University, Yantai, 264003, China
| | - Yurong Liu
- School of Pharmacy, Binzhou Medical University, Yantai, 264003, China
| | - Longqing Si
- School of Pharmacy, Binzhou Medical University, Yantai, 264003, China
| | - Shaojing Jiang
- Yantai Engineering Research Center for Digital Technology of Stomatology, School of Stomatology, Binzhou Medical University, Yantai, 264003, China
| | - Aoya Wang
- School of Pharmacy, Binzhou Medical University, Yantai, 264003, China
| | - Xukai Che
- School of Pharmacy, Binzhou Medical University, Yantai, 264003, China
| | - Jing Chen
- School of Pharmacy, Binzhou Medical University, Yantai, 264003, China.
| | - Jinghui Hu
- Yantai Engineering Research Center for Digital Technology of Stomatology, School of Stomatology, Binzhou Medical University, Yantai, 264003, China.
| |
Collapse
|
2
|
Chen Y, Shi L, Yin W, Xia H, Lin C. PD‑1/PD‑L1 inhibitor‑based immunotherapy in locally advanced or metastatic triple‑negative breast cancer: A meta‑analysis. Oncol Lett 2025; 29:57. [PMID: 39606565 PMCID: PMC11600704 DOI: 10.3892/ol.2024.14803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 10/29/2024] [Indexed: 11/29/2024] Open
Abstract
Triple-negative breast cancer (TNBC) is a subtype of breast cancer that is negative for oestrogen receptor, progesterone receptor and human epidermal growth factor receptor 2 expression. Locally advanced and metastatic TNBC not only have a worse prognosis and are more invasive than TNBC, but are also the most immunogenic subtypes of breast cancer. There is still a lack of clarity regarding the optimal treatment of locally advanced or metastatic TNBC. The present study aimed to assess the efficacy and safety of programmed cell death protein 1 (PD-1)/programmed death ligand 1 (PD-L1) inhibitor-based immunotherapy [i.e., immune checkpoint inhibitors (ICIs)] alone or in combination with other therapies for the treatment of locally advanced or metastatic TNBC. The PubMed, Cochrane Library, Embase and MEDLINE databases were searched up to July 19, 2023 to identify studies that examined the efficacy and safety of ICIs for treating TNBC. The primary outcomes were progression-free survival (PFS) and overall survival (OS). The secondary outcomes were safety and adverse events. The data were analysed using Review Manager 5.4. A total of 8 studies (3,338 patients) were included in the present meta-analysis. Compared with other therapies, ICIs had a significantly different effect on OS [hazard ratio (HR)=0.83; 95% confidence interval (CI)=0.69-1.00; P<0.05; I2=59%] in patients with locally advanced or metastatic TNBC. In addition, ICIs significantly prolonged PFS compared with other therapies (intent-to-treat: HR=0.81; 95% CI=0.75-0.88; P<0.00001; I2=0%). Immunotherapy based on PD-1/PD-L1 inhibitors showed variable efficacy on OS and PFS in TNBC, while a significant improvement was observed for PD-L1(+). Future studies should focus on PD-L1 subgroup status, which may help optimize personalized treatment regimens for TNBC.
Collapse
Affiliation(s)
- Yonghui Chen
- School of Chemical and Biological Engineering, Yichun College, Yichun, Jiangxi 336000, P.R. China
| | - Liji Shi
- School of Chemical and Biological Engineering, Yichun College, Yichun, Jiangxi 336000, P.R. China
| | - Weihua Yin
- School of Chemical and Biological Engineering, Yichun College, Yichun, Jiangxi 336000, P.R. China
- Department of Oncology, Baoan Central Hospital of Shenzhen, Shenzhen, Guangdong 518102, P.R. China
| | - Hongmei Xia
- Department of Oncology, Baoan Central Hospital of Shenzhen, Shenzhen, Guangdong 518102, P.R. China
| | - Canling Lin
- School of Chemical and Biological Engineering, Yichun College, Yichun, Jiangxi 336000, P.R. China
| |
Collapse
|
3
|
Rogers MC, Lamens KD, Tollefson SJ, Williams JV. Genetic absence of PD-L1 does not restore CD8 + T cell function during respiratory virus infection and delays virus clearance. J Virol 2024; 98:e0079724. [PMID: 39311697 PMCID: PMC11495042 DOI: 10.1128/jvi.00797-24] [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: 05/17/2024] [Accepted: 08/25/2024] [Indexed: 10/23/2024] Open
Abstract
A key mediator of T cell impairment during respiratory virus infection is the inhibitory receptor PD-1. PD-1 is induced on T cells following antigen exposure, whereas proinflammatory cytokines upregulate the ligands PD-L1 and PD-L2. Respiratory virus infection leads to upregulation of PD-L1 on airway epithelial cells, dendritic cells, and alveolar macrophages. However, the role of PD-L1 on different cell types in acute respiratory virus infections is not known. We sought to determine the role of PD-L1 on different cell types in CD8+ T cell impairment. We found that PD-L1-/- mice challenged with human metapneumovirus or influenza showed a similar level of CD8+ T cell impairment compared to wild-type (WT) mice. Moreover, virus clearance was delayed in PD-L1-/- mice compared to WT. CD8+ T cells from PD-L1-deficient mice expressed higher levels of inhibitory receptors both at baseline and after respiratory virus infection. The antibody blockade of PD-L2 failed to restore function to the impaired cells. While reciprocal bone marrow chimeras between WT and PD-L1-/- mice did not restore CD8+ T cell function after the respiratory virus challenge, mice that received the PD-L1-/- bone marrow had higher inhibitory receptor expression on CD8+ cells. This discrepancy in the inhibitory receptor expression suggests that cells of the hematopoietic compartment contribute to T cell impairment on CD8+ T cells.IMPORTANCEThe phenomenon of pulmonary CD8+ T cell impairment with diminished antiviral function occurs during acute respiratory virus infection mediated by Programmed Cell Death-1 (PD-1) signaling. Moreover, PD-1 blockade enhances T cell function to hasten viral clearance. The ligand PD-L1 is expressed in many cell types, but which cells drive lung T cell impairment is not known. We used genetic approaches to determine the contribution of PD-L1 on lung T cell impairment. We found that PD-L2 cannot compensate for the loss of PD-L1, and PD-L1-deficient mice exhibit increased expression of other inhibitory receptors. Bone marrow chimeras between PD-L1-deficient and wild-type mice indicated that hematopoietic PD-L1 expression is associated with inhibitory receptor upregulation and impairment.
Collapse
Affiliation(s)
- Meredith C. Rogers
- Department of Pediatrics, University of Pittsburgh School of Medicine, UPMC Children’s Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Kristina D. Lamens
- Department of Pediatrics, University of Pittsburgh School of Medicine, UPMC Children’s Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Sharon J. Tollefson
- Department of Pediatrics, University of Pittsburgh School of Medicine, UPMC Children’s Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - John V. Williams
- Department of Pediatrics, University of Pittsburgh School of Medicine, UPMC Children’s Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Institute for Infection, Inflammation, and Immunity in Children (i4Kids), Pittsburgh, Pennsylvania, USA
| |
Collapse
|
4
|
DeNiro G, Que K, Fujimoto T, Koo SM, Schneider B, Mukhopadhyay A, Kim J, Sawant A, Nguyen TA. OMIP-105: A 30-color full-spectrum flow cytometry panel to characterize the immune cell landscape in spleen and tumor within a syngeneic MC-38 murine colon carcinoma model. Cytometry A 2024; 105:659-665. [PMID: 39107997 DOI: 10.1002/cyto.a.24886] [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: 12/15/2023] [Revised: 06/12/2024] [Accepted: 06/20/2024] [Indexed: 10/25/2024]
Abstract
This panel was designed to characterize the immune cell landscape in the mouse tumor microenvironment as well as mouse lymphoid tissues (e.g., spleen). As an example, using the MC-38 mouse syngeneic tumor model, we demonstrated that we could measure the frequency and characterize the functional status of CD4 T cells, CD8 T cells, regulatory T cells, NK cells, B cells, macrophages, granulocytes, monocytes, and dendritic cells. This panel is especially useful for understanding the immune landscape in "cold" preclinical tumor models with very low immune cell infiltration and for investigating how therapeutic treatments may modulate the immune landscape.
Collapse
Affiliation(s)
| | - Kathryn Que
- Bristol-Myers Squibb, Redwood City, California, USA
| | | | - Soo Min Koo
- Bristol-Myers Squibb, Redwood City, California, USA
| | | | | | - Jeong Kim
- Bristol-Myers Squibb, Redwood City, California, USA
| | | | | |
Collapse
|
5
|
Alanzi A, Moussa AY, Mothana RA, Abbas M, Ali I. In silico exploration of PD-L1 binding compounds: Structure-based virtual screening, molecular docking, and MD simulation. PLoS One 2024; 19:e0306804. [PMID: 39121024 PMCID: PMC11315321 DOI: 10.1371/journal.pone.0306804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 06/23/2024] [Indexed: 08/11/2024] Open
Abstract
Programmed death-ligand 1 (PD-L1), a transmembrane protein, is associated with the regulation of immune system. It frequently has overexpression in various cancers, allowing tumor cells to avoid immune detection. PD-L1 inhibition has risen as a potential strategy in the field of therapeutic immunology for cancer. In the current study, structure-based virtual screening of drug libraries was conducted and then the screened hits were docked to the active residues of PD-L1 to select the optimal binding poses. The top ten compounds with binding affinities ranging from -10.734 to -10.398 kcal/mol were selected for further analysis. The ADMET analysis of selected compounds showed the compounds meet the criteria of ADMET properties. Further, the conformational changes and binding stability of the top two compounds was analyzed by conducting 200 ns simulation and it was observed that the hits did not exert conformational changes to the protein structure. All the results suggest that the chosen hits can be considered as lead compounds for the inhibition of biological activity of PD-L1 in in vitro studies.
Collapse
Affiliation(s)
- Abdullah Alanzi
- Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Ashaimaa Y. Moussa
- Department of Pharmacognosy, Faculty of Pharmacy, Ain-Shams University, Cairo, Egypt
| | - Ramzi A. Mothana
- Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Munawar Abbas
- College of Food Science and Technology, Henan University of Technology, Zhengzhou, Henan, China
| | - Ijaz Ali
- Centre for Applied Mathematics and Bioinformatics (CAMB), Gulf University for Science and Technology, Hawally, Kuwait
| |
Collapse
|
6
|
Ding X, Zhang L, Fan M, Li L. TME-NET: an interpretable deep neural network for predicting pan-cancer immune checkpoint inhibitor responses. Brief Bioinform 2024; 25:bbae410. [PMID: 39167797 PMCID: PMC11337220 DOI: 10.1093/bib/bbae410] [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: 05/24/2024] [Revised: 07/17/2024] [Accepted: 08/02/2024] [Indexed: 08/23/2024] Open
Abstract
Immunotherapy with immune checkpoint inhibitors (ICIs) is increasingly used to treat various tumor types. Determining patient responses to ICIs presents a significant clinical challenge. Although components of the tumor microenvironment (TME) are used to predict patient outcomes, comprehensive assessments of the TME are frequently overlooked. Using a top-down approach, the TME was divided into five layers-outcome, immune role, cell, cellular component, and gene. Using this structure, a neural network called TME-NET was developed to predict responses to ICIs. Model parameter weights and cell ablation studies were used to investigate the influence of TME components. The model was developed and evaluated using a pan-cancer cohort of 948 patients across four cancer types, with Area Under the Curve (AUC) and accuracy as performance metrics. Results show that TME-NET surpasses established models such as support vector machine and k-nearest neighbors in AUC and accuracy. Visualization of model parameter weights showed that at the cellular layer, Th1 cells enhance immune responses, whereas myeloid-derived suppressor cells and M2 macrophages show strong immunosuppressive effects. Cell ablation studies further confirmed the impact of these cells. At the gene layer, the transcription factors STAT4 in Th1 cells and IRF4 in M2 macrophages significantly affect TME dynamics. Additionally, the cytokine-encoding genes IFNG from Th1 cells and ARG1 from M2 macrophages are crucial for modulating immune responses within the TME. Survival data from immunotherapy cohorts confirmed the prognostic ability of these markers, with p-values <0.01. In summary, TME-NET performs well in predicting immunotherapy responses and offers interpretable insights into the immunotherapy process. It can be customized at https://immbal.shinyapps.io/TME-NET.
Collapse
Affiliation(s)
- Xiaobao Ding
- Institute of Biomedical Engineering and Instrumentation, Hangzhou Dianzi University, Hangzhou 310018, Zhejiang, China
- Institute of Big Data and Artificial Intelligence in Medicine, School of Electronics and Information Engineering, Taizhou University, Taizhou 318000, Zhejiang, China
- School of Computer Science and Technology, Hangzhou Dianzi University, Hangzhou, 310018, China
| | - Lin Zhang
- Institute of Biomedical Engineering and Instrumentation, Hangzhou Dianzi University, Hangzhou 310018, Zhejiang, China
| | - Ming Fan
- Institute of Biomedical Engineering and Instrumentation, Hangzhou Dianzi University, Hangzhou 310018, Zhejiang, China
| | - Lihua Li
- Institute of Biomedical Engineering and Instrumentation, Hangzhou Dianzi University, Hangzhou 310018, Zhejiang, China
- School of Computer Science and Technology, Hangzhou Dianzi University, Hangzhou, 310018, China
| |
Collapse
|
7
|
Fan Y, Dai T, Zhang D, Guo H, Zhou F, Shi B, Wang S, Ji Z, Wang C, Yao X, Wei Q, Chen N, Xing J, Yang J, Kong C, Huang J, Ye D, Zhou L. PD-L1 expression and its correlation with tumor biomarkers in Chinese urothelial bladder cancer. Sci Rep 2024; 14:16753. [PMID: 39033240 PMCID: PMC11271459 DOI: 10.1038/s41598-024-67508-6] [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: 09/08/2023] [Accepted: 07/11/2024] [Indexed: 07/23/2024] Open
Abstract
Data on prevalence of programmed death ligand-1 (PD-L1) expression and its correlation with tumor biomarkers in Chinese patients with muscle-invasive urothelial bladder cancer (MIUBC) are scarce. We investigated the prevalence of PD-L1 expression, PD-L1 expression in tumor cells (TC) and immune cells (IC), and its correlation with tumor biomarkers (CD8+ T cells and tumor mutation burden [TMB]) in Chinese patients with newly diagnosed MIUBC (NCT03433924). Of 248 patients enrolled, 229 with PD-L1 data available were analysed. High PD-L1 expression (≥ 25% of TC or IC with PD-L1 expression) was observed in 120 (52.4%) patients. 59 cases showed positive staining in ≥ 25% of TC, and 82 cases had positive staining in ≥ 25% of IC. High expression of CD8+ T cell and TMB (> 10 mutations/megabase) was observed in 44.5% and 54.1% patients, respectively. A positive correlation was observed between percentage of TC with membrane PD-L1 positivity and CD8+ T cells (0.34; P < 0.001) and between IC with membrane PD-L1 positivity and CD8+ T cells (0.44; P < 0.001). There is high prevalence of PD-L1 expression in Chinese patients with MIUBC, suggesting that a sizable subset of patients could benefit from immunotherapy. The correlation of PD-L1 expression with tumor biomarkers provide clues for mechanisms underlying the effects of biomarkers for predicting efficacy.
Collapse
Affiliation(s)
- Yu Fan
- Department of Urology, Peking University First Hospital, Beijing, 100034, China
| | - Tao Dai
- Department of Urology, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University and Hunan Cancer Hospital, Changsha, 410006, China
| | - Dahong Zhang
- Department of Urology, Zhejiang Provincial People's Hospital, Hangzhou, 310005, Zhejiang, China
| | - Hongqian Guo
- Department of Urology, Drum Tower Hospital, Medical School of Nanjing University, Institute of Urology, Nanjing University, Nanjing, 210000, Jiangsu, China
| | - Fangjian Zhou
- Department of Urology, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Benkang Shi
- Department of Urology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Shaogang Wang
- Department of Urology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiafang Avenue, Qiaokou, Wuhan, 430000, Hubei, China
| | - Zhigang Ji
- Department of Urology, Peking Union Medical College Hospital, Chinese Academy of Science, Beijing, 100005, China
| | - Chunxi Wang
- Department of Urology, First Hospital of Jilin University, Changchun, 130000, Jilin, China
| | - Xudong Yao
- Department of Urology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200040, China
| | - Qiang Wei
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Nanhui Chen
- Department of Urology, Meizhou People's Hospital, Meizhou, 514031, Guangdong, China
| | - Jinchun Xing
- Department of Urology Surgery, The First Affiliated Hospital of Xiamen University, Xiamen, 361003, Fujian, China
| | - Jinjian Yang
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Chuize Kong
- Department of Urology, The First Hospital of China Medical University, Shenyang, 110001, China
| | - Jian Huang
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510000, China
| | - Dingwei Ye
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
| | - Liqun Zhou
- Department of Urology, Peking University First Hospital, Beijing, 100034, China.
| |
Collapse
|
8
|
Wang X, Xu W, Wang Z, Yu Q, Yuan L, Liu Y, Sang J, Li W, Zhu S, Jiang W, Li Z, Zhang W, Dang Y. Sokotrasterol Sulfate Suppresses IFN-γ-Induced PD-L1 Expression by Inhibiting JAK Activity. JOURNAL OF NATURAL PRODUCTS 2024; 87:713-721. [PMID: 38417168 DOI: 10.1021/acs.jnatprod.3c00811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/01/2024]
Abstract
PD-1/PD-L1 monoclonal antibodies exhibit promising therapeutic effectiveness in multiple cancers. However, developing a simple and efficient non-antibody treatment strategy using the PD-1/PD-L1 signaling pathway still remains challenging. In this study, we developed a flow cytometry assay to screen bioactive compounds with PD-L1 inhibitory activity. A total of 409 marine natural products were screened, and sokotrasterol sulfate (SKS) was found to efficiently suppress the IFN-γ-induced PD-L1 expression. SKS sensitizes the tumor cells to antigen-specific T-cell killing in the T cell-tumor cell coculture system. Mechanistically, SKS directly targeted Janus kinase (JAK) to inhibit the downstream activation of signal transducer and activator of transcription (STAT) and the subsequent transcription of PDL1. Our findings highlight the immunological role of SKS that may act as a basis for a potential immunotherapeutic agent.
Collapse
Affiliation(s)
- Xiaobo Wang
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, 131 Dong-An Road, Shanghai 200032, People's Republic of China
| | - Wenlong Xu
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, 131 Dong-An Road, Shanghai 200032, People's Republic of China
| | - Zengyiyi Wang
- School of Medicine, Tongji University, 1238 Si-Ping Road, Shanghai 200092, People's Republic of China
| | - Qian Yu
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, 131 Dong-An Road, Shanghai 200032, People's Republic of China
| | - Li Yuan
- School of Pharmacy, Naval Medical University, 325 Guo-He Road, Shanghai 200433, People's Republic of China
| | - Yihang Liu
- Basic Medicine Research and Innovation Center for Novel Target and Therapeutic Intervention, Ministry of Education, Institute of Life Sciences, The Second Affiliated Hospital of Chongqing Medical University, College of Pharmacy, Chongqing Medical University, 1 Yi-Xue-Yuan Road, Chongqing 400010, People's Republic of China
| | - Jinpeng Sang
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, People's Republic of China
| | - Wei Li
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, People's Republic of China
| | - Sanyong Zhu
- Basic Medicine Research and Innovation Center for Novel Target and Therapeutic Intervention, Ministry of Education, Institute of Life Sciences, The Second Affiliated Hospital of Chongqing Medical University, College of Pharmacy, Chongqing Medical University, 1 Yi-Xue-Yuan Road, Chongqing 400010, People's Republic of China
- Key Laboratory of Marine Drugs, Ministry of Education & Shandong Provincial Key Laboratory of Glycoscience and Glycotechnology, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, People's Republic of China
| | - Wei Jiang
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, 131 Dong-An Road, Shanghai 200032, People's Republic of China
| | - Zengxia Li
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, 131 Dong-An Road, Shanghai 200032, People's Republic of China
| | - Wen Zhang
- School of Medicine, Tongji University, 1238 Si-Ping Road, Shanghai 200092, People's Republic of China
- School of Pharmacy, Naval Medical University, 325 Guo-He Road, Shanghai 200433, People's Republic of China
| | - Yongjun Dang
- Basic Medicine Research and Innovation Center for Novel Target and Therapeutic Intervention, Ministry of Education, Institute of Life Sciences, The Second Affiliated Hospital of Chongqing Medical University, College of Pharmacy, Chongqing Medical University, 1 Yi-Xue-Yuan Road, Chongqing 400010, People's Republic of China
| |
Collapse
|
9
|
Stergiou IE, Palamaris K, Levidou G, Tzimou M, Papadakos SP, Mandrakis G, Masaoutis C, Rontogianni D, Theocharis S. PD-L1 Expression in Neoplastic and Immune Cells of Thymic Epithelial Tumors: Correlations with Disease Characteristics and HDAC Expression. Biomedicines 2024; 12:772. [PMID: 38672128 PMCID: PMC11048374 DOI: 10.3390/biomedicines12040772] [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: 02/21/2024] [Revised: 03/26/2024] [Accepted: 03/28/2024] [Indexed: 04/28/2024] Open
Abstract
BACKGROUND Programmed death-ligand 1 (PD-L1) expression in neoplastic and immune cells of the tumor microenvironment determines the efficacy of antitumor immunity, while it can be regulated at the epigenetic level by various factors, including HDACs. In this study, we aim to evaluate the expression patterns of PD-L1 in thymic epithelial tumors (TETs), while we attempt the first correlation analysis between PD-L1 and histone deacetylases (HDACs) expression. METHODS Immunohistochemistry was used to evaluate the expression of PD-L1 in tumor and immune cells of 91 TETs with SP263 and SP142 antibody clones, as well as the expressions of HDCA1, -2, -3, -4, -5, and -6. RESULTS The PD-L1 tumor proportion score (TPS) was higher, while the immune cell score (IC-score) was lower in the more aggressive TET subtypes and in more advanced Masaoka-Koga stages. A positive correlation between PD-L1 and HDAC-3, -4, and -5 cytoplasmic expression was identified. CONCLUSIONS Higher PD-L1 expression in neoplastic cells and lower PD-L1 expression in immune cells of TETs characterizes more aggressive and advanced neoplasms. Correlations between PD-L1 and HDAC expression unravel the impact of epigenetic regulation on the expression of immune checkpoint molecules in TETs, with possible future applications in combined therapeutic targeting.
Collapse
Affiliation(s)
- Ioanna E. Stergiou
- Department of Pathophysiology, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece;
- First Department of Pathology, School of Medicine, National and Kapodistrian University of Athens, 10679 Athens, Greece; (K.P.); (M.T.); (S.P.P.); (G.M.); (C.M.)
| | - Kostas Palamaris
- First Department of Pathology, School of Medicine, National and Kapodistrian University of Athens, 10679 Athens, Greece; (K.P.); (M.T.); (S.P.P.); (G.M.); (C.M.)
| | - Georgia Levidou
- Second Department of Pathology, Paracelsus Medical University, 90419 Nurenberg, Germany
| | - Maria Tzimou
- First Department of Pathology, School of Medicine, National and Kapodistrian University of Athens, 10679 Athens, Greece; (K.P.); (M.T.); (S.P.P.); (G.M.); (C.M.)
| | - Stavros P. Papadakos
- First Department of Pathology, School of Medicine, National and Kapodistrian University of Athens, 10679 Athens, Greece; (K.P.); (M.T.); (S.P.P.); (G.M.); (C.M.)
| | - Georgios Mandrakis
- First Department of Pathology, School of Medicine, National and Kapodistrian University of Athens, 10679 Athens, Greece; (K.P.); (M.T.); (S.P.P.); (G.M.); (C.M.)
| | - Christos Masaoutis
- First Department of Pathology, School of Medicine, National and Kapodistrian University of Athens, 10679 Athens, Greece; (K.P.); (M.T.); (S.P.P.); (G.M.); (C.M.)
| | - Dimitra Rontogianni
- Department of Pathology, Evangelismos General Hospital of Athens, 10676 Athens, Greece;
| | - Stamatios Theocharis
- First Department of Pathology, School of Medicine, National and Kapodistrian University of Athens, 10679 Athens, Greece; (K.P.); (M.T.); (S.P.P.); (G.M.); (C.M.)
| |
Collapse
|
10
|
Zhang H, Luo Y, Zhao X, Liu X. Engineering Proteus mirabilis improves antitumor efficacy via enhancing cytotoxic T cell responses. MOLECULAR THERAPY. ONCOLOGY 2024; 32:200770. [PMID: 38596299 PMCID: PMC10937320 DOI: 10.1016/j.omton.2024.200770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 01/19/2024] [Accepted: 02/01/2024] [Indexed: 04/11/2024]
Abstract
Cancer immunotherapy based on bioengineering of bacteria can effectively increase anticancer immune responses. However, few studies have investigated the antitumor potential of engineering Proteus mirabilis. Here, we genetically engineered P. mirabilis to overexpress Vibrio vulnificus flagellin B (FlaB) protein in a murine CT26 tumor model. We found that a large number of FlaB-expressing P. mirabilis colonized tumor tissues, enhanced T cell infiltration and secretion of cytokines and cytotoxic proteins in tumors, and significantly restrained tumor growth. Our results also showed that programmed death ligand 1 (PD-L1) expression in tumor-infiltrating immune cells was elevated after treatment with FlaB-expressing P. mirabilis. In addition, combination therapy with FlaB-expressing P. mirabilis and PD-L1 blockade synergistically improved antitumor efficacy by enhancing infiltration of CD8+ cells. Furthermore, serum liver biochemical indices of mice increased in the short term in both the P. mirabilis and the FlaB-expressing P. mirabilis treatment groups but gradually recovered in the later stage of treatment so that FlaB protein expression did not increase the toxicity of P. mirabilis in vivo. Taken together, our results suggest that P. mirabilis could serve as an engineered bacterium for bacterium-based cancer immunotherapy.
Collapse
Affiliation(s)
- Hong Zhang
- Institute of Tropical Agriculture and Forestry, Hainan University, Haikou, Hainan 570228, P.R. China
- College of Animal Science and Technology, Anhui Agricultural University, Heifei, Anhui 230036, P.R. China
| | - Yinlin Luo
- Institute of Tropical Agriculture and Forestry, Hainan University, Haikou, Hainan 570228, P.R. China
| | - Xincheng Zhao
- China Animal Disease Control Center, Beijing 100026, P.R. China
| | - Xiande Liu
- School of Life and Health, Hainan University, Haikou, Hainan 570228, P.R. China
| |
Collapse
|
11
|
Wang Y, Zhu T, Shi Q, Zhu G, Zhu S, Hou F. Tumor-draining lymph nodes: opportunities, challenges, and future directions in colorectal cancer immunotherapy. J Immunother Cancer 2024; 12:e008026. [PMID: 38242718 PMCID: PMC10806546 DOI: 10.1136/jitc-2023-008026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/07/2024] [Indexed: 01/21/2024] Open
Abstract
Tumor-draining lymph nodes (TDLNs) are potential immunotherapy targets that could expand the population of patients with colorectal cancer (CRC) who may benefit from immunotherapy. Currently, pathological detection of tumor cell infiltration limits the acquisition of immune information related to the resected lymph nodes. Understanding the immune function and metastatic risk of specific stages of lymph nodes can facilitate better discussions on the removal or preservation of lymph nodes, as well as the timing of immunotherapy. This review summarized the contribution of TDLNs to CRC responses to immune checkpoint blockade therapy, local immunotherapy, adoptive cell therapy, and cancer vaccines, and discussed the significance of these findings for the development of diagnostics based on TDLNs and the potential implications for guiding immunotherapy after a definitive diagnosis. Molecular pathology and immune spectrum diagnosis of TDLNs will promote significant advances in the selection of immunotherapy options and predicting treatment efficacy.
Collapse
Affiliation(s)
- Yao Wang
- Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Tingting Zhu
- Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Qi Shi
- Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Guanghui Zhu
- Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Siwei Zhu
- Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Fenggang Hou
- Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
12
|
Freen-van Heeren JJ. Employing CRISPR-Cas9 to Enhance T Cell Effector Function. Methods Mol Biol 2024; 2782:195-208. [PMID: 38622404 DOI: 10.1007/978-1-0716-3754-8_16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/17/2024]
Abstract
As part of the adaptive immune system, T cells are critical to maintain immune homeostasis. T cells provide protective immunity by killing infected cells and combatting cancerous cells. To do so, T cells produce and secrete effector molecules, such as granzymes, perforin, and cytokines such as tumor necrosis factor α and interferon γ. However, in immune suppressive environments, such as tumors, T cells gradually lose the capacity to perform their effector function. One way T cell effector function can be enhanced is through genetic engineering with tools such as clustered regularly interspaced short palindromic repeat (CRISPR)/CRISPR-associated protein 9 (Cas9). This protocol explains in a step-by-step fashion how to perform a controlled electroporation-based CRISPR experiment to enhance human T cell effector function. Of note, these steps are suitable for CRISPR-mediated genome editing in T cells in general and can thus also be used to study proteins of interest that do not influence T cell effector function.
Collapse
|
13
|
Bredel D, Tihic E, Mouraud S, Danlos FX, Susini S, Aglave M, Alfaro A, Mohamed-Djalim C, Rouanne M, Halse H, Bigorgne A, Tselikas L, Dalle S, Hartl DM, Baudin E, Guettier C, Vibert E, Rosmorduc O, Robert C, Ferlicot S, Parier B, Albiges L, de Montpreville VT, Besse B, Mercier O, Even C, Breuskin I, Classe M, Radulescu C, Lebret T, Pautier P, Gouy S, Scoazec JY, Zitvogel L, Marabelle A, Bonvalet M. Immune checkpoints are predominantly co-expressed by clonally expanded CD4 +FoxP3 + intratumoral T-cells in primary human cancers. J Exp Clin Cancer Res 2023; 42:333. [PMID: 38057799 DOI: 10.1186/s13046-023-02897-6] [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: 07/14/2023] [Accepted: 11/11/2023] [Indexed: 12/08/2023] Open
Abstract
BACKGROUND In addition to anti-PD(L)1, anti-CTLA-4 and anti-LAG-3, novel immune checkpoint proteins (ICP)-targeted antibodies have recently failed to demonstrate significant efficacy in clinical trials. In these trials, patients were enrolled without screening for drug target expression. Although these novel ICP-targeted antibodies were expected to stimulate anti-tumor CD8 + T-cells, the rationale for their target expression in human tumors relied on pre-clinical IHC stainings and transcriptomic data, which are poorly sensitive and specific techniques for assessing membrane protein expression on immune cell subsets. Our aim was to describe ICP expression on intratumoral T-cells from primary solid tumors to better design upcoming neoadjuvant cancer immunotherapy trials. METHODS We prospectively performed multiparameter flow cytometry and single-cell RNA sequencing (scRNA-Seq) paired with TCR sequencing on freshly resected human primary tumors of various histological types to precisely determine ICP expression levels within T-cell subsets. RESULTS Within a given tumor type, we found high inter-individual variability for tumor infiltrating CD45 + cells and for T-cells subsets. The proportions of CD8+ T-cells (~ 40%), CD4+ FoxP3- T-cells (~ 40%) and CD4+ FoxP3+ T-cells (~ 10%) were consistent across patients and indications. Intriguingly, both stimulatory (CD25, CD28, 4-1BB, ICOS, OX40) and inhibitory (PD-1, CTLA-4, PD-L1, CD39 and TIGIT) checkpoint proteins were predominantly co-expressed by intratumoral CD4+FoxP3+ T-cells. ScRNA-Seq paired with TCR sequencing revealed that T-cells with high clonality and high ICP expressions comprised over 80% of FoxP3+ cells among CD4+ T-cells. Unsupervised clustering of flow cytometry and scRNAseq data identified subsets of CD8+ T-cells and of CD4+ FoxP3- T-cells expressing certain checkpoints, though these expressions were generally lower than in CD4+ FoxP3+ T-cell subsets, both in terms of proportions among total T-cells and ICP expression levels. CONCLUSIONS Tumor histology alone does not reveal the complete picture of the tumor immune contexture. In clinical trials, assumptions regarding target expression should rely on more sensitive and specific techniques than conventional IHC or transcriptomics. Flow cytometry and scRNAseq accurately characterize ICP expression within immune cell subsets. Much like in hematology, flow cytometry can better describe the immune contexture of solid tumors, offering the opportunity to guide patient treatment according to drug target expression rather than tumor histological type.
Collapse
Affiliation(s)
- Delphine Bredel
- Gustave Roussy, 114 Rue Édouard Vaillant, 94805, Villejuif, France
- Institut National de La Santé Et de La Recherche Médicale (INSERM) U1015, Laboratoire de Recherche Translationnelle en Immunothérapie (LRTI), 94805, Villejuif, France
| | - Edi Tihic
- Gustave Roussy, 114 Rue Édouard Vaillant, 94805, Villejuif, France
- Institut National de La Santé Et de La Recherche Médicale (INSERM) U1015, Laboratoire de Recherche Translationnelle en Immunothérapie (LRTI), 94805, Villejuif, France
- Institut National de La Santé Et de La Recherche Médicale (INSERM) CIC1428, Centre d'Investigation Clinique BIOTHERIS, 94805, Villejuif, France
| | - Séverine Mouraud
- Gustave Roussy, 114 Rue Édouard Vaillant, 94805, Villejuif, France
- Institut National de La Santé Et de La Recherche Médicale (INSERM) U1015, Laboratoire de Recherche Translationnelle en Immunothérapie (LRTI), 94805, Villejuif, France
- Institut National de La Santé Et de La Recherche Médicale (INSERM) CIC1428, Centre d'Investigation Clinique BIOTHERIS, 94805, Villejuif, France
| | - François-Xavier Danlos
- Gustave Roussy, 114 Rue Édouard Vaillant, 94805, Villejuif, France
- Institut National de La Santé Et de La Recherche Médicale (INSERM) U1015, Laboratoire de Recherche Translationnelle en Immunothérapie (LRTI), 94805, Villejuif, France
- Institut National de La Santé Et de La Recherche Médicale (INSERM) CIC1428, Centre d'Investigation Clinique BIOTHERIS, 94805, Villejuif, France
- Université Paris-Saclay, Faculté de Médecine, 94270, Le Kremlin-Bicêtre, France
- Gustave Roussy, Département d'Innovation Thérapeutique Et d'Essais Précoces (DITEP), 94805, Villejuif, France
| | - Sandrine Susini
- Gustave Roussy, 114 Rue Édouard Vaillant, 94805, Villejuif, France
- Institut National de La Santé Et de La Recherche Médicale (INSERM) U1015, Laboratoire de Recherche Translationnelle en Immunothérapie (LRTI), 94805, Villejuif, France
- Institut National de La Santé Et de La Recherche Médicale (INSERM) CIC1428, Centre d'Investigation Clinique BIOTHERIS, 94805, Villejuif, France
| | - Marine Aglave
- Gustave Roussy, Plateforme de bioinformatique, F-94805, Villejuif, France
| | - Alexia Alfaro
- Gustave Roussy, Université Paris-Saclay, UMS 23/3655, Plateforme Imagerie Et Cytométrie, Villejuif, France
| | - Chifaou Mohamed-Djalim
- Gustave Roussy, 114 Rue Édouard Vaillant, 94805, Villejuif, France
- Institut National de La Santé Et de La Recherche Médicale (INSERM) CIC1428, Centre d'Investigation Clinique BIOTHERIS, 94805, Villejuif, France
| | - Mathieu Rouanne
- Gustave Roussy, 114 Rue Édouard Vaillant, 94805, Villejuif, France
- Institut National de La Santé Et de La Recherche Médicale (INSERM) U1015, Laboratoire de Recherche Translationnelle en Immunothérapie (LRTI), 94805, Villejuif, France
- Institut National de La Santé Et de La Recherche Médicale (INSERM) CIC1428, Centre d'Investigation Clinique BIOTHERIS, 94805, Villejuif, France
- Department of Microbiology and Immunology, Vagelos College of Physicians and Surgeons, Columbia University, New York, USA
| | - Héloise Halse
- Gustave Roussy, 114 Rue Édouard Vaillant, 94805, Villejuif, France
- Institut National de La Santé Et de La Recherche Médicale (INSERM) U1015, Laboratoire de Recherche Translationnelle en Immunothérapie (LRTI), 94805, Villejuif, France
- Institut National de La Santé Et de La Recherche Médicale (INSERM) CIC1428, Centre d'Investigation Clinique BIOTHERIS, 94805, Villejuif, France
- Institut National de La Santé Et de La Recherche Médicale (INSERM) U1163, Institut Imagine, Université Paris Descartes, 75015, Paris, France
| | - Amélie Bigorgne
- Gustave Roussy, 114 Rue Édouard Vaillant, 94805, Villejuif, France
- Institut National de La Santé Et de La Recherche Médicale (INSERM) U1015, Laboratoire de Recherche Translationnelle en Immunothérapie (LRTI), 94805, Villejuif, France
- Institut National de La Santé Et de La Recherche Médicale (INSERM) CIC1428, Centre d'Investigation Clinique BIOTHERIS, 94805, Villejuif, France
- Institut National de La Santé Et de La Recherche Médicale (INSERM) U1163, Institut Imagine, Université Paris Descartes, 75015, Paris, France
| | - Lambros Tselikas
- Gustave Roussy, 114 Rue Édouard Vaillant, 94805, Villejuif, France
- Institut National de La Santé Et de La Recherche Médicale (INSERM) U1015, Laboratoire de Recherche Translationnelle en Immunothérapie (LRTI), 94805, Villejuif, France
- Institut National de La Santé Et de La Recherche Médicale (INSERM) CIC1428, Centre d'Investigation Clinique BIOTHERIS, 94805, Villejuif, France
- Université Paris-Saclay, Faculté de Médecine, 94270, Le Kremlin-Bicêtre, France
- Gustave Roussy, Université Paris Saclay, Département d'Anesthésie, Chirurgie et Imagerie Interventionnelle, F-94805, Villejuif, France
| | - Stéphane Dalle
- Department of Dermatology, HCL Cancer Institute, Lyon Cancer Research Center, 69495, Lyon, France
| | - Dana M Hartl
- Gustave Roussy, Université Paris Saclay, Département d'Anesthésie, Chirurgie et Imagerie Interventionnelle, F-94805, Villejuif, France
| | - Eric Baudin
- Gustave Roussy, Département d'Oncologie Médicale, F-94805, Villejuif, France
| | - Catherine Guettier
- Université Paris-Saclay, Faculté de Médecine, 94270, Le Kremlin-Bicêtre, France
- Service d'Anatomie Pathologique, Hôpital Bicêtre, AP-HP, 94270, Le Kremlin-Bicêtre, France
- UMR-S 1193, Hôpital Paul Brousse Université Paris Saclay, 94800, Villejuif, France
| | - Eric Vibert
- UMR-S 1193, Hôpital Paul Brousse Université Paris Saclay, 94800, Villejuif, France
- Centre Hépato-Biliaire, Hôpital Paul Brousse, AP-HP, 94800, Villejuif, France
| | - Olivier Rosmorduc
- Centre Hépato-Biliaire, Hôpital Paul Brousse, AP-HP, 94800, Villejuif, France
| | - Caroline Robert
- Université Paris-Saclay, Faculté de Médecine, 94270, Le Kremlin-Bicêtre, France
- Gustave Roussy, Département d'Oncologie Médicale, F-94805, Villejuif, France
- Institut National de La Santé Et de La Recherche Médicale (INSERM) U981, Gustave Roussy, 94805, Villejuif, France
| | - Sophie Ferlicot
- Université Paris-Saclay, Faculté de Médecine, 94270, Le Kremlin-Bicêtre, France
- Service d'Anatomie Pathologique, Hôpital Bicêtre, AP-HP, 94270, Le Kremlin-Bicêtre, France
- Centre National de Recherche Scientifique (CNRS), Gustave Roussy, Université Paris-Saclay, UMR 9019, 94805, Villejuif, France
| | - Bastien Parier
- Service de Chirurgie Urologique, Hôpital Bicêtre, AP-HP, Le Kremlin-Bicêtre, France
| | - Laurence Albiges
- Université Paris-Saclay, Faculté de Médecine, 94270, Le Kremlin-Bicêtre, France
- Gustave Roussy, Département d'Oncologie Médicale, F-94805, Villejuif, France
| | | | - Benjamin Besse
- Université Paris-Saclay, Faculté de Médecine, 94270, Le Kremlin-Bicêtre, France
- Gustave Roussy, Département d'Oncologie Médicale, F-94805, Villejuif, France
| | - Olaf Mercier
- Université Paris-Saclay, Faculté de Médecine, 94270, Le Kremlin-Bicêtre, France
- Service de Chirurgie Thoracique Et Transplantation Cardio-Pulmonaire, Hôpital Marie-Lannelongue, UMR_S 999 INSERM, Université Paris-Saclay, GHPSJ, 92350, Le Plessis-Robinson, France
| | - Caroline Even
- Gustave Roussy, Département d'Oncologie Médicale, F-94805, Villejuif, France
| | - Ingrid Breuskin
- Gustave Roussy, Université Paris Saclay, Département d'Anesthésie, Chirurgie et Imagerie Interventionnelle, F-94805, Villejuif, France
| | - Marion Classe
- Gustave Roussy, Département de Biopathologie, F-94805, Villejuif, France
| | - Camélia Radulescu
- Département de Pathologie, Hôpital Foch, UVSQ, Université Paris-Saclay, 92150, Suresnes, France
| | - Thierry Lebret
- Département d'Urologie, Hôpital Foch, UVSQ-Université Paris-Saclay, 92150, Suresnes, France
| | - Patricia Pautier
- Gustave Roussy, Département d'Oncologie Médicale, F-94805, Villejuif, France
| | - Sébastien Gouy
- Gustave Roussy, Université Paris Saclay, Département d'Anesthésie, Chirurgie et Imagerie Interventionnelle, F-94805, Villejuif, France
| | - Jean-Yves Scoazec
- Gustave Roussy, Département de Biopathologie, F-94805, Villejuif, France
| | - Laurence Zitvogel
- Gustave Roussy, 114 Rue Édouard Vaillant, 94805, Villejuif, France
- Institut National de La Santé Et de La Recherche Médicale (INSERM) U1015, Laboratoire de Recherche Translationnelle en Immunothérapie (LRTI), 94805, Villejuif, France
- Institut National de La Santé Et de La Recherche Médicale (INSERM) CIC1428, Centre d'Investigation Clinique BIOTHERIS, 94805, Villejuif, France
- Université Paris-Saclay, Faculté de Médecine, 94270, Le Kremlin-Bicêtre, France
| | - Aurélien Marabelle
- Gustave Roussy, 114 Rue Édouard Vaillant, 94805, Villejuif, France.
- Institut National de La Santé Et de La Recherche Médicale (INSERM) U1015, Laboratoire de Recherche Translationnelle en Immunothérapie (LRTI), 94805, Villejuif, France.
- Institut National de La Santé Et de La Recherche Médicale (INSERM) CIC1428, Centre d'Investigation Clinique BIOTHERIS, 94805, Villejuif, France.
- Université Paris-Saclay, Faculté de Médecine, 94270, Le Kremlin-Bicêtre, France.
- Gustave Roussy, Département d'Innovation Thérapeutique Et d'Essais Précoces (DITEP), 94805, Villejuif, France.
| | - Mélodie Bonvalet
- Gustave Roussy, 114 Rue Édouard Vaillant, 94805, Villejuif, France
- Institut National de La Santé Et de La Recherche Médicale (INSERM) U1015, Laboratoire de Recherche Translationnelle en Immunothérapie (LRTI), 94805, Villejuif, France
- Institut National de La Santé Et de La Recherche Médicale (INSERM) CIC1428, Centre d'Investigation Clinique BIOTHERIS, 94805, Villejuif, France
| |
Collapse
|
14
|
Samejima H, Kojima K, Fujiwara A, Tokunaga T, Okishio K, Yoon H. The combination of PD-L1 expression and the neutrophil-to-lymphocyte ratio as a prognostic factor of postoperative recurrence in non-small-cell lung cancer: a retrospective cohort study. BMC Cancer 2023; 23:1107. [PMID: 37964220 PMCID: PMC10644552 DOI: 10.1186/s12885-023-11604-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 11/02/2023] [Indexed: 11/16/2023] Open
Abstract
BACKGROUND While PD-L1 expression and neutrophil-to-lymphocyte ratio (NLR) are prognostic biomarkers for lung cancer, few studies have considered their interaction. We hypothesized that the product of PD-L1 expression (tumor proportion score) and the NLR (PD-L1 × NLR) might be a postoperative prognostic marker reflecting the immune microenvironment of lung cancer. METHODS We analyzed the association between PD-L1 × NLR and postoperative recurrence-free survival in 647 patients with NSCLC using multivariable Cox proportional hazards models. RESULTS In the analysis of PD-L1 × NLR as a categorical variable, the group with PD-L1 × NLR ≥ 25.8 had a significantly higher hazard ratio (HR) than the group with < 25.8 (adjusted HR 1.78, 95% confidence interval [CI] 1.23-2.60). The adjusted HR for PD-L1 × NLR, considered a continuous variable, was 1.004 (95% CI, 1.002-1.006). The risk of postoperative recurrence increased by 1.004-fold for each unit increase in PD-L1 × NLR, and a more than 2-fold increase in risk was observed for values ≥ 170. CONCLUSIONS PD-L1 × NLR may be used in real-world clinical practice as a novel factor for predicting the risk of postoperative recurrence after lung cancer surgery.
Collapse
Affiliation(s)
- Hironobu Samejima
- Department of General Thoracic Surgery, National Hospital Organization Kinki-Chuo Chest Medical Center, 1180 Nagasone-Cho, Kita-Ku, Sakai-Shi, Osaka, 591-8555, Japan
| | - Kensuke Kojima
- Department of General Thoracic Surgery, National Hospital Organization Kinki-Chuo Chest Medical Center, 1180 Nagasone-Cho, Kita-Ku, Sakai-Shi, Osaka, 591-8555, Japan.
| | - Ayako Fujiwara
- Department of General Thoracic Surgery, National Hospital Organization Kinki-Chuo Chest Medical Center, 1180 Nagasone-Cho, Kita-Ku, Sakai-Shi, Osaka, 591-8555, Japan
| | - Toshiteru Tokunaga
- Department of General Thoracic Surgery, National Hospital Organization Kinki-Chuo Chest Medical Center, 1180 Nagasone-Cho, Kita-Ku, Sakai-Shi, Osaka, 591-8555, Japan
| | - Kyoichi Okishio
- Clinical Research Center, National Hospital Organization Kinki-Chuo Chest Medical Center, Osaka, Japan
- Department of Thoracic Oncology, National Hospital Organization Kinki-Chuo Chest Medical Center, Osaka, Japan
| | - Hyungeun Yoon
- Department of General Thoracic Surgery, National Hospital Organization Kinki-Chuo Chest Medical Center, 1180 Nagasone-Cho, Kita-Ku, Sakai-Shi, Osaka, 591-8555, Japan
| |
Collapse
|
15
|
Allemailem KS, Alsahli MA, Almatroudi A, Alrumaihi F, Al Abdulmonem W, Moawad AA, Alwanian WM, Almansour NM, Rahmani AH, Khan AA. Innovative Strategies of Reprogramming Immune System Cells by Targeting CRISPR/Cas9-Based Genome-Editing Tools: A New Era of Cancer Management. Int J Nanomedicine 2023; 18:5531-5559. [PMID: 37795042 PMCID: PMC10547015 DOI: 10.2147/ijn.s424872] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2023] [Accepted: 09/16/2023] [Indexed: 10/06/2023] Open
Abstract
The recent developments in the study of clustered regularly interspaced short palindromic repeats/associated protein 9 (CRISPR/Cas9) system have revolutionized the art of genome-editing and its applications for cellular differentiation and immune response behavior. This technology has further helped in understanding the mysteries of cancer progression and possible designing of novel antitumor immunotherapies. CRISPR/Cas9-based genome-editing is now often used to engineer universal T-cells, equipped with recombinant T-cell receptor (TCR) or chimeric antigen receptor (CAR). In addition, this technology is used in cytokine stimulation, antibody designing, natural killer (NK) cell transfer, and to overcome immune checkpoints. The innovative potential of CRISPR/Cas9 in preparing the building blocks of adoptive cell transfer (ACT) immunotherapy has opened a new window of antitumor immunotherapy and some of them have gained FDA approval. The manipulation of immunogenetic regulators has opened a new interface for designing, implementation and interpretation of CRISPR/Cas9-based screening in immuno-oncology. Several cancers like lymphoma, melanoma, lung, and liver malignancies have been treated with this strategy, once thought to be impossible. The safe and efficient delivery of CRISPR/Cas9 system within the immune cells for the genome-editing strategy is a challenging task which needs to be sorted out for efficient immunotherapy. Several targeting approaches like virus-mediated, electroporation, microinjection and nanoformulation-based methods have been used, but each procedure offers some limitations. Here, we elaborate the recent updates of cancer management through immunotherapy in partnership with CRISPR/Cas9 technology. Further, some innovative methods of targeting this genome-editing system within the immune system cells for reprogramming them, as a novel strategy of anticancer immunotherapy is elaborated. In addition, future prospects and clinical trials are also discussed.
Collapse
Affiliation(s)
- Khaled S Allemailem
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah, Saudi Arabia
| | - Mohammed A Alsahli
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah, Saudi Arabia
| | - Ahmad Almatroudi
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah, Saudi Arabia
| | - Faris Alrumaihi
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah, Saudi Arabia
| | - Waleed Al Abdulmonem
- Department of Pathology, College of Medicine, Qassim University, Buraydah, Saudi Arabia
| | - Amira A Moawad
- Friedrich-Loeffler-Institut, Institute of Bacterial Infections and Zoonoses, Jena, Germany
| | - Wanian M Alwanian
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah, Saudi Arabia
| | - Nahlah Makki Almansour
- Department of Biology, College of Science, University of Hafr Al Batin, Hafr Al Batin, Saudi Arabia
| | - Arshad Husain Rahmani
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah, Saudi Arabia
| | - Amjad Ali Khan
- Department of Basic Health Sciences, College of Applied Medical Sciences, Qassim University, Buraydah, Saudi Arabia
| |
Collapse
|
16
|
Sharma P, Joshi RV, Pritchard R, Xu K, Eicher MA. Therapeutic Antibodies in Medicine. Molecules 2023; 28:6438. [PMID: 37764213 PMCID: PMC10535987 DOI: 10.3390/molecules28186438] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Revised: 08/05/2023] [Accepted: 08/28/2023] [Indexed: 09/29/2023] Open
Abstract
Antibody engineering has developed into a wide-reaching field, impacting a multitude of industries, most notably healthcare and diagnostics. The seminal work on developing the first monoclonal antibody four decades ago has witnessed exponential growth in the last 10-15 years, where regulators have approved monoclonal antibodies as therapeutics and for several diagnostic applications, including the remarkable attention it garnered during the pandemic. In recent years, antibodies have become the fastest-growing class of biological drugs approved for the treatment of a wide range of diseases, from cancer to autoimmune conditions. This review discusses the field of therapeutic antibodies as it stands today. It summarizes and outlines the clinical relevance and application of therapeutic antibodies in treating a landscape of diseases in different disciplines of medicine. It discusses the nomenclature, various approaches to antibody therapies, and the evolution of antibody therapeutics. It also discusses the risk profile and adverse immune reactions associated with the antibodies and sheds light on future applications and perspectives in antibody drug discovery.
Collapse
Affiliation(s)
- Prerna Sharma
- Geisinger Commonwealth School of Medicine, Scranton, PA 18509, USA
| | | | | | | | | |
Collapse
|
17
|
Varlı M, Ngo MT, Kim SM, Taş İ, Zhou R, Gamage CD, Pulat S, Park SY, Sesal NC, Hur JS, Kang KB, Kim H. A fatty acid-rich fraction of an endolichenic fungus Phoma sp. suppresses immune checkpoint markers via AhR/ARNT and ESR1. Heliyon 2023; 9:e19185. [PMID: 37662726 PMCID: PMC10474435 DOI: 10.1016/j.heliyon.2023.e19185] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 07/29/2023] [Accepted: 08/15/2023] [Indexed: 09/05/2023] Open
Abstract
Lung cancer has the highest mortality rates worldwide. The disease is caused by environmental pollutants, smoking, and many other factors. Recent treatments include immunotherapeutics, which have shown some success; however, the search for new therapeutics is ongoing. Endolichenic fungi produce a whale of a lot of secondary metabolites, the therapeutic effects of which are being evaluated. Here, we used a crude extract and subfractions of the endolichenic fungus, Phoma sp. (EL006848), isolated from the Pseudevernia furfuracea. It was identified the fatty acid components, palmitic acid, stearic acid, and oleic acid, exist in subfractions E1 and E2. In addition, EL006848 and its fatty acids fractions suppressed benzo[a]pyrene (an AhR ligand)- induced expression of PD-L1 to inhibit the activity of multiple immune checkpoints. E2 subfraction, which had a higher fatty acid content than E1, downregulated expression of AhR/ARNT and several human transcription factors related to ESR1. Moreover, E2 showed a strong inhibitory effect on STAT3 expression and mild effect on NF-kB activity. These results suggest that fatty acids extracted from an endolichenic fungus can exert strong immunotherapeutic effects.
Collapse
Affiliation(s)
- Mücahit Varlı
- College of Pharmacy, Sunchon National University, 255 Jungang-ro, Sunchon, Jeonnam 57922, Republic of Korea
| | - Men Thi Ngo
- College of Pharmacy, Sookmyung Women's University, 100 Cheongpa-ro 47 gil, Seoul 04310, Republic of Korea
| | - Seong-Min Kim
- College of Pharmacy, Sunchon National University, 255 Jungang-ro, Sunchon, Jeonnam 57922, Republic of Korea
| | - İsa Taş
- College of Pharmacy, Sunchon National University, 255 Jungang-ro, Sunchon, Jeonnam 57922, Republic of Korea
| | - Rui Zhou
- College of Pharmacy, Sunchon National University, 255 Jungang-ro, Sunchon, Jeonnam 57922, Republic of Korea
| | - Chathurika D.B. Gamage
- College of Pharmacy, Sunchon National University, 255 Jungang-ro, Sunchon, Jeonnam 57922, Republic of Korea
| | - Sultan Pulat
- College of Pharmacy, Sunchon National University, 255 Jungang-ro, Sunchon, Jeonnam 57922, Republic of Korea
| | - So-Yeon Park
- College of Pharmacy, Sunchon National University, 255 Jungang-ro, Sunchon, Jeonnam 57922, Republic of Korea
| | - Nüzhet Cenk Sesal
- Faculty of Arts and Sciences, Department of Biology, Marmara University, Istanbul, Turkey
| | - Jae-Seoun Hur
- Korean Lichen Research Institute, Sunchon National University, 255 Jungang-ro, Sunchon, Jeonnam 57922, Republic of Korea
| | - Kyo Bin Kang
- College of Pharmacy, Sookmyung Women's University, 100 Cheongpa-ro 47 gil, Seoul 04310, Republic of Korea
| | - Hangun Kim
- College of Pharmacy, Sunchon National University, 255 Jungang-ro, Sunchon, Jeonnam 57922, Republic of Korea
| |
Collapse
|
18
|
Puopolo T, Li H, Gutkowski J, Cai A, Seeram NP, Ma H, Liu C. Establishment of Human PD-1/PD-L1 Blockade Assay Based on Surface Plasmon Resonance (SPR) Biosensor. Bio Protoc 2023; 13:e4765. [PMID: 37575393 PMCID: PMC10415205 DOI: 10.21769/bioprotoc.4765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 05/03/2023] [Accepted: 05/31/2023] [Indexed: 08/15/2023] Open
Abstract
Blockade of the programmed cell death protein 1 (PD-1)/PD-ligand 1 (PD-L1) axis is a promising strategy for cancer immunotherapy. Although antibody-based PD-1/PD-L1 inhibitors have shown remarkable results in clinical cancer studies, their inherent limitations underscore the significance of developing novel PD-1/PD-L1 inhibitors. Small molecule inhibitors have several advantages over antibody-based inhibitors, including favorable tumor penetration and oral bioavailability, fewer side effects, easier administration, preferred biological half-life, and lower cost. However, small molecule inhibitors that directly target the PD-1/PD-L1 interaction are still in the early development stage, partially due to the lack of reliable biophysical assays. Herein, we present a novel PD-1/PD-L1 blockade assay using a surface plasmon resonance (SPR)-based technique. This blockade assay immobilizes human PD-1 on a sensor chip, which interacts with PD-L1 inhibitors or negative PD-L1 binders with human PD-L1 protein at a range of molecular ratios. The binding kinetics of PD-L1 to PD-1 and the blockade rates of small molecules were determined. Compared to other techniques such as PD-1/PD-L1 pair enzyme-linked immunosorbent assay (ELISA) and AlphaLISA immunoassays, our SPR-based method offers real-time and label-free detection with advantages including shorter experimental runs and smaller sample quantity requirements. Key features A SPR protocol screens compounds for their capacity to block the PD-1/PD-L1 interaction. Validation of PD-1/PD-L1 interaction, followed by assessing blockade effects with known inhibitors BMS-1166 and BMS-202, and a negative control NO-Losartan A. Analysis of percentage blockade of PD-1/PD-L1 of the samples to obtain the IC50. Broad applications in the discovery of small molecule-based PD-1/PD-L1 inhibitors for cancer immunotherapy. Graphical overview.
Collapse
Affiliation(s)
- Tess Puopolo
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI 02881, USA
| | - Huifang Li
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI 02881, USA
| | - Justin Gutkowski
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI 02881, USA
| | - Ang Cai
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI 02881, USA
| | - Navindra P. Seeram
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI 02881, USA
| | - Hang Ma
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI 02881, USA
| | - Chang Liu
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI 02881, USA
| |
Collapse
|
19
|
Fernandez-Rodriguez L, Cianciaruso C, Bill R, Trefny MP, Klar R, Kirchhammer N, Buchi M, Festag J, Michel S, Kohler RH, Jones E, Maaske A, Vom Berg J, Kobold S, Kashyap AS, Jaschinski F, Dixon KO, Pittet MJ, Zippelius A. Dual TLR9 and PD-L1 targeting unleashes dendritic cells to induce durable antitumor immunity. J Immunother Cancer 2023; 11:e006714. [PMID: 37208130 PMCID: PMC10201251 DOI: 10.1136/jitc-2023-006714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/16/2023] [Indexed: 05/21/2023] Open
Abstract
BACKGROUND Although immune checkpoint inhibitors have been a breakthrough in clinical oncology, these therapies fail to produce durable responses in a significant fraction of patients. This lack of long-term efficacy may be due to a poor pre-existing network linking innate and adaptive immunity. Here, we present an antisense oligonucleotide (ASO)-based strategy that dually targets toll-like receptor 9 (TLR9) and programmed cell death ligand 1 (PD-L1), aiming to overcome resistance to anti-PD-L1 monoclonal therapy. METHODS We designed a high-affinity immunomodulatory IM-TLR9:PD-L1-ASO antisense oligonucleotide (hereafter, IM-T9P1-ASO) targeting mouse PD-L1 messenger RNA and activating TLR9. Then, we performed in vitro and in vivo studies to validate the IM-T9P1-ASO activity, efficacy, and biological effects in tumors and draining lymph nodes. We also performed intravital imaging to study IM-T9P1-ASO pharmacokinetics in the tumor. RESULTS IM-T9P1-ASO therapy, unlike PD-L1 antibody therapy, results in durable antitumor responses in multiple mouse cancer models. Mechanistically, IM-T9P1-ASO activates a state of tumor-associated dendritic cells (DCs), referred to here as DC3s, which have potent antitumor potential but express the PD-L1 checkpoint. IM-T9P1-ASO has two roles: it triggers the expansion of DC3s by engaging with TLR9 and downregulates PD-L1, thereby unleashing the antitumor functions of DC3s. This dual action leads to tumor rejection by T cells. The antitumor efficacy of IM-T9P1-ASO depends on the antitumor cytokine interleukin-12 (IL-12), produced by DC3s, and Batf3, a transcription factor required for DC development. CONCLUSIONS By simultaneously targeting TLR9 and PD-L1, IM-T9P1-ASO amplifies antitumor responses via DC activation, leading to sustained therapeutic efficacy in mice. By highlighting differences and similarities between mouse and human DCs, this study could serve to develop similar therapeutic strategies for patients with cancer.
Collapse
Affiliation(s)
| | - Chiara Cianciaruso
- Department of Pathology and Immunology, University of Geneva, Geneve, Switzerland
- AGORA Cancer Research Center, Lausanne, Switzerland
- Center for Systems Biology, Massachusetts General Hospital Research Institute and Harvard Medical School, Boston, Massachusetts, USA
| | - Ruben Bill
- Department of Pathology and Immunology, University of Geneva, Geneve, Switzerland
- AGORA Cancer Research Center, Lausanne, Switzerland
- Center for Systems Biology, Massachusetts General Hospital Research Institute and Harvard Medical School, Boston, Massachusetts, USA
| | - Marcel P Trefny
- Department of Biomedicine, University of Basel and University Hospital of Basel, Basel, Switzerland
| | - Richard Klar
- Secarna Pharmaceuticals GmbH & Co KG, Planegg Martinsried, Germany
| | - Nicole Kirchhammer
- Department of Biomedicine, University of Basel and University Hospital of Basel, Basel, Switzerland
| | - Mélanie Buchi
- Department of Biomedicine, University of Basel and University Hospital of Basel, Basel, Switzerland
| | - Julia Festag
- Secarna Pharmaceuticals GmbH & Co KG, Planegg Martinsried, Germany
| | - Sven Michel
- Secarna Pharmaceuticals GmbH & Co KG, Planegg Martinsried, Germany
| | - Rainer H Kohler
- Center for Systems Biology, Massachusetts General Hospital Research Institute and Harvard Medical School, Boston, Massachusetts, USA
| | - Elham Jones
- Department of Biomedicine, University of Basel and University Hospital of Basel, Basel, Switzerland
| | - Andre Maaske
- Secarna Pharmaceuticals GmbH & Co KG, Planegg Martinsried, Germany
| | - Johannes Vom Berg
- Institute of Laboratory Animal Science, University of Zurich, Zürich, Switzerland
| | - Sebastian Kobold
- Center of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, Department of Medicine IV, Klinikum der Universität München, LMU, Munich, Germany
- German Center for Lung Research (DZL), German Center for Translational Cancer Research (DKTK), partner site Munich, Munich, Germany
| | - Abhishek S Kashyap
- Department of Biomedicine, University of Basel and University Hospital of Basel, Basel, Switzerland
| | - Frank Jaschinski
- Secarna Pharmaceuticals GmbH & Co KG, Planegg Martinsried, Germany
| | - Karen O Dixon
- Department of Biomedicine, University of Basel and University Hospital of Basel, Basel, Switzerland
| | - Mikael J Pittet
- Department of Pathology and Immunology, University of Geneva, Geneve, Switzerland
- AGORA Cancer Research Center, Lausanne, Switzerland
- Center for Systems Biology, Massachusetts General Hospital Research Institute and Harvard Medical School, Boston, Massachusetts, USA
- Ludwig Institute for Cancer Research Lausanne Branch, Lausanne, Switzerland
| | - Alfred Zippelius
- Department of Biomedicine, University of Basel and University Hospital of Basel, Basel, Switzerland
- Department of Medical Oncology, University Hospital Basel, Basel, Switzerland
| |
Collapse
|
20
|
Said SS, Ibrahim WN. Cancer Resistance to Immunotherapy: Comprehensive Insights with Future Perspectives. Pharmaceutics 2023; 15:pharmaceutics15041143. [PMID: 37111629 PMCID: PMC10141036 DOI: 10.3390/pharmaceutics15041143] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Revised: 03/24/2023] [Accepted: 03/31/2023] [Indexed: 04/08/2023] Open
Abstract
Cancer immunotherapy is a type of treatment that harnesses the power of the immune systems of patients to target cancer cells with better precision compared to traditional chemotherapy. Several lines of treatment have been approved by the US Food and Drug Administration (FDA) and have led to remarkable success in the treatment of solid tumors, such as melanoma and small-cell lung cancer. These immunotherapies include checkpoint inhibitors, cytokines, and vaccines, while the chimeric antigen receptor (CAR) T-cell treatment has shown better responses in hematological malignancies. Despite these breakthrough achievements, the response to treatment has been variable among patients, and only a small percentage of cancer patients gained from this treatment, depending on the histological type of tumor and other host factors. Cancer cells develop mechanisms to avoid interacting with immune cells in these circumstances, which has an adverse effect on how effectively they react to therapy. These mechanisms arise either due to intrinsic factors within cancer cells or due other cells within the tumor microenvironment (TME). When this scenario is used in a therapeutic setting, the term “resistance to immunotherapy” is applied; “primary resistance” denotes a failure to respond to treatment from the start, and “secondary resistance” denotes a relapse following the initial response to immunotherapy. Here, we provide a thorough summary of the internal and external mechanisms underlying tumor resistance to immunotherapy. Furthermore, a variety of immunotherapies are briefly discussed, along with recent developments that have been employed to prevent relapses following treatment, with a focus on upcoming initiatives to improve the efficacy of immunotherapy for cancer patients.
Collapse
Affiliation(s)
- Sawsan Sudqi Said
- Department of Biomedical Sciences, College of Health Sciences, QU Health, Qatar University, Doha P.O. Box 2713, Qatar
| | - Wisam Nabeel Ibrahim
- Department of Biomedical Sciences, College of Health Sciences, QU Health, Qatar University, Doha P.O. Box 2713, Qatar
| |
Collapse
|
21
|
Khan M, Du K, Ai M, Wang B, Lin J, Ren A, Chen C, Huang Z, Qiu W, Yuan Y, Tian Y. PD-L1 expression as biomarker of efficacy of PD-1/PD-L1 checkpoint inhibitors in metastatic triple negative breast cancer: A systematic review and meta-analysis. Front Immunol 2023; 14:1060308. [PMID: 36949944 PMCID: PMC10027008 DOI: 10.3389/fimmu.2023.1060308] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 02/13/2023] [Indexed: 03/08/2023] Open
Abstract
Background Inhibitors of programmed cell death 1 (PD-1)/programmed cell death ligand 1(PD-L1) checkpoint have been approved for metastatic triple negative breast cancer (mTNBC) in patients positive for PD-L1 expression. Negative results from the recent phase III trials (IMPassion131 and IMPassion132) have raises questions on the efficacy of PD-1/PD-L1 checkpoint inhibitors and the predictive value of PD-L1 expression. Here we attempt to systematically analyze the biomarker value of PD-L1 expression for predicting the response of PD-1/PD-L1 checkpoint inhibitors in mTNBC. Materials and methods PubMed database was searched until Dec 2021 for studies evaluating PD-1/PD-L1 checkpoint inhibitors plus/minus chemotherapy in mTNBC. Outcome of interest included objective response rate (ORR), progression-free survival (PFS), and overall survival (OS). Review Manager (RevMan) version 5.4. was used for data-analysis. Results In total, 20 clinical trials comprising 3962 mTNBC patients (ICT: 2665 (67%); CT: 1297 (33%) were included in this study. Overall ORR was 22% (95%CI, 14-30%) and significant improvement was observed for PD-L1+ patients (ORR 1.78 [95%CI, 1.45-2.19], p<0.00001) as compared to PD-L1- cohort. Pooled outcome also indicated a significant 1-year PFS and 2-year OS advantage for patients with PD-L1 expression (1-year PFS: ORR 1.39 [95%CI, 1.04-1.85], p=0.02; I2 = 0%; 2-year OS: (ORR 2.47 [95%CI, 1.30-4.69], p=0.006; I2 = 63%). Subgroup analysis indicated that PD-L1 expression can successfully predict tumor response and 2-year OS benefit in mTNBC patients regardless of the type of investigating agent, line of treatment administration, and to some extent the type of treatment. Biomarker ability of PD-L1 expression to predict 1-year PFS was slightly better with pembrolizumab (p=0.09) than atezolizumab (p=0.18), and significantly better when treatment was administered in the first-line setting (OR 1.38 [95%CI, 1.02-1.87], p=0.04) and chemotherapy was added (OR 1.38 [95%CI, 1.02-1.86], p=0.03). Immune-related toxicity of any grade and grade≥3 was 39% (95%CI, 26%-52%) and 10% (95%CI, 8%-13%), respectively. Conclusions PD-L1 expression can predict objective response rate and 2-year OS in mTNBC patients receiving PD-1/PD-L1 checkpoint inhibitors. One-year PFS is also predicted in selected patients. PD-L1 expression can be a useful biomarker of efficacy of PD-1/PD-L1 checkpoint inhibitors in mTNBC.
Collapse
Affiliation(s)
- Muhammad Khan
- Department of Radiation Oncology, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, China
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Disease, Guangzhou Medical University, Guangzhou, China
| | - Kunpeng Du
- Department of Radiation Oncology, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, China
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Disease, Guangzhou Medical University, Guangzhou, China
| | - Meiling Ai
- Department of Radiation Oncology, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, China
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Disease, Guangzhou Medical University, Guangzhou, China
| | - Baiyao Wang
- Department of Radiation Oncology, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, China
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Disease, Guangzhou Medical University, Guangzhou, China
| | - Jie Lin
- Department of Radiation Oncology, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, China
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Disease, Guangzhou Medical University, Guangzhou, China
| | - Anbang Ren
- Department of Radiation Oncology, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, China
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Disease, Guangzhou Medical University, Guangzhou, China
| | - Chengcong Chen
- Department of Radiation Oncology, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, China
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Disease, Guangzhou Medical University, Guangzhou, China
| | - Zhong Huang
- Department of Radiation Oncology, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, China
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Disease, Guangzhou Medical University, Guangzhou, China
| | - Wenze Qiu
- Department of Radiation Oncology, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, China
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Disease, Guangzhou Medical University, Guangzhou, China
| | - Yawei Yuan
- Department of Radiation Oncology, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, China
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Disease, Guangzhou Medical University, Guangzhou, China
| | - Yunhong Tian
- Department of Radiation Oncology, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, China
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Disease, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
22
|
Rashid S, Shaughnessy M, Tsao H. Melanoma classification and management in the era of molecular medicine. Dermatol Clin 2022; 41:49-63. [DOI: 10.1016/j.det.2022.07.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
|
23
|
Zhang D, Lindstrom A, Kim EJ, Hwang CI, Hall ML, Lin TY, Li Y. SEMA3C Supports Pancreatic Cancer Progression by Regulating the Autophagy Process and Tumor Immune Microenvironment. Front Oncol 2022; 12:890154. [PMID: 35785187 PMCID: PMC9243227 DOI: 10.3389/fonc.2022.890154] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Accepted: 05/16/2022] [Indexed: 01/26/2023] Open
Abstract
To date, driver genes for pancreatic cancer treatment are difficult to pursue therapeutically. Targeting mutated KRAS, the most renowned driver gene in pancreatic cancer, is an active area of study. We discovered a gene named SEMA3C was highly expressed in pancreatic cancer cell lines and patients with a G12D mutation in KRAS. High expression of SEMA3C in patients was significantly associated with the decreased survival of pancreatic cancer patients based on the TCGA database. In pancreatic cancer cells, SEMA3C knockdown or inhibition exhibited growth/colony inhibition and cell cycle arrest. In addition, SEMA3C inhibition sensitized KRAS or MEK1/2 inhibition in pancreatic cancer cells. Overexpression of SEMA3C resulted in the induction of autophagy, whereas depletion of SEMA3C compromised induction of autophagy. SEMA3C modified the PD-L1 expression in tumor and immune cells and is correlated with the M2-like macrophage marker ARG1/CD163 expression, which could reshape the tumor microenvironment. Inhibition of SEMA3C decreased tumor formation in the xenograft model in vivo. Taken together, our data suggest that SEMA3C plays a substantial role in promoting cancer cell survival by regulating the autophagy process and impacting the tumor environment immune response. SEMA3C can be used as a novel target or marker with therapeutic or diagnostic potential in pancreatic cancer especially in tumors harboring the specific KRAS G12D mutation.
Collapse
Affiliation(s)
- Dalin Zhang
- Department of Biochemistry and Molecular Medicine, UC Davis Comprehensive Cancer Center, University of California, Davis, Sacramento, CA, United States
| | - Aaron Lindstrom
- Department of Biochemistry and Molecular Medicine, UC Davis Comprehensive Cancer Center, University of California, Davis, Sacramento, CA, United States
| | - Edward J Kim
- Division of Hematology and Oncology, Department of Internal Medicine, University of California, Davis, Sacramento, CA, United States
| | - Chang-il Hwang
- Department of Microbiology and Molecular Genetics, University of California, Davis, Davis, CA, United States
| | - Madison Lee Hall
- Department of Microbiology and Molecular Genetics, University of California, Davis, Davis, CA, United States
| | - Tzu-Yin Lin
- Division of Hematology and Oncology, Department of Internal Medicine, University of California, Davis, Sacramento, CA, United States
| | - Yuanpei Li
- Department of Biochemistry and Molecular Medicine, UC Davis Comprehensive Cancer Center, University of California, Davis, Sacramento, CA, United States,*Correspondence: Yuanpei Li,
| |
Collapse
|
24
|
Yamaguchi Y, Gibson J, Ou K, Lopez LS, Ng RH, Leggett N, Jonsson VD, Zarif JC, Lee PP, Wang X, Martinez C, Dorff TB, Forman SJ, Priceman SJ. PD-L1 blockade restores CAR T cell activity through IFN-γ-regulation of CD163+ M2 macrophages. J Immunother Cancer 2022; 10:e004400. [PMID: 35738799 PMCID: PMC9226933 DOI: 10.1136/jitc-2021-004400] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/08/2022] [Indexed: 11/04/2022] Open
Abstract
BACKGROUND The immune suppressive tumor microenvironment (TME) that inhibits T cell infiltration, survival, and antitumor activity has posed a major challenge for developing effective immunotherapies for solid tumors. Chimeric antigen receptor (CAR)-engineered T cell therapy has shown unprecedented clinical response in treating patients with hematological malignancies, and intense investigation is underway to achieve similar responses with solid tumors. Immunologically cold tumors, including prostate cancers, are often infiltrated with abundant tumor-associated macrophages (TAMs), and infiltration of CD163+ M2 macrophages correlates with tumor progression and poor responses to immunotherapy. However, the impact of TAMs on CAR T cell activity alone and in combination with TME immunomodulators is unclear. METHODS To model this in vitro, we utilized a novel co-culture system with tumor cells, CAR T cells, and polarized M1 or M2 macrophages from CD14+ peripheral blood mononuclear cells collected from healthy human donors. Tumor cell killing, T cell activation and proliferation, and macrophage phenotypes were evaluated by flow cytometry, cytokine production, RNA sequencing, and functional blockade of signaling pathways using antibodies and small molecule inhibitors. We also evaluated the TME in humanized mice following CAR T cell therapy for validation of our in vitro findings. RESULTS We observed inhibition of CAR T cell activity with the presence of M2 macrophages, but not M1 macrophages, coinciding with a robust induction of programmed death ligand-1 (PD-L1) in M2 macrophages. We observed similar PD-L1 expression in TAMs following CAR T cell therapy in the TME of humanized mice. PD-L1, but not programmed cell death protein-1, blockade in combination with CAR T cell therapy altered phenotypes to more M1-like subsets and led to loss of CD163+ M2 macrophages via interferon-γ signaling, resulting in improved antitumor activity of CAR T cells. CONCLUSION This study reveals an alternative mechanism by which the combination of CAR T cells and immune checkpoint blockade modulates the immune landscape of solid tumors to enhance therapeutic efficacy of CAR T cells.
Collapse
Affiliation(s)
- Yukiko Yamaguchi
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope, Duarte, California, USA
| | - Jackson Gibson
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope, Duarte, California, USA
| | - Kevin Ou
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope, Duarte, California, USA
| | - Lupita S Lopez
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope, Duarte, California, USA
| | - Rachel H Ng
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope, Duarte, California, USA
- Department of Bioengineering, University of Washington, Seattle, Washington, USA
| | - Neena Leggett
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope, Duarte, California, USA
| | - Vanessa D Jonsson
- Department of Applied Mathematics, University of California, Santa Cruz, California, USA
- Department of Biomolecular Engineering, University of California, Santa Cruz, California, USA
| | - Jelani C Zarif
- Department of Oncology, Johns Hopkins University School of Medicine and The Sidney Kimmel Comprehensive Cancer Center, Baltimore, Maryland, USA
| | - Peter P Lee
- Department of Immuno-Oncology, Beckman Research Institute of City of Hope, Duarte, California, USA
| | - Xiuli Wang
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope, Duarte, California, USA
| | - Catalina Martinez
- Department of Clinical and Translational Project Development, City of Hope, Duarte, CA, USA
| | - Tanya B Dorff
- Department of Medical Oncology and Therapeutics Research, City of Hope National Medical Center, Duarte, California, USA
| | - Stephen J Forman
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope, Duarte, California, USA
- Department of Immuno-Oncology, Beckman Research Institute of City of Hope, Duarte, California, USA
| | - Saul J Priceman
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope, Duarte, California, USA
- Department of Immuno-Oncology, Beckman Research Institute of City of Hope, Duarte, California, USA
| |
Collapse
|
25
|
Bazhin AV, von Ahn K, Fritz J, Bunge H, Maier C, Isayev O, Neff F, Siveke JT, Karakhanova S. Pivotal antitumor role of the immune checkpoint molecule B7-H1 in pancreatic cancer. Oncoimmunology 2022; 11:2043037. [PMID: 35251770 PMCID: PMC8890402 DOI: 10.1080/2162402x.2022.2043037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Immune checkpoint molecule B7-H1 plays a decisive immune regulatory role in different pathologies including cancer, and manipulation of B7-H1 expression became an attractive approach in cancer immunotherapy. Pancreatic cancer (PDAC) is characterized by pronounced immunosuppressive environment and B7-H1 expression correlates with PDAC prognosis. However, the first attempts to diminish B7-H1 expression in patients were not so successful. This points the complicity of PDAC immunosuppressive network and requires further examinations. We investigated the effect of B7-H1 deficiency in PDAC. Our results clearly show that partial or complete B7-H1 inhibition in vivo let to reduced tumor volume and improved survival of PDAC-bearing mice. This oncological benefit is due to the abrogation of immunosuppression provided by MDSC, macrophages, DC and Treg, which resulted in simultaneous restoration of anti-tumor immune response, namely improved accumulation and functionality of effector-memory CD4 and CD8 T cells. Our results underline the potential of B7-H1 molecule to control immunosuppressive network in PDAC and provide new issues for further clinical investigations.
Collapse
Affiliation(s)
- Alexandr V. Bazhin
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Katharina von Ahn
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Jasmin Fritz
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Henriette Bunge
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Caroline Maier
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Orkhan Isayev
- Department of Cytology, Embryology and Histology, Azerbaijan Medical University, Baku, Azerbaijan
| | - Florian Neff
- Division of Solid Tumor Translational Oncology, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), partner site University Hospital Essen, Heidelberg, Germany
| | - Jens T. Siveke
- Division of Solid Tumor Translational Oncology, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), partner site University Hospital Essen, Heidelberg, Germany
- Bridge Institute of Experimental Tumor Therapy, West German Cancer Center, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Svetlana Karakhanova
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| |
Collapse
|
26
|
Wenthe J, Naseri S, Hellström AC, Moreno R, Ullenhag G, Alemany R, Lövgren T, Eriksson E, Loskog A. Immune priming using DC- and T cell-targeting gene therapy sensitizes both treated and distant B16 tumors to checkpoint inhibition. Mol Ther Oncolytics 2022; 24:429-442. [PMID: 35141399 PMCID: PMC8810301 DOI: 10.1016/j.omto.2022.01.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 01/07/2022] [Indexed: 01/06/2023] Open
Abstract
Immune checkpoint inhibitors have revolutionized the treatment of metastatic melanoma, but most tumors show resistance. Resistance is connected to a non-T cell inflamed phenotype partially caused by a lack of functional dendritic cells (DCs) that are crucial for T cell priming. Herein, we investigated whether the adenoviral gene vehicle mLOAd703 carrying both DC- and T cell-activating genes can lead to inflammation in a B16-CD46 model and thereby overcome resistance to checkpoint inhibition therapy. B16-CD46 cells were injected subcutaneously in one or both flanks of immunocompetent C57BL/6J mice. mLOAd703 treatments were given intratumorally alone or in combination with intraperitoneal checkpoint inhibition therapy (anti-PD-1, anti-PD-L1, or anti-TIM-3). Tumor, lymph node, spleen, and serum samples were analyzed for the presence of immune cells and cytokines/chemokines. B16-CD46 tumors were non-inflamed and resistant to checkpoint blockade. In contrast, mLOAd703 treatment led to infiltration of the tumor by CD8+ T cells, natural killer (NK) cells, and CD103+ DCs, accompanied by a systemic increase of pro-inflammatory cytokines interferon γ (IFN-γ), tumor necrosis factor alpha (TNF-α), and interleukin-27 (IL-27). This response was even more pronounced after combining the virus with checkpoint therapy, in particular with anti-PD-L1 and anti-TIM-3, leading to further reduced tumor growth in injected lesions. Moreover, anti-PD-L1 combination also facilitated abscopal responses in non-injected lesions.
Collapse
Affiliation(s)
- Jessica Wenthe
- Uppsala University, Department of Immunology, Genetics and Pathology, Science for Life Laboratory, 751 85 Uppsala, Sweden
- Corresponding author Jessica Wenthe, MSc, Uppsala University, Department of Immunology, Genetics and Pathology, Science for Life Laboratory, 751 85 Uppsala, Sweden.
| | - Sedigheh Naseri
- Uppsala University, Department of Immunology, Genetics and Pathology, Science for Life Laboratory, 751 85 Uppsala, Sweden
| | - Ann-Charlotte Hellström
- Uppsala University, Department of Immunology, Genetics and Pathology, Science for Life Laboratory, 751 85 Uppsala, Sweden
| | - Rafael Moreno
- IDIBELL-Institute Català d'Oncologia, 08908 L'Hospitalet de Llobregat, Barcelona, Spain
| | - Gustav Ullenhag
- Uppsala University, Department of Immunology, Genetics and Pathology, Science for Life Laboratory, 751 85 Uppsala, Sweden
- Uppsala University Hospital, Department of Oncology, 751 85 Uppsala, Sweden
| | - Ramon Alemany
- IDIBELL-Institute Català d'Oncologia, 08908 L'Hospitalet de Llobregat, Barcelona, Spain
| | - Tanja Lövgren
- Uppsala University, Department of Immunology, Genetics and Pathology, Science for Life Laboratory, 751 85 Uppsala, Sweden
| | - Emma Eriksson
- Uppsala University, Department of Immunology, Genetics and Pathology, Science for Life Laboratory, 751 85 Uppsala, Sweden
- Lokon Pharma AB, 753 20 Uppsala, Sweden
| | - Angelica Loskog
- Uppsala University, Department of Immunology, Genetics and Pathology, Science for Life Laboratory, 751 85 Uppsala, Sweden
- Lokon Pharma AB, 753 20 Uppsala, Sweden
| |
Collapse
|
27
|
Kurozumi S, Kaira K, Matsumoto H, Kurosumi M, Yokobori T, Kanai Y, Sekine C, Honda C, Katayama A, Furuya M, Shiino S, Makiguchi T, Mongan NP, Rakha EA, Oyama T, Fujii T, Shirabe K, Horiguchi J. Association of L-type amino acid transporter 1 (LAT1) with the immune system and prognosis in invasive breast cancer. Sci Rep 2022; 12:2742. [PMID: 35177712 PMCID: PMC8854643 DOI: 10.1038/s41598-022-06615-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Accepted: 02/02/2022] [Indexed: 01/04/2023] Open
Abstract
L-type amino acid transporter 1 (LAT1), also referred to as SLC7A5, is believed to regulate tumor metabolism and be associated with tumor proliferation. In invasive breast cancer, we clinicopathologically investigated the utility of LAT1 expression. LAT1 expression was evaluated via immunohistochemistry analyses in 250 breast cancer patients undergoing long-term follow-up. We assessed the relationships between LAT1 expression and patient outcomes and clinicopathological factors. Breast cancer-specific survival stratified by LAT1 expression was assessed. Human epidermal growth factor receptor 2 (HER2)-positive patients with metastasis received trastuzumab therapy. The density of tumor-infiltrating lymphocytes (TILs) was evaluated according to the International Working Group guidelines. In the current study, high LAT1 expression was significantly correlated with estrogen receptor (ER) negativity, progesterone receptor negativity, high histological grade, increased TILs, and programmed death ligand 1 positivity. Among the ER-positive and HER2-negative patients, high LAT1 was an independent indicator of poor outcomes (hazard ratio (HR) = 2.97; 95% confidence interval (CI), 1.16-7.62; p = 0.023). Moreover, high LAT1 expression was an independent poor prognostic factor in luminal B-like breast cancer with aggressive features (HR = 3.39; 95% CI 1.35-8.52; p = 0.0094). In conclusion, high LAT1 expression could be used to identify a subgroup of invasive breast cancer characterized by aggressive behavior and high tumor immunoreaction. Our findings suggest that LAT1 might be a candidate therapeutic target for breast cancer patients, particularly those with luminal B-like type breast cancer.
Collapse
Affiliation(s)
- Sasagu Kurozumi
- Department of Breast Surgery, International University of Health and Welfare, 852, Hatakeda, Narita, Chiba, 286-8520, Japan. .,Department of General Surgical Science, Gunma University Graduate School of Medicine, Gunma, Japan.
| | - Kyoichi Kaira
- Department of Respiratory Medicine, Comprehensive Cancer Center, International Medical Center, Saitama Medical University, Saitama, Japan
| | | | | | - Takehiko Yokobori
- Gunma University Initiative for Advanced Research (GIAR), Maebashi, Gunma, Japan
| | - Yoshikatsu Kanai
- Division of Bio-System Pharmacology, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Chikako Sekine
- Department of Breast Surgery, International University of Health and Welfare, 852, Hatakeda, Narita, Chiba, 286-8520, Japan
| | - Chikako Honda
- Department of General Surgical Science, Gunma University Graduate School of Medicine, Gunma, Japan
| | - Ayaka Katayama
- Department of Diagnostic Pathology, Gunma University Graduate School of Medicine, Gunma, Japan
| | - Mio Furuya
- Department of Diagnostic Pathology, Gunma University Graduate School of Medicine, Gunma, Japan
| | - Sho Shiino
- Nottingham Breast Cancer Research Centre, School of Medicine, University of Nottingham, Nottingham, UK
| | - Takaya Makiguchi
- Department of Oral and Maxillofacial Surgery and Plastic Surgery, Gunma University Graduate School of Medicine, Gunma, Japan
| | - Nigel P Mongan
- Biodiscovery Institute, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham, UK
| | - Emad A Rakha
- Nottingham Breast Cancer Research Centre, School of Medicine, University of Nottingham, Nottingham, UK
| | - Tetsunari Oyama
- Department of Diagnostic Pathology, Gunma University Graduate School of Medicine, Gunma, Japan
| | - Takaaki Fujii
- Department of General Surgical Science, Gunma University Graduate School of Medicine, Gunma, Japan
| | - Ken Shirabe
- Department of General Surgical Science, Gunma University Graduate School of Medicine, Gunma, Japan
| | - Jun Horiguchi
- Department of Breast Surgery, International University of Health and Welfare, 852, Hatakeda, Narita, Chiba, 286-8520, Japan
| |
Collapse
|
28
|
Ponce LF, Leon K, Valiente PA. Unraveling a Conserved Conformation of the FG Loop upon the Binding of Natural Ligands to the Human and Murine PD1. J Phys Chem B 2022; 126:1441-1446. [PMID: 35167293 DOI: 10.1021/acs.jpcb.1c09463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The activation of T cells is normally accompanied by inhibitory mechanisms within which the PD1 receptor stands out. PD1 drives T cells to an unresponsive state called exhaustion, characterized by a markedly decreased capacity to exert effector functions upon binding the ligands PDL1 and PDL2. For this reason, PD1 has become one of the most important targets in cancer immunotherapy. Despite the numerous studies about PD1 signaling modulation, how the PD1 signaling pathway is activated upon the ligands' binding remains an open question. In this work, we used molecular dynamics simulations to assess the differences of the PD1 motion in the free state and in complex with the ligands. We found that, in both human and murine systems, the binding of PDL1 and PDL2 stabilizes the conformation of the FG loop similarly. This result, combined with the conservation of the FG loop residues across species, suggests that the conformation of the FG loop is somehow related to the signaling process. We also found a high similarity between the PD1-PDL1 structures with the variable region of an antibody structure, where the FG loop occupies a similar position to the CDR3 light chain.
Collapse
Affiliation(s)
- Luis F Ponce
- Molecular System Biology Department, Center of Molecular Immunology, Havana, Havana 11600, Cuba.,Center for Molecular Simulations, Biological Science Department, University of Calgary, Calgary, Alberta T2N 1N4, Canada
| | - Kalet Leon
- Molecular System Biology Department, Center of Molecular Immunology, Havana, Havana 11600, Cuba
| | - Pedro A Valiente
- Center for Protein Studies, Faculty of Biology, University of Havana, Havana, Havana 10400, Cuba.,Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario M5S 3E1, Canada
| |
Collapse
|
29
|
Yamaguchi H, Hsu JM, Yang WH, Hung MC. Mechanisms regulating PD-L1 expression in cancers and associated opportunities for novel small-molecule therapeutics. Nat Rev Clin Oncol 2022; 19:287-305. [DOI: 10.1038/s41571-022-00601-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/06/2022] [Indexed: 02/06/2023]
|
30
|
Liu W, Stachura P, Xu HC, Váraljai R, Shinde P, Ganesh NU, Mack M, Van Lierop A, Huang A, Sundaram B, Lang KS, Picard D, Fischer U, Remke M, Homey B, Roesch A, Häussinger D, Lang PA, Borkhardt A, Pandyra AA. BAFF Attenuates Immunosuppressive Monocytes in the Melanoma Tumor Microenvironment. Cancer Res 2022; 82:264-277. [PMID: 34810198 PMCID: PMC9397630 DOI: 10.1158/0008-5472.can-21-1171] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 08/17/2021] [Accepted: 11/15/2021] [Indexed: 01/07/2023]
Abstract
Emerging evidence indicates B-cell activating factor (BAFF, Tnfsf13b) to be an important cytokine for antitumor immunity. In this study, we generated a BAFF-overexpressing B16.F10 melanoma cell model and found that BAFF-expressing tumors grow more slowly in vivo than control tumors. The tumor microenvironment (TME) of BAFF-overexpressing tumors had decreased myeloid infiltrates with lower PD-L1 expression. Monocyte depletion and anti-PD-L1 antibody treatment confirmed the functional importance of monocytes for the phenotype of BAFF-mediated tumor growth delay. RNA sequencing analysis confirmed that monocytes isolated from BAFF-overexpressing tumors were characterized by a less exhaustive phenotype and were enriched for in genes involved in activating adaptive immune responses and NF-κB signaling. Evaluation of patients with late-stage metastatic melanoma treated with inhibitors of the PD-1/PD-L1 axis demonstrated a stratification of patients with high and low BAFF plasma levels. Patients with high BAFF levels experienced lower responses to anti-PD-1 immunotherapies. In summary, these results show that BAFF, through its effect on tumor-infiltrating monocytes, not only impacts primary tumor growth but can serve as a biomarker to predict response to anti-PD-1 immunotherapy in advanced disease. SIGNIFICANCE: The BAFF cytokine regulates monocytes in the melanoma microenvironment to suppress tumor growth, highlighting the importance of BAFF in antitumor immunity.
Collapse
Affiliation(s)
- Wei Liu
- Department of Molecular Medicine II, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | - Paweł Stachura
- Department of Molecular Medicine II, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | - Haifeng C. Xu
- Department of Molecular Medicine II, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | - Renáta Váraljai
- Department of Dermatology, University Hospital Essen, West German Cancer Center, University of Duisburg-Essen and the German Cancer Consortium (DKTK), Essen, Germany
| | - Prashant Shinde
- Department of Molecular Medicine II, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | - Nikkitha Umesh Ganesh
- Department of Molecular Medicine II, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | - Matthias Mack
- Department of Nephrology, Universitätsklinikum Regensburg, Regensburg, Germany
| | - Anke Van Lierop
- Department of Dermatology, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | - Anfei Huang
- Department of Molecular Medicine II, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | - Balamurugan Sundaram
- Department of Molecular Medicine II, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | - Karl S. Lang
- Institute of Immunology, Medical Faculty, University of Duisburg-Essen, Essen, Germany
| | - Daniel Picard
- Department of Pediatric Oncology, Hematology and Clinical Immunology, Medical Faculty, Center of Child and Adolescent Health, Heinrich-Heine-University, Düsseldorf, Germany.,Division of Pediatric Neuro-Oncogenomics, German Cancer Research Center (DKFZ), Heidelberg, Germany.,German Consortium for Translational Cancer Research (DKTK), partner site Essen/Düsseldorf, Düsseldorf, Germany.,Department of Neuropathology, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | - Ute Fischer
- Department of Pediatric Oncology, Hematology and Clinical Immunology, Medical Faculty, Center of Child and Adolescent Health, Heinrich-Heine-University, Düsseldorf, Germany
| | - Marc Remke
- Department of Pediatric Oncology, Hematology and Clinical Immunology, Medical Faculty, Center of Child and Adolescent Health, Heinrich-Heine-University, Düsseldorf, Germany.,Division of Pediatric Neuro-Oncogenomics, German Cancer Research Center (DKFZ), Heidelberg, Germany.,German Consortium for Translational Cancer Research (DKTK), partner site Essen/Düsseldorf, Düsseldorf, Germany.,Department of Neuropathology, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | - Bernhard Homey
- Department of Dermatology, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | - Alexander Roesch
- Department of Dermatology, University Hospital Essen, West German Cancer Center, University of Duisburg-Essen and the German Cancer Consortium (DKTK), Essen, Germany
| | - Dieter Häussinger
- Department of Gastroenterology, Hepatology, and Infectious Diseases, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | | | - Arndt Borkhardt
- Department of Pediatric Oncology, Hematology and Clinical Immunology, Medical Faculty, Center of Child and Adolescent Health, Heinrich-Heine-University, Düsseldorf, Germany
| | - Aleksandra A. Pandyra
- Department of Pediatric Oncology, Hematology and Clinical Immunology, Medical Faculty, Center of Child and Adolescent Health, Heinrich-Heine-University, Düsseldorf, Germany.,Corresponding Author: Aleksandra A. Pandyra, Department of Pediatric Oncology, Hematology and Clinical Immunology, Medical Faculty, Center of Child and Adolescent Health, Heinrich-Heine-University, Universitätsstraβe 1, Düsseldorf, 40225, Germany. E-mail:
| |
Collapse
|
31
|
Zhu L, Liu J, Chen J, Zhou Q. The developing landscape of combinatorial therapies of immune checkpoint blockade with DNA damage repair inhibitors for the treatment of breast and ovarian cancers. J Hematol Oncol 2021; 14:206. [PMID: 34930377 PMCID: PMC8686226 DOI: 10.1186/s13045-021-01218-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Accepted: 12/05/2021] [Indexed: 02/07/2023] Open
Abstract
The use of immune checkpoint blockade (ICB) using antibodies against programmed death receptor (PD)-1, PD ligand (PD-L)-1, and cytotoxic T-lymphocyte antigen 4 (CTLA-4) has redefined the therapeutic landscape in solid tumors, including skin, lung, bladder, liver, renal, and breast tumors. However, overall response rates to ICB therapy remain limited in PD-L1-negative patients. Thus, rational and effective combination therapies will be needed to address ICB treatment resistance in these patients, as well as in PD-L1-positive patients who have progressed under ICB treatment. DNA damage repair inhibitors (DDRis) may activate T-cell responses and trigger inflammatory cytokines release and eventually immunogenic cancer cell death by amplifying DNA damage and generating immunogenic neoantigens, especially in DDR-defective tumors. DDRi may also lead to adaptive PD-L1 upregulation, providing a rationale for PD-L1/PD-1 blockade. Thus, based on preclinical evidence of efficacy and no significant overlapping toxicity, some ICB/DDRi combinations have rapidly progressed to clinical testing in breast and ovarian cancers. Here, we summarize the available clinical data on the combination of ICB with DDRi agents for treating breast and ovarian cancers and discuss the mechanisms of action and other lessons learned from translational studies conducted to date. We also review potential biomarkers to select patients most likely to respond to ICB/DDRi combination therapy.
Collapse
Affiliation(s)
- Lingling Zhu
- Lung Cancer Center, West China Hospital of Sichuan University, Chengdu, 610041, Sichuan Province, China
| | - Jiewei Liu
- Lung Cancer Center, West China Hospital of Sichuan University, Chengdu, 610041, Sichuan Province, China
| | - Jiang Chen
- Department of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, 310016, Zhejiang Province, China.
| | - Qinghua Zhou
- Lung Cancer Center, West China Hospital of Sichuan University, Chengdu, 610041, Sichuan Province, China.
| |
Collapse
|
32
|
Wong JJW, Selbo PK. Light-controlled elimination of PD-L1+ cells. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY B-BIOLOGY 2021; 225:112355. [PMID: 34768077 DOI: 10.1016/j.jphotobiol.2021.112355] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 10/11/2021] [Accepted: 11/03/2021] [Indexed: 12/31/2022]
Abstract
The programmed death ligand-1 (PD-L1), also known as CD274 or B7-H1, is mainly expressed on cancer cells and/or immunosuppressive cells in the tumor microenvironment (TME) and plays an essential role in tumor progression and immune escape. Immune checkpoint inhibitors (ICIs) of the PD-1/PD-L1 axis have shown impressive clinical success, however, the majority of the patients do not respond to immune checkpoint therapy (ICT). Thus, to overcome ICT resistance there is a high need for potent and novel strategies that simultaneously target both tumor cells and immunosuppressive cells in the TME. In this study, we show that the intracellular light-controlled drug delivery method photochemical internalization (PCI) induce specific and strongly enhanced cytotoxic effects of the PD-L1-targeting immunotoxin, anti-PD-L1-saporin (Anti-PDL1-SAP), in the PD-L1+ triple-negative breast cancer MDA-MB-231 cell line, while no enhanced efficacy was obtained in the PD-L1 negative control cell line MDA-MB-453. Using fluorescence microscopy, we reveal that the anti-PD-L1 antibody binds to PD-L1 on the surface of the MDA-MD-231 cells and overnight accumulates in late endosomes and lysosomes where it co-localizes with the PCI photosensitizer fimaporfin (TPCS2a). Moreover, light-controlled endosomal/lysosomal escape of the anti-PD-L1 antibody and fimaporfin into the cytosol was obtained. We also confirm that the breast MDA-MB-468 and the prostate PC-3 and DU-145 cancer cell lines have subpopulations with PD-L1 expression. In addition, we show that interferon-gamma strongly induce PD-L1 expression in the per se PD-L1 negative CT26.WT cells and enhance the PD-L1 expression in MC-38 cells, of which both are murine colon cancer cell lines. In conclusion, our work provides an in vitro proof-of-concept of PCI-enhanced targeting and eradication of PD-L1 positive immunosuppressive cells. This light-controlled combinatorial strategy has a potential to advance cancer immunotherapy and should be explored in preclinical studies.
Collapse
Affiliation(s)
- Judith Jing Wen Wong
- Department of Radiation Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Norway.
| | - Pål Kristian Selbo
- Department of Radiation Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Norway.
| |
Collapse
|
33
|
Wang W, Jing H, Liu J, Bu D, Zhang Y, Zhu T, Lu K, Xu Y, Cheng M, Liu J, Yao J, Huang S, Wang L. Correlation between schistosomiasis and CD8+ T cell and stromal PD-L1 as well as the different prognostic role of CD8+ T cell and PD-L1 in schistosomal-associated colorectal cancer and non-schistosomal-associated colorectal cancer. World J Surg Oncol 2021; 19:321. [PMID: 34743724 PMCID: PMC8573878 DOI: 10.1186/s12957-021-02433-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Accepted: 10/24/2021] [Indexed: 02/08/2023] Open
Abstract
Background The effect of schistosomiasis on CD8+ T cells and then on PD-L1 expression was unknown, and the utility of CD8+ TILs as a biomarker for schistosomal-associated colorectal cancer (SCRC) rarely has been reported. Methods Three hundred thirty-eight patients with colorectal cancer (CRC) were enrolled. Immunohistochemical analysis was conducted to evaluate the expression of PD-L1 and the infiltration of CD8+ T cells. Results In the total cohort, the results showed that CD8+ TIL density was positively correlated with tumoral (p = 0.0001) and stromal PD-L1 expression (p = 0.0102). But there were no correlation between schistosomiasis and CD8+ TILs and PD-L1. Furthermore, CD8+ TIL density (p = 0.010), schistosomiasis (p = 0.042) were independent predictive factors for overall survival (OS). Stromal PD-L1 (sPD-L1) was correlated with OS (p = 0.046), but it was not an independent predictor. In patients without schistosomiasis, CD8 + T cells (p = 0.002) and sPD-L1 (p = 0.005) were associated with better OS. In patients with schistosomiasis, CD8 + T cells were independent prognosis factor (p = 0.045). Conclusions The study showed that CD8+ TILs was an independent predictive factor for OS in CRC and SCRC patients. The expression of PD-L1 was positively associated with CD8 + TILs density. There were no correlation between schistosomiasis and CD8 + TILs and PD-L1. Stromal PD-L1 but not tPD-L1 was significantly associated with OS, whereas it was not an independent prognostic factor. Supplementary Information The online version contains supplementary material available at 10.1186/s12957-021-02433-w.
Collapse
Affiliation(s)
- Weixia Wang
- Department of Pathology, Qingpu Branch of Zhongshan Hospital, Fudan University, No. 1158 East Park Road, Qingpu District, Shanghai, 201700, People's Republic of China
| | - Hongyan Jing
- Department of Pathology, Qingpu Branch of Zhongshan Hospital, Fudan University, No. 1158 East Park Road, Qingpu District, Shanghai, 201700, People's Republic of China
| | - Jican Liu
- Department of Pathology, Qingpu Branch of Zhongshan Hospital, Fudan University, No. 1158 East Park Road, Qingpu District, Shanghai, 201700, People's Republic of China
| | - Dacheng Bu
- Department of Pathology, Qingpu Branch of Zhongshan Hospital, Fudan University, No. 1158 East Park Road, Qingpu District, Shanghai, 201700, People's Republic of China
| | - Yingyi Zhang
- Department of Pathology, Qingpu Branch of Zhongshan Hospital, Fudan University, No. 1158 East Park Road, Qingpu District, Shanghai, 201700, People's Republic of China
| | - Ting Zhu
- Department of Pathology, Qingpu Branch of Zhongshan Hospital, Fudan University, No. 1158 East Park Road, Qingpu District, Shanghai, 201700, People's Republic of China
| | - Kui Lu
- Department of Pathology, Qingpu Branch of Zhongshan Hospital, Fudan University, No. 1158 East Park Road, Qingpu District, Shanghai, 201700, People's Republic of China
| | - Yanchao Xu
- Department of Pathology, Qingpu Branch of Zhongshan Hospital, Fudan University, No. 1158 East Park Road, Qingpu District, Shanghai, 201700, People's Republic of China
| | - Meihong Cheng
- Department of Pathology, Qingpu Branch of Zhongshan Hospital, Fudan University, No. 1158 East Park Road, Qingpu District, Shanghai, 201700, People's Republic of China
| | - Jing Liu
- Department of Pathology, Qingpu Branch of Zhongshan Hospital, Fudan University, No. 1158 East Park Road, Qingpu District, Shanghai, 201700, People's Republic of China
| | - Junxia Yao
- Department of Pathology, Qingpu Branch of Zhongshan Hospital, Fudan University, No. 1158 East Park Road, Qingpu District, Shanghai, 201700, People's Republic of China
| | - Sinian Huang
- Department of Pathology, Qingpu Branch of Zhongshan Hospital, Fudan University, No. 1158 East Park Road, Qingpu District, Shanghai, 201700, People's Republic of China
| | - Limei Wang
- Department of Pathology, Qingpu Branch of Zhongshan Hospital, Fudan University, No. 1158 East Park Road, Qingpu District, Shanghai, 201700, People's Republic of China.
| |
Collapse
|
34
|
Nienhuis PH, Antunes IF, Glaudemans AWJM, Jalving M, Leung D, Noordzij W, Slart RHJA, de Vries EF, Hospers GAP. 18F-BMS986192 PET imaging of PD-L1 in metastatic melanoma patients with brain metastases treated with immune checkpoint inhibitors. A pilot study. J Nucl Med 2021; 63:899-905. [PMID: 34503964 DOI: 10.2967/jnumed.121.262368] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 08/24/2021] [Indexed: 11/16/2022] Open
Abstract
Immune checkpoint inhibitors (ICI) targeting PD-1/PD-L1 frequently induces tumor response in metastatic melanoma patients. However, tumor response often takes months and may be heterogeneous. Consequently, additional local treatment for non-responsive metastases may be needed, especially in the case of brain metastases. Non-invasive imaging may allow the characterization of (brain) metastases to predict response. This pilot study uses 18F-BMS986192 PET for PD-L1 expression to explore the variability in metastatic tracer uptake and its relation to tumor response, with a special focus on brain metastases. Methods: Metastatic melanoma patients underwent whole-body 18F-BMS986192 PET/CT scanning before and 6 weeks after starting ICI therapy. 18F-BMS986192 uptake was measured in healthy tissues, organs, and tumor lesions. Tumor response was evaluated at 12 weeks using CT thorax/abdomen and MRI brain. RECIST v 1.1 was used to define therapy response per patient. Response per lesion was measured by the percentage change in lesion diameter. Toxicity was assessed according to Common Terminology Criteria for Adverse Events version 4.0. Results: Baseline 18F-BMS986192 PET/CT was performed in 8 patients, with follow-up scans in 4 patients. The highest tracer uptake was observed in the spleen, bone marrow, kidneys, and liver. Tracer uptake in tumor lesions was heterogeneous. In total, 42 tumor lesions were identified at baseline with most lesions in the lungs (n = 21) and brain (n = 14). Tracer uptake was similar between tumor locations. 18F-BMS986192 uptake in lesions at baseline, corrected for blood pool activity, was negatively correlated with the change lesion diameter at response evaluation (r=-0.49, P = 0.005), both in intra- and extracerebral lesions. Receiver operating characteristic (ROC) analysis demonstrated that 18F-BMS986192 uptake can discriminate between responding and nonresponding lesions with an area under the curve of 0.82. At the follow-up scan an increased 18F-BMS986192 uptake compared to baseline scan was correlated with an increased lesion diameter at response evaluation. In the follow-up 18F-BMS986192-PET scan of two patients, ICI-related toxicity (thyroiditis and colitis) was detected. Conclusion: In this pilot study, 18F-BMS986192 PET showed heterogeneous uptake in intra and extracerebral metastatic lesions in melanoma patients. Baseline 18F-BMS986192 uptake was able to predict an ICI treatment-induced reduction in lesion volume, whereas the follow-up PET scan allowed the detection of treatment-induced toxicity.
Collapse
|
35
|
Huang TC, Liang CW, Li YI, Guo JC, Lin CC, Chen YJ, Cheng AL, Hsu CH. Prognostic value of PD-L1 expression on immune cells or tumor cells for locally advanced esophageal squamous cell carcinoma in patients treated with neoadjuvant chemoradiotherapy. J Cancer Res Clin Oncol 2021; 148:1803-1811. [PMID: 34432128 DOI: 10.1007/s00432-021-03772-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Accepted: 08/17/2021] [Indexed: 11/25/2022]
Abstract
PURPOSE Programmed death-ligand 1 (PD-L1) expression may influence the prognosis of patients with localized esophageal cancer. The current study compared the prognostic value of PD-L1 expression between tumor cells and immune cells. METHODS Archival esophageal tumor tissue samples were collected from patients who received paclitaxel and cisplatin-based neoadjuvant chemoradiotherapy (CRT) for locally advanced esophageal squamous cell carcinoma (ESCC) in three prospective phase II trials. PD-L1 expression on tumor and immune cells was examined immunohistochemically by using the SP142 antibody and scored by two independent pathologists. The association of PD-L1 expression with patient's outcomes was analyzed using a log-rank test and Cox regression multivariate analysis. RESULTS A total of 100 patients were included. PD-L1 expression on tumor cells was positive (≥ 1%, TC-positive) in 55 patients; PD-L1 expression on immune cells was high (≥ 5%, IC-high) in 30 patients. TC-positive status was associated with poor overall survival (OS) (HR: 1.63, P = 0.035), whereas IC-high status was associated with improved OS (HR: 0.44, P = 0.0024). Multivariate analysis revealed that TC-positive, IC-high, and performance status were independent prognostic factors for progression-free survival and that IC-high and performance status were independent factors for OS. Furthermore, the combination of IC-high and TC-negative status was associated with the optimal OS, whereas that of TC-positive and IC-low status was associated with the worst OS. CONCLUSION PD-L1 expression on tumor and immune cells may have different prognostic value for patients with locally advanced ESCC receiving neoadjuvant CRT. A combination of these two indexes may further improve the prognostic prediction.
Collapse
Affiliation(s)
- Ta-Chen Huang
- Graduate Institute of Oncology, National Taiwan University College of Medicine, Taipei, Taiwan.,Department of Oncology, National Taiwan University Hospital, Taipei, Taiwan
| | - Cher-Wei Liang
- Department of Pathology, Fu-Jen Catholic University Hospital, New Taipei City, Taiwan
| | - Yu-I Li
- Department of Pathology, Fu-Jen Catholic University Hospital, New Taipei City, Taiwan
| | - Jhe-Cyuan Guo
- Department of Oncology, National Taiwan University Hospital, Taipei, Taiwan.,National Taiwan University Cancer Center, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Chia-Chi Lin
- Department of Oncology, National Taiwan University Hospital, Taipei, Taiwan
| | - Ya-Jhen Chen
- Graduate Institute of Oncology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Ann-Lii Cheng
- Graduate Institute of Oncology, National Taiwan University College of Medicine, Taipei, Taiwan. .,Department of Oncology, National Taiwan University Hospital, Taipei, Taiwan. .,Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan. .,National Taiwan University Cancer Center, National Taiwan University College of Medicine, Taipei, Taiwan.
| | - Chih-Hung Hsu
- Graduate Institute of Oncology, National Taiwan University College of Medicine, Taipei, Taiwan. .,Department of Oncology, National Taiwan University Hospital, Taipei, Taiwan. .,National Taiwan University Cancer Center, National Taiwan University College of Medicine, Taipei, Taiwan.
| |
Collapse
|
36
|
Li T, Liu T, Zhu W, Xie S, Zhao Z, Feng B, Guo H, Yang R. Targeting MDSC for Immune-Checkpoint Blockade in Cancer Immunotherapy: Current Progress and New Prospects. CLINICAL MEDICINE INSIGHTS-ONCOLOGY 2021; 15:11795549211035540. [PMID: 34408525 PMCID: PMC8365012 DOI: 10.1177/11795549211035540] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Accepted: 07/07/2021] [Indexed: 01/06/2023]
Abstract
Immune-checkpoint blockade (ICB) demonstrated inspiring effect and great promise in anti-cancer therapy. However, many obstacles, such as drug resistance and difficulty in patient selection, limited the efficacy of ICB therapy and awaited to be overcome. By timely identification and intervention of the key immune-suppressive promotors in the tumor microenvironment (TME), we may better understand the mechanisms of cancer immune-escape and use novel strategies to enhance the therapeutic effect of ICB. Myeloid-derived suppressor cell (MDSC) is recognized as a major immune suppressor in the TME. In this review, we summarized the roles MDSC played in the cancer context, focusing on its negative biologic functions in ICB therapy, discussed the strategies targeted on MDSC to optimize the diagnosis and therapy process of ICB and improve the efficacy of ICB therapy against malignancies.
Collapse
Affiliation(s)
- Tianhang Li
- Department of Urology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Institute of Urology, Nanjing University, Nanjing, 210008, People's Republic of China
| | - Tianyao Liu
- Department of Urology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Institute of Urology, Nanjing University, Nanjing, 210008, People's Republic of China
| | - Wenjie Zhu
- Department of Urology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Institute of Urology, Nanjing University, Nanjing, 210008, People's Republic of China
| | - Shangxun Xie
- Department of Urology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Institute of Urology, Nanjing University, Nanjing, 210008, People's Republic of China
| | - Zihan Zhao
- Department of Urology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Institute of Urology, Nanjing University, Nanjing, 210008, People's Republic of China
| | - Baofu Feng
- Department of Urology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Institute of Urology, Nanjing University, Nanjing, 210008, People's Republic of China
| | - Hongqian Guo
- Department of Urology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Institute of Urology, Nanjing University, Nanjing, 210008, People's Republic of China
| | - Rong Yang
- Department of Urology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Institute of Urology, Nanjing University, Nanjing, 210008, People's Republic of China
| |
Collapse
|
37
|
Baru A, Sharma S, Purakayastha BPD, Khan S, Mazumdar S, Gupta R, Kundu PK, Arora NM. AXTEX-4D: A Three-Dimensional Ex Vivo Platform for Preclinical Investigations of Immunotherapy Agents. Assay Drug Dev Technol 2021; 19:361-372. [PMID: 34319797 DOI: 10.1089/adt.2021.031] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The latest advancements in oncology are majorly focused on immuno-oncology (I-O) therapies. However, only ∼7% of drugs are being approved from the preclinical discovery phase to phase 1. The most challenging issues in I-O are the development of active and efficient drugs in an economically feasible way and in a comparatively short time for testing and validation. This mandates an urgent need for the upgradation of preclinical screening models that closely mimic the in vivo tumor microenvironment (TME). The established and most common methods for investigating the tumoricidal activity of I-O drugs are either two-dimensional systems or primary tumor cells in standard tissue culture vessels. Unfortunately, they do not mimic the TME. Consequently, the more in vivo-like three-dimensional (3D) multicellular tumor spheroids are quickly becoming the favored model to examine immune cell-mediated responses in reaction to the administration of I-O drugs. Despite many advantages of multicellular spheroids, challenges (e.g., incompatibility of quantitative assays with spheroid platforms) are still involved in the tedious procedures required for the spheroid culture that is holding back the biological community from adapting the well-recognized spheroid tissue models for studying drug delivery more widely. To this end, we have demonstrated the utility of the 3D ex vivo oncology model, developed on our novel AXTEX-4D™ platform to assess therapeutic efficacies of I-O drugs by investigating immune cell proliferation, migration, infiltration, cytokine profiling, and cytotoxicity of tumor tissueoids. The platform eliminates the need for additional biomolecules such as hydrogels and instead relies on the cancer cells themselves to create their own gradients and microenvironmental factors. In effect, the more comprehensive and ex vivo-like immune-oncology model developed on AXTEX-4D platform can be utilized for high-throughput screening of immunotherapeutic drugs.
Collapse
Affiliation(s)
- Ambica Baru
- Mammalian Cell Culture Lab, Premas Biotech Pvt, Ltd., Sector IV, IMT, Manesar, India
| | - Swati Sharma
- Mammalian Cell Culture Lab, Premas Biotech Pvt, Ltd., Sector IV, IMT, Manesar, India
| | | | - Sameena Khan
- Mammalian Cell Culture Lab, Premas Biotech Pvt, Ltd., Sector IV, IMT, Manesar, India
| | - Saumyabrata Mazumdar
- Mammalian Cell Culture Lab, Premas Biotech Pvt, Ltd., Sector IV, IMT, Manesar, India
| | - Reeshu Gupta
- Mammalian Cell Culture Lab, Premas Biotech Pvt, Ltd., Sector IV, IMT, Manesar, India
| | - Prabuddha K Kundu
- Mammalian Cell Culture Lab, Premas Biotech Pvt, Ltd., Sector IV, IMT, Manesar, India
| | - Nupur Mehrotra Arora
- Mammalian Cell Culture Lab, Premas Biotech Pvt, Ltd., Sector IV, IMT, Manesar, India
| |
Collapse
|
38
|
Von CE, Fuang HG. A case of remarkable response to atezolizumab in ALK-translocated metastatic lung adenocarcinoma. Respir Med Case Rep 2021; 34:101478. [PMID: 34336591 PMCID: PMC8318922 DOI: 10.1016/j.rmcr.2021.101478] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 07/04/2021] [Accepted: 07/11/2021] [Indexed: 11/20/2022] Open
Abstract
The development of resistance to tyrosine kinase inhibitors (TKIs) in metastatic non-small cell lung cancer (NSCLC) with oncogenic driver mutations highlights the challenge in improving the survival of these patients. The standard of care for ALK-rearranged advanced NSCLC refractory to various generations of ALK TKIs falls back to the use of chemotherapy and the prognosis remains poor. We report the case of a 41-year-old lady with an ALK-translocated metastatic lung adenocarcinoma, who demonstrated good response to an immune checkpoint inhibitor, atezolizumab in combination with bevacizumab and chemotherapy (pemetrexed and carboplatin), following disease progression on three generations of ALK TKIs. Six months into treatment, she continues to show improvement in her health-related quality of life and is tolerating treatment well. Our case suggests that this treatment regimen is a potential treatment option for TKI-refractory driver-mutated NSCLC.
Collapse
Affiliation(s)
- Cheong E Von
- Department of Clinical Oncology, First Floor Menara Timur, University of Malaya Medical Centre, Lembah Pantai, 59100, Kuala Lumpur, Malaysia
| | - Ho Gwo Fuang
- Department of Clinical Oncology, First Floor Menara Timur, University of Malaya Medical Centre, Lembah Pantai, 59100, Kuala Lumpur, Malaysia
- Faculty of Medicine, University of Malaya, Malaysia
| |
Collapse
|
39
|
PD-L1 regulation revisited: impact on immunotherapeutic strategies. Trends Mol Med 2021; 27:868-881. [PMID: 34187739 DOI: 10.1016/j.molmed.2021.06.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 05/29/2021] [Accepted: 06/07/2021] [Indexed: 02/06/2023]
Abstract
A particularly promising cancer treatment is the use of monoclonal antibodies (mAbs) against immune checkpoints (i.e., immune checkpoint inhibitors; ICIs). However, many patients experience relapse and severe adverse events. To overcome these negative issues and improve efficiency, current approaches rely on combinatorial treatments, including some modulating the expression of programmed cell death receptor 1 (PD-1)/programmed death ligand 1 (PD-L1) immune checkpoints directly. In this review, we examine the recently discovered pathways involved in PD-L1 expression and highlight the relevant druggable strategies that are being developed to both improve the response rate and avoid the onset of resistance. Altogether, these new strategies will pave the way for effective treatment combinations in future oncology clinical trials.
Collapse
|
40
|
Kim DH, Bae GE, Suh KS, Ryuman D, Song KS, Kim JS, Lee SI, Yeo MK. Clinical Significance of Tumor and Immune Cell PD-L1 Expression in Gastric Adenocarcinoma. In Vivo 2021; 34:3171-3180. [PMID: 33144421 DOI: 10.21873/invivo.12152] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Revised: 07/28/2020] [Accepted: 07/29/2020] [Indexed: 12/18/2022]
Abstract
BACKGROUND/AIM The prognostic relevance of programmed cell death ligand-1 (PD-L1) protein expression in gastric cancer (GC) remains controversial. The aims of the present study were to determine the correlations between tumor cell (TC) and immune cell (IC) PD-L1 protein levels with prognosis, and to determine the correlation between PD-L1 expression and different molecular GC subtypes. MATERIALS AND METHODS TC and IC PD-L1 protein levels were measured in 286 GC patients. The patients were classified according to the Cancer Genome Atlas and Asian Cancer Research Group guidelines using immunohistochemistry and in situ hybridization. RESULTS TC and IC PD-L1 protein levels were positively correlated with patient survival. TC PD-L1 expression was negatively correlated with tumor grade. TC and IC PD-L1 expression was associated with improved prognosis in Epstein-Barr virus negative (EBV-), microsatellite instability (MSI) rather than microsatellite stability (MSS) subgroup GC patients. CONCLUSION PD-L1 protein expression in TCs and ICs can be used as a prognostic indicator for GC patients, particularly in the EBV-, MSI, and MSS subgroups.
Collapse
Affiliation(s)
- Dong Hyun Kim
- Department of Pathology, Chungnam National University School of Medicine, Daejeon, Republic of Korea
| | - Go Eun Bae
- Department of Pathology, Chungnam National University School of Medicine, Daejeon, Republic of Korea
| | - Kwang Sun Suh
- Department of Pathology, Chungnam National University School of Medicine, Daejeon, Republic of Korea
| | - David Ryuman
- Department of Pathology, Chungnam National University School of Medicine, Daejeon, Republic of Korea
| | | | - Ju Seok Kim
- Department of Internal Medicine, Chungnam National University School of Medicine, Daejeon, Republic of Korea
| | - Sang-Il Lee
- Department of Surgery, Chungnam National University College of Medicine, Daejeon, Republic of Korea
| | - Min-Kyung Yeo
- Department of Pathology, Chungnam National University School of Medicine, Daejeon, Republic of Korea
| |
Collapse
|
41
|
Freen-van Heeren JJ. Post-transcriptional control of T-cell cytokine production: Implications for cancer therapy. Immunology 2021; 164:57-72. [PMID: 33884612 DOI: 10.1111/imm.13339] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Revised: 03/22/2021] [Accepted: 03/30/2021] [Indexed: 01/05/2023] Open
Abstract
As part of the adaptive immune system, T cells are vital for the eradication of infected and malignantly transformed cells. To perform their protective function, T cells produce effector molecules that are either directly cytotoxic, such as granzymes, perforin, interferon-γ and tumour necrosis factor α, or attract and stimulate (immune) cells, such as interleukin-2. As these molecules can also induce immunopathology, tight control of their production is required. Indeed, inflammatory cytokine production is regulated on multiple levels. Firstly, locus accessibility and transcription factor availability and activity determine the amount of mRNA produced. Secondly, post-transcriptional mechanisms, influencing mRNA splicing/codon usage, stability, decay, localization and translation rate subsequently determine the amount of protein that is produced. In the immune suppressive environments of tumours, T cells gradually lose the capacity to produce effector molecules, resulting in tumour immune escape. Recently, the role of post-transcriptional regulation in fine-tuning T-cell effector function has become more appreciated. Furthermore, several groups have shown that exhausted or dysfunctional T cells from cancer patients or murine models possess mRNA for inflammatory mediators, but fail to produce effector molecules, hinting that post-transcriptional events also play a role in hampering tumour-infiltrating lymphocyte effector function. Here, the post-transcriptional regulatory events governing T-cell cytokine production are reviewed, with a specific focus on the importance of post-transcriptional regulation in anti-tumour responses. Furthermore, potential approaches to circumvent tumour-mediated dampening of T-cell effector function through the (dis)engagement of post-transcriptional events are explored, such as CRISPR/Cas9-mediated genome editing or chimeric antigen receptors.
Collapse
|
42
|
Liu C, Seeram NP, Ma H. Small molecule inhibitors against PD-1/PD-L1 immune checkpoints and current methodologies for their development: a review. Cancer Cell Int 2021; 21:239. [PMID: 33906641 PMCID: PMC8077906 DOI: 10.1186/s12935-021-01946-4] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 04/19/2021] [Indexed: 12/12/2022] Open
Abstract
Programmed death-1/programmed death ligand-1 (PD-1/PD-L1) based immunotherapy is a revolutionary cancer therapy with great clinical success. The majority of clinically used PD-1/PD-L1 inhibitors are monoclonal antibodies but their applications are limited due to their poor oral bioavailability and immune-related adverse effects (irAEs). In contrast, several small molecule inhibitors against PD-1/PD-L1 immune checkpoints show promising blockage effects on PD-1/PD-L1 interactions without irAEs. However, proper analytical methods and bioassays are required to effectively screen small molecule derived PD-1/PD-L1 inhibitors. Herein, we summarize the biophysical and biochemical assays currently employed for the measurements of binding capacities, molecular interactions, and blocking effects of small molecule inhibitors on PD-1/PD-L1. In addition, the discovery of natural products based PD-1/PD-L1 antagonists utilizing these screening assays are reviewed. Potential pitfalls for obtaining false leading compounds as PD-1/PD-L1 inhibitors by using certain binding bioassays are also discussed in this review.
Collapse
Affiliation(s)
- Chang Liu
- Bioactive Botanical Research Laboratory, Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Avedisian Hall Lab 440, 7 Greenhouse Road, Kingston, RI, 02881, USA.
| | - Navindra P Seeram
- Bioactive Botanical Research Laboratory, Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Avedisian Hall Lab 440, 7 Greenhouse Road, Kingston, RI, 02881, USA
| | - Hang Ma
- Bioactive Botanical Research Laboratory, Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Avedisian Hall Lab 440, 7 Greenhouse Road, Kingston, RI, 02881, USA.
| |
Collapse
|
43
|
Fan C, Qu H, Wang X, Sobhani N, Wang L, Liu S, Xiong W, Zeng Z, Li Y. Cancer/testis antigens: from serology to mRNA cancer vaccine. Semin Cancer Biol 2021; 76:218-231. [PMID: 33910064 DOI: 10.1016/j.semcancer.2021.04.016] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 04/19/2021] [Accepted: 04/20/2021] [Indexed: 02/01/2023]
Abstract
Cancer/testis antigens (CTAs) are a group of tumor antigens expressed in numerous cancer tissues, as well as in the testis and placental tissues. There are over 200 CTAs supported by serology and expression data. The expression patterns of CTAs reflect the similarities between the processes of gametogenesis and tumorigenesis. It is notable that CTAs are highly expressed in three types of cancers (lung cancer, bladder cancer, and skin cancer), all of which have a metal etiology. Here, we review the expression, regulation, and function of CTAs and their translational prospects as cancer biomarkers and treatment targets. Many CTAs are highly immunogenic, tissue-specific, and frequently expressed in cancer tissues but not under physiological conditions, rendering them promising candidates for cancer detection. Some CTAs are associated with clinical outcomes, so they may serve as prognostic biomarkers. A small number of CTAs are membrane-bound, making them ideal targets for chimeric antigen receptor (CAR) T cells. Mounting evidence suggests that CTAs induce humoral or cellular immune responses, providing cancer immunotherapeutic opportunities for T-cell receptors (TCRs), CAR T cell, antibody-based therapy and peptide- or mRNA-based vaccines. Indeed, CTAs are the dominating non-mutated targets in mRNA cancer vaccine development. Clinical trials on CTA TCR and vaccines have shown effectiveness, safety, and tolerance, but these successes are limited to a small number of patients. In-depth studies on CTA expression and function are needed to improve CTA-based immunotherapy.
Collapse
Affiliation(s)
- Chunmei Fan
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medicine, Central South University, Changsha, Hunan, China; Section of Epidemiology and Population Science, Department of Medicine, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, United States
| | - Hongke Qu
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medicine, Central South University, Changsha, Hunan, China
| | - Xu Wang
- Section of Epidemiology and Population Science, Department of Medicine, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, United States
| | - Navid Sobhani
- Section of Epidemiology and Population Science, Department of Medicine, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, United States
| | - Leiming Wang
- Section of Epidemiology and Population Science, Department of Medicine, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, United States
| | - Shuanglin Liu
- Section of Epidemiology and Population Science, Department of Medicine, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, United States
| | - Wei Xiong
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medicine, Central South University, Changsha, Hunan, China
| | - Zhaoyang Zeng
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medicine, Central South University, Changsha, Hunan, China.
| | - Yong Li
- Section of Epidemiology and Population Science, Department of Medicine, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, United States.
| |
Collapse
|
44
|
Mo D, Zhu H, Wang J, Hao H, Guo Y, Wang J, Han X, Zou L, Li Z, Yao H, Zhu J, Zhou J, Peng Y, Li J, Meng K. Icaritin inhibits PD-L1 expression by Targeting Protein IκB Kinase α. Eur J Immunol 2021; 51:978-988. [PMID: 33354776 PMCID: PMC8248075 DOI: 10.1002/eji.202048905] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 10/30/2020] [Accepted: 12/15/2020] [Indexed: 12/26/2022]
Abstract
Icaritin, a small molecule currently being investigated in phase III clinical trials in China (NCT03236636 and NCT03236649) for treatment of advanced hepatocellular carcinoma (HCC), is a prenylflavonoid derivative obtained from the Epimedium genus. Previously, it was found that Icaritin decreased the expression of PD-L1, but its direct molecular targets and the underlying mechanisms have not been identified. In this study, we report the identification of IKK-α as the protein target of Icaritin by biotin-based affinity binding assay. The further mutagenesis assay has provided evidence that C46 and C178 in IKK-α were essential amino acids for Icaritin binding to IKK-α, revealing the binding sites of Icaritin to IKK-α for the first time. Functionally, Icaritin inhibited the NF-κB signalling pathway by blocking IKK complex formation, which led to decreased nuclear translocation of NF-κB p65, and subsequent downregulation of PD-L1 expression in a dose-dependent manner. More importantly, PD-L1-positive patients exhibited longer overall survival upon Icaritin therapy. Finally, Icaritin in combination with checkpoints antibodies, such as α-PD-1, has demonstrated much better efficacy than any single therapy in animal models. This is the first report that anticancer effects of Icaritin are mediated, at least in part, by impairing functions of IKK-α.
Collapse
Affiliation(s)
- Dongliang Mo
- Beijing Shenogen Pharma Group. LtdBeijingP. R. China
| | - Hai Zhu
- Beijing Shenogen Pharma Group. LtdBeijingP. R. China
| | - Jun Wang
- Beijing Shenogen Pharma Group. LtdBeijingP. R. China
| | - Haibang Hao
- Beijing Shenogen Pharma Group. LtdBeijingP. R. China
| | - Yuming Guo
- Beijing Shenogen Pharma Group. LtdBeijingP. R. China
| | - Jiaojiao Wang
- Beijing Shenogen Pharma Group. LtdBeijingP. R. China
| | - Xu Han
- Beijing Shenogen Pharma Group. LtdBeijingP. R. China
| | - Liangfeng Zou
- Beijing Shenogen Pharma Group. LtdBeijingP. R. China
| | - Zhongwan Li
- Beijing Shenogen Pharma Group. LtdBeijingP. R. China
| | - Hua Yao
- Beijing Shenogen Pharma Group. LtdBeijingP. R. China
| | - Jinsong Zhu
- 13110 NE 177th Place #100Plexera LLCWoodinvilleWAUSA
| | - Junma Zhou
- Beijing Shenogen Pharma Group. LtdBeijingP. R. China
| | - Yong Peng
- Beijing Shenogen Pharma Group. LtdBeijingP. R. China
| | - Jian Li
- Institute of ImmunologyPLAThird Military Medical University (Army Medical University)ChongqingP. R. China
| | - Kun Meng
- Beijing Shenogen Pharma Group. LtdBeijingP. R. China
| |
Collapse
|
45
|
Freen-van Heeren JJ. Using CRISPR to enhance T cell effector function for therapeutic applications. Cytokine X 2021; 3:100049. [PMID: 33604565 PMCID: PMC7885876 DOI: 10.1016/j.cytox.2020.100049] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Accepted: 12/15/2020] [Indexed: 12/12/2022] Open
Abstract
T cells are critical to fight pathogenic microbes and combat malignantly transformed cells in the fight against cancer. To exert their effector function, T cells produce effector molecules, such as the pro-inflammatory cytokines IFN-γ, TNF-α and IL-2. Tumors possess many inhibitory mechanisms that dampen T cell effector function, limiting the secretion of cytotoxic molecules. As a result, the control and elimination of tumors is impaired. Through recent advances in genomic editing, T cells can now be successfully modified via CRISPR/Cas9 technology. For instance, engaging (post-)transcriptional mechanisms to enhance T cell cytokine production, the retargeting of T cell antigen specificity or rendering T cells refractive to inhibitory receptor signaling can augment T cell effector function. Therefore, CRISPR/Cas9-mediated genome editing might provide novel strategies for cancer immunotherapy. Recently, the first-in-patient clinical trial was successfully performed with CRISPR/Cas9-modified human T cell therapy. In this review, a brief overview of currently available techniques is provided, and recent advances in T cell genomic engineering for the enhancement of T cell effector function for therapeutic purposes are discussed.
Collapse
Key Words
- AP-1, activator protein 1
- ARE, AU-rich element
- ARE-Del, deletion of the 3′UTR AREs from the Ifng/IFNG gene
- CAR T cells
- CAR, Chimeric Antigen Receptor
- CRISPR
- CRISPR, Clustered Regularly Interspaced Short Palindromic Repeat
- CRS, cytokine release syndrome
- CTLA-4, cytotoxic T-lymphocyte-associated protein 4
- Cas, CRISPR-associated
- Cas9
- Cytokines
- DGK, Diacylglycerol kinase
- DHX37, DEAH-box helicase 37
- EBV, Epstein Barr virus
- FOXP3, Forkhead box P3
- GATA, GATA binding protein
- Genome editing
- IFN, interferon
- IL, interleukin
- LAG-3, Lymphocyte Activating 3
- NF-κB, nuclear factor of activated B cells
- PD-1, Programmed cell Death 1
- PD-L1, Programmed Death Ligand 1
- PTPN2, Protein Tyrosine Phosphatase Non-Receptor 2
- Pdia3, Protein Disulfide Isomerase Family A Member 3
- RBP, RNA-binding protein
- RNP, ribonuclear protein
- T cell effector function
- T cells
- TCR, T cell receptor
- TGF, transforming growth factor
- TIL, Tumor Infiltrating Lymphocyte
- TLRs, Toll-like receptors
- TNF, tumor necrosis factor
- TRAC, TCR-α chain
- TRBC, TCR-β chain
- UTR, untranslated region
- tTCR, transgenic TCR
Collapse
|
46
|
Fabian KP, Padget MR, Fujii R, Schlom J, Hodge JW. Differential combination immunotherapy requirements for inflamed (warm) tumors versus T cell excluded (cool) tumors: engage, expand, enable, and evolve. J Immunother Cancer 2021; 9:jitc-2020-001691. [PMID: 33602696 PMCID: PMC7896589 DOI: 10.1136/jitc-2020-001691] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/06/2021] [Indexed: 02/07/2023] Open
Abstract
Background Different types of tumors have varying susceptibility to immunotherapy and hence require different treatment strategies; these cover a spectrum ranging from ‘hot’ tumors or those with high mutational burden and immune infiltrates that are more amenable to targeting to ‘cold’ tumors that are more difficult to treat due to the fewer targetable mutations and checkpoint markers. We hypothesized that an effective anti-tumor response requires multiple agents that would (1) engage the immune response and generate tumor-specific effector cells; (2) expand the number and breadth of the immune effector cells; (3) enable the anti-tumor activity of these immune cells in the tumor microenvironment; and (4) evolve the tumor response to widen immune effector repertoire. Methods A hexatherapy combination was designed and administered to MC38-CEA (warm) and 4T1 (cool) murine tumor models. The hexatherapy regimen was composed of adenovirus-based vaccine and IL-15 (interleukin-15) superagonist (N-803) to engage the immune response; anti-OX40 and anti-4-1BB to expand effector cells; anti-PD-L1 (anti-programmed death-ligand 1) to enable anti-tumor activity; and docetaxel to promote antigen spread. Primary and metastatic tumor growth inhibition were measured. The generation of anti-tumor immune effector cells was analyzed using flow cytometry, ELISpot (enzyme-linked immunospot), and RNA analysis. Results The MC38-CEA and 4T1 tumor models have differential sensitivities to the combination treatments. In the ‘warm’ MC38-CEA, combinations with two to five agents resulted in moderate therapeutic benefit while the hexatherapy regimen outperformed all these combinations. On the other hand, the hexatherapy regimen was required in order to decrease the primary and metastatic tumor burden in the ‘cool’ 4T1 model. In both models, the hexatherapy regimen promoted CD4+ and CD8+ T cell proliferation and activity. Furthermore, the hexatherapy regimen induced vaccine-specific T cells and stimulated antigen cascade. The hexatherapy regimen also limited the immunosuppressive T cell and myeloid derived suppressor cell populations, and also decreased the expression of exhaustion markers in T cells in the 4T1 model. Conclusion The hexatherapy regimen is a strategic combination of immuno-oncology agents that can engage, expand, enable, and evolve the immune response and can provide therapeutic benefits in both MC38-CEA (warm) and 4T1 (cool) tumor models.
Collapse
Affiliation(s)
- Kellsye P Fabian
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Michelle R Padget
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Rika Fujii
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Jeffrey Schlom
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - James W Hodge
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| |
Collapse
|
47
|
Wu Y, Sang M, Liu F, Zhang J, Li W, Li Z, Gu L, Zheng Y, Li J, Shan B. Epigenetic modulation combined with PD-1/PD-L1 blockade enhances immunotherapy based on MAGE-A11 antigen-specific CD8+T cells against esophageal carcinoma. Carcinogenesis 2021; 41:894-903. [PMID: 32529260 DOI: 10.1093/carcin/bgaa057] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 04/15/2020] [Accepted: 06/10/2020] [Indexed: 12/24/2022] Open
Abstract
Cancer testis antigens (CTAs) are promising targets for T cell-based immunotherapy and studies have shown that certain CT genes are epigenetically depressed in cancer cells through DNA demethylation. Melanoma-associated antigen A11 (MAGE-A11) is a CTA that is frequently expressed in esophageal cancer and is correlated with a poor esophageal cancer prognosis. Consequently, MAGE-A11 is a potential immunotherapy target. In this study, we evaluated MAGE-A11 expression in esophageal cancer cells and found that it was downregulated in several tumor cell lines, which restricted the effect of immunotherapy. Additionally, the specific recognition and lytic potential of cytotoxic T lymphocytes (CTLs) derived from the MAGE-A11 was determined. Specific CTLs could kill esophageal cancer cells expressing MAGE-A11 but rarely lysed MAGE-A11-negative tumor cells. Therefore, induction of MAGE-A11 expression is critical for CTLs recognition and lysis of esophageal cancer cells. Treatment with the DNA methyltransferase inhibitor 5-aza-2'-deoxycytidine increased MAGE-A11 expression in esophageal cancer cells and subsequently enhanced the cytotoxicity of MAGE-A11-specific CD8+T cells against cancer cell lines. Furthermore, we found that PD-L1 expression in esophageal cancer cells affected the antitumor function of CTLs. programmed death-1 (PD-1)/PD-L1 blockade could increase the specific CTL-induced lysis of HLA-A2+/MAGE-A11+ tumor cell lines treated with 5-aza-2'-deoxycytidine. These findings indicate that the treatment of tumor cells with the DNA methyltransferase inhibitor 5-aza-2'-deoxycytidine augments MAGE-A11 expression in esophageal cancer cells. The combination of epigenetic modulation by 5-aza-2'-deoxycytidine and PD-1/PD-L1 blockade may be useful for T cell-based immunotherapy against esophageal cancer.
Collapse
Affiliation(s)
- Yunyan Wu
- Department of Research Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, People's Republic of China
| | - Meixiang Sang
- Department of Research Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, People's Republic of China.,Institute of Tumor Research, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, People's Republic of China
| | - Fei Liu
- Department of Research Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, People's Republic of China
| | - Jiandong Zhang
- Department of Clinical Laboratory, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, People's Republic of China
| | - Weijing Li
- Department of Anesthesiology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, People's Republic of China
| | - Zhenhua Li
- Department of Thoracic Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, People's Republic of China
| | - Lina Gu
- Department of Research Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, People's Republic of China
| | - Yang Zheng
- Department of Research Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, People's Republic of China
| | - Juan Li
- Department of Research Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, People's Republic of China
| | - Baoen Shan
- Department of Research Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, People's Republic of China.,Institute of Tumor Research, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, People's Republic of China
| |
Collapse
|
48
|
Freen-van Heeren JJ. Toll-like receptor-2/7-mediated T cell activation: An innate potential to augment CD8 + T cell cytokine production. Scand J Immunol 2021; 93:e13019. [PMID: 33377182 DOI: 10.1111/sji.13019] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 12/10/2020] [Accepted: 12/26/2020] [Indexed: 12/11/2022]
Abstract
CD8+ T cells are critical to combat pathogens and eradicate malignantly transformed cells. To exert their effector function and kill target cells, T cells produce copious amounts of effector molecules, including the pro-inflammatory cytokines interferon γ, tumour necrosis factor α and interleukin 2. TCR triggering alone is sufficient to induce cytokine secretion by effector and memory CD8+ T cells. However, T cells can also be directly activated by pathogen-derived molecules, such as through the triggering of Toll-like receptors (TLRs). TLR-mediated pathogen sensing by T cells results in the production of only interferon γ. However, in particular when the antigen load on target cells is low, or when TCR affinity to the antigen is limited, antigen-experienced T cells can benefit from costimulatory signals. TLR stimulation can also function in a costimulatory fashion to enhance TCR triggering. Combined TCR and TLR triggering enhances the proliferation, memory formation and effector function of T cells, resulting in enhanced production of interferon γ, tumour necrosis factor α and interleukin 2. Therefore, TLR ligands or the exploitation of TLR signalling could provide novel opportunities for immunotherapy approaches. In fact, CD19 CAR T cells bearing an intracellular TLR2 costimulatory domain were recently employed to treat cancer patients in a clinical trial. Here, the current knowledge regarding TLR2/7 stimulation-induced cytokine production by T cells is reviewed. Specifically, the transcriptional and post-transcriptional pathways engaged upon TLR2/7 sensing and TLR2/7 signalling are discussed. Finally, the potential uses of TLRs to enhance the anti-tumor effector function of T cells are explored.
Collapse
|
49
|
Tarhoni I, Wakefield CJ, Kollipara R, Fidler MJ, Batus M, Bonomi P, Borgia JA. Relationship between circulating tumor-associated autoantibodies and clinical outcomes in advanced-stage NSCLC patients receiving PD-1/-L1 directed immune checkpoint inhibition. J Immunol Methods 2021; 490:112956. [PMID: 33434603 DOI: 10.1016/j.jim.2021.112956] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 12/20/2020] [Accepted: 12/31/2020] [Indexed: 12/17/2022]
Abstract
BACKGROUND Durable tumor regressions are observed in a subset of advanced-stage non-small cell lung cancer (NSCLC) patients receiving PD-1/-L1 targeted immune checkpoint inhibitors (or 'immunotherapy') alone or in combination with chemotherapy. However, the majority of advanced NSCLC patients receiving these agents do not experience long-term disease control. Existing methods to identify patients most likely to gain clinical benefit from PD-1/-L1 immunotherapy have limitations, creating a need for improved methods to guide treatment selection, particularly for those likely to benefit from single-agent immunotherapy. Here, we describe the development of a series of novel assays for tumor-associated autoantibodies as part of an exploratory study intended to determine if these biomarkers have potential prognostic value in this setting. METHOD A selection of recombinant tumor autoantigens previously characterized for their diagnostic utility were developed and preliminarily evaluated by this study. These include: Fumarate Dehydrogenase (FH), Hydroxysteroid 17-Beta Dehydrogenase 10 (HSD17B10), Inosine Monophosphate Dehydrogenase 2 (IMPDH2), New York Esophageal Squamous Cell Carcinoma-1 (NY ESO-1), Phosphoglycerate Mutase 1 (PGAM1), and Vimentin. Custom Luminex immunobead assays were developed for these targets to quantitatively assess autoantibody levels in individual patient sera. Assays were erected as indirect immunoassays on MagPlex® Microspheres using standard carbodiimide/NHS-based chemistries, utilizing a biotin-conjugated secondary (i.e. anti-human IgG) antibody and R-phycoerythrin-conjugated streptavidin reporter system. Standard curves were created for quantitative purposes using commercially-available anti-antigen antibodies and permitted analytical performance characteristics to be calculated. These assays were used to preliminarily evaluate a series of pretreatment serum samples from stage IV NSCLC patients receiving anti PD-1/-L1 therapy after failure of at least one prior line of therapy (n = 40) and their classification efficiency calculated based on 12 months overall survival (OS) threshold. RESULTS Six assays were developed that each showed dynamic ranges of four orders of magnitude and provided more than 90% classification accuracy based on the observed clinical outcome data. Inter- and intra-assay precision was assessed within these standards and overall %CVs of ≤7% and ≤ 10%, respectively, were calculated. Generally, the baseline level of autoantibodies were significantly (p < 0.05) lower in the ≥12 months survival group relative to the <12 months survival groups. Serum titers of FH, HSD170B, NY-ESO-1, and vimentin were significantly correlated with ≥12 month survival (p-value 0.0038, 0.0061, 0.0073, and 0.022, respectively). IMPDH2 and PGAM1 were found to have marginal significance (p-value 0.08 and 0.076, respectively). CONCLUSION This study demonstrates an efficient and promising means for assessing circulating autoantibody titers that could be useful in selecting advanced NSCLC patients for PD-1/-L1 directed immunotherapy. Further exploration and validation of this paradigm is warranted to further refine current treatment selection methods for this therapeutic strategy.
Collapse
Affiliation(s)
- Imad Tarhoni
- Department of Cell and Molecular Medicine, Rush University Medical Center, Chicago, IL 606012, United States of America
| | - Connor J Wakefield
- Department of Cell and Molecular Medicine, Rush University Medical Center, Chicago, IL 606012, United States of America
| | - Revathi Kollipara
- Department of Medical Oncology, Rush University Medical Center, Chicago, IL 606012, United States of America
| | - Mary Jo Fidler
- Department of Medical Oncology, Rush University Medical Center, Chicago, IL 606012, United States of America
| | - Marta Batus
- Department of Medical Oncology, Rush University Medical Center, Chicago, IL 606012, United States of America
| | - Philip Bonomi
- Department of Medical Oncology, Rush University Medical Center, Chicago, IL 606012, United States of America
| | - Jeffrey A Borgia
- Department of Cell and Molecular Medicine, Rush University Medical Center, Chicago, IL 606012, United States of America; Department of Pathology, Rush University Medical Center, Chicago, IL 606012, United States of America.
| |
Collapse
|
50
|
Saito A, Tojo M, Kumagai Y, Ohzawa H, Yamaguchi H, Miyato H, Sadatomo A, Naoi D, Ota G, Koinuma K, Horie H, Lefor AK, Sata N, Kitayama J. Flow cytometry detection of cell type-specific expression of programmed death receptor ligand-1 (PD-L1) in colorectal cancer specimens. Heliyon 2020; 7:e05880. [PMID: 33458446 PMCID: PMC7797507 DOI: 10.1016/j.heliyon.2020.e05880] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 11/28/2020] [Accepted: 12/24/2020] [Indexed: 02/06/2023] Open
Abstract
Aim PD-1/PD-L1 blockade therapy is now widely used for the treatment of advanced malignancies. Although PD-L1 is known to be expressed by various host cells as well as tumor cells, the role of PD-L1 on non-malignant cells and its clinical significance is unknown. We evaluated cell type-specific expression of PD-L1 in colorectal cancer (CRC) specimens using multicolor flow cytometry. Methods Single cell suspensions were made from 21 surgically resected CRC specimens, and immunostained with various mAbs conjugated with different fluorescent dyes. Tumor cells, stromal cells, and immune cells were identified as CD326(+), CD90(+) and CD45(+) phenotype, respectively. CD11b(+) myeloid cells, CD19(+) B cells and CD4(+) or CD8(+) T cells were also stained in different samples, and their frequencies in the total cell population and the ratio of PD-L1(+) cells to each phenotype were determined. Results PD-L1 was expressed by all the cell types. The ratio of PD-L1(+) cells to CD326(+) tumor cells was 19.1% ± 14.0%, lower than those for CD90(+) stromal cells (39.6% ± 16.0%) and CD11b(+) myeloid cells (31.9% ± 14.3%). The ratio of PD-L1(+) cells in tumor cells correlated strongly with the ratio in stromal cells, while only weakly with that in myeloid cells. Tumor cells were divided into two populations by CD326 expression levels, and the PD-L1 positive ratios were inversely correlated with the rate of CD326 highly expressing cells as well as mean fluorescein intensity of CD326 in tumor cells, while positively correlated with the frequencies of stromal cells or myeloid cells in CRC. Conclusion PD-L1 is differentially expressed on various cell types in CRC. PD-L1 on tumor cells may be upregulated together with CD326 downregulation in the process of epithelial mesenchymal transition. Quantification of cell type-specific expression of PD-L1 using multicolor flow cytometry may provide useful information for the immunotherapy of solid tumors.
Collapse
Affiliation(s)
- Akira Saito
- Department of Gastrointestinal Surgery, Jichi Medical University, Japan
| | - Mineyuki Tojo
- Department of Gastrointestinal Surgery, Jichi Medical University, Japan
| | - Yuko Kumagai
- Department of Gastrointestinal Surgery, Jichi Medical University, Japan
| | - Hideyuki Ohzawa
- Department of Clinical Oncology, Jichi Medical University, Japan
| | | | - Hideyo Miyato
- Department of Gastrointestinal Surgery, Jichi Medical University, Japan
| | - Ai Sadatomo
- Department of Gastrointestinal Surgery, Jichi Medical University, Japan
| | - Daishi Naoi
- Department of Gastrointestinal Surgery, Jichi Medical University, Japan
| | - Gaku Ota
- Department of Gastrointestinal Surgery, Jichi Medical University, Japan
| | - Koji Koinuma
- Department of Gastrointestinal Surgery, Jichi Medical University, Japan
| | - Hisanaga Horie
- Department of Gastrointestinal Surgery, Jichi Medical University, Japan
| | | | - Naohiro Sata
- Department of Gastrointestinal Surgery, Jichi Medical University, Japan
| | - Joji Kitayama
- Department of Gastrointestinal Surgery, Jichi Medical University, Japan
- Corresponding author.
| |
Collapse
|