101
|
Immunotherapeutic Approaches in Ovarian Cancer. Curr Issues Mol Biol 2023; 45:1233-1249. [PMID: 36826026 PMCID: PMC9955550 DOI: 10.3390/cimb45020081] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 01/27/2023] [Accepted: 01/31/2023] [Indexed: 02/05/2023] Open
Abstract
Ovarian cancer (OC) is gynecological cancer, and diagnosis and treatment are continuously advancing. Next-generation sequencing (NGS)-based diagnoses have emerged as novel methods for identifying molecules and pathways in cancer research. The NGS-based applications have expanded in OC research for early detection and identification of aberrant genes and dysregulation pathways, demonstrating comprehensive views of the entire transcriptome, such as fusion genes, genetic mutations, and gene expression profiling. Coinciding with advances in NGS-based diagnosis, treatment strategies for OC, such as molecular targeted therapy and immunotherapy, have also advanced. Immunotherapy is effective against many other cancers, and its efficacy against OC has also been demonstrated at the clinical phase. In this review, we describe several NGS-based applications for therapeutic targets of OC, and introduce current immunotherapeutic strategies, including vaccines, checkpoint inhibitors, and chimeric antigen receptor (CAR)-T cell transplantation, for effective diagnosis and treatment of OC.
Collapse
|
102
|
Abiko K, Hamanishi J, Matsumura N, Mandai M. Dynamic host immunity and PD-L1/PD-1 blockade efficacy: developments after "IFN-γ from lymphocytes induces PD-L1 expression and promotes progression of ovarian cancer". Br J Cancer 2023; 128:461-467. [PMID: 36068276 PMCID: PMC9938281 DOI: 10.1038/s41416-022-01960-x] [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: 07/06/2022] [Revised: 08/01/2022] [Accepted: 08/11/2022] [Indexed: 11/09/2022] Open
Abstract
In the article titled "IFN-γ from lymphocytes induces PD-L1 expression and promotes progression of ovarian cancer" in 2015, we showed that PD-L1 expression is induced by IFN-γ from lymphocytes in the tumour microenvironment. This article proposed that PD-L1 expression in cancer cells is not stable but varies among cases, or even within a case, which is influenced by the stromal infiltration of cytotoxic lymphocytes. Immune-checkpoint inhibitors, especially anti-PD-1/PD-L1 therapies, are now widely used to treat various types of cancer. Predictive biomarkers for the efficacy of immune-checkpoint inhibitors include PD-L1 expression, MSI/mismatch repair deficiency and high tumour mutation burden. However, clinical trials have proven that their use in ovarian cancer is still challenging. Reliable biomarkers and new treatment strategies may be sought by elucidating the complex immune microenvironment of ovarian cancer. Although the interaction between cytotoxic lymphocytes and PD-1/PD-L1 on tumour cells is at the centre of therapeutic targets, other immune checkpoints and various immunosuppressive cells also play important roles in ovarian cancer. Targeting these role players in combination with PD-1/PD-L1 blockade may be a promising therapeutic strategy.
Collapse
Affiliation(s)
- Kaoru Abiko
- Department of Obstetrics and Gynecology, National Hospital Organization Kyoto Medical Center, Kyoto, Japan.
| | - Junzo Hamanishi
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Noriomi Matsumura
- Department of Obstetrics and Gynecology, Kindai University Faculty of Medicine, Osaka-sayama, Osaka prefecture, Japan
| | - Masaki Mandai
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| |
Collapse
|
103
|
Predicting Prognosis and Platinum Resistance in Ovarian Cancer: Role of Immunohistochemistry Biomarkers. Int J Mol Sci 2023; 24:ijms24031973. [PMID: 36768291 PMCID: PMC9916805 DOI: 10.3390/ijms24031973] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 12/17/2022] [Accepted: 12/20/2022] [Indexed: 01/20/2023] Open
Abstract
Ovarian cancer is a lethal reproductive tumour affecting women worldwide. The advancement in presentation and occurrence of chemoresistance are the key factors for poor survival among ovarian cancer women. Surgical debulking was the mainstay of systemic treatment for ovarian cancer, which was followed by a successful start to platinum-based chemotherapy. However, most women develop platinum resistance and relapse within six months of receiving first-line treatment. Thus, there is a great need to identify biomarkers to predict platinum resistance before enrolment into chemotherapy, which would facilitate individualized targeted therapy for these subgroups of patients to ensure better survival and an improved quality of life and overall outcome. Harnessing the immune response through immunotherapy approaches has changed the treatment way for patients with cancer. The immune outline has emerged as a beneficial tool for recognizing predictive and prognostic biomarkers clinically. Studying the tumour microenvironment (TME) of ovarian cancer tissue may provide awareness of actionable targets for enhancing chemotherapy outcomes and quality of life. This review analyses the relevance of immunohistochemistry biomarkers as prognostic biomarkers in predicting chemotherapy resistance and improving the quality of life in ovarian cancer.
Collapse
|
104
|
Soldi LR, Silva VLC, Rabelo DH, Uehara IA, Silva MJB. Reactivation of natural killer cells with monoclonal antibodies in the microenvironment of malignant neoplasms. J Cancer Res Clin Oncol 2023:10.1007/s00432-023-04575-8. [PMID: 36633682 DOI: 10.1007/s00432-023-04575-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 01/05/2023] [Indexed: 01/13/2023]
Abstract
Natural killer cells are critical players in the antitumor immune response due to their ability to destroy target cells through cytotoxic activity and other means. However, this response is inhibited in the tumor microenvironment, where a crippling hypoxic environment and several inhibitory molecules bind to NK cells to trigger an anergic state. Inhibitory receptors such as PD-1, NK2GA, KIR, TIGIT, and LAG-3 have been associated with inhibition of NK cells in multiple cancer types. Binding to these receptors leads to loss of cytotoxicity, lower proliferation and metabolic rates, and even apoptosis. While these receptors are important for avoiding auto-immunity, in a pathological setting like malignant neoplasms they are disadvantageous for the individual's immune system to combat cancer cells. The use of monoclonal antibodies to block these receptors contributes to cancer therapy by preventing the inhibition of NK cells. In this review, the impact of NK cell inhibition and activation on cancer therapy was summarized and an overview of the blockade of inhibitory pathways by monoclonal antibodies was provided.
Collapse
Affiliation(s)
- Luiz Ricardo Soldi
- Institute of Biomedical Sciences, Federal University of Uberlândia, Uberlândia, MG, Brazil.,Tumor Biomarkers and Osteoimmunology Laboratory, Av. Pará - 1720 - Block 6T, Room 07 - District Umuarama, Uberlândia, MG, Brazil.,Graduate Program in Applied Immunology and Parasitology, Universidade Federal de Uberlândia, Uberlândia, Brazil
| | - Victor Luigi Costa Silva
- Institute of Biomedical Sciences, Federal University of Uberlândia, Uberlândia, MG, Brazil.,Tumor Biomarkers and Osteoimmunology Laboratory, Av. Pará - 1720 - Block 6T, Room 07 - District Umuarama, Uberlândia, MG, Brazil
| | - Diogo Henrique Rabelo
- Institute of Biomedical Sciences, Federal University of Uberlândia, Uberlândia, MG, Brazil.,Tumor Biomarkers and Osteoimmunology Laboratory, Av. Pará - 1720 - Block 6T, Room 07 - District Umuarama, Uberlândia, MG, Brazil.,Graduate Program in Applied Immunology and Parasitology, Universidade Federal de Uberlândia, Uberlândia, Brazil
| | - Isadora Akemi Uehara
- Institute of Biomedical Sciences, Federal University of Uberlândia, Uberlândia, MG, Brazil.,Tumor Biomarkers and Osteoimmunology Laboratory, Av. Pará - 1720 - Block 6T, Room 07 - District Umuarama, Uberlândia, MG, Brazil
| | - Marcelo José Barbosa Silva
- Institute of Biomedical Sciences, Federal University of Uberlândia, Uberlândia, MG, Brazil. .,Tumor Biomarkers and Osteoimmunology Laboratory, Av. Pará - 1720 - Block 6T, Room 07 - District Umuarama, Uberlândia, MG, Brazil.
| |
Collapse
|
105
|
Wu W, Wang X, Le W, Lu C, Li H, Zhu Y, Chen X, An W, Xu C, Wu Q, Wang L. Immune microenvironment infiltration landscape and immune-related subtypes in prostate cancer. Front Immunol 2023; 13:1001297. [PMID: 36700224 PMCID: PMC9868452 DOI: 10.3389/fimmu.2022.1001297] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 11/22/2022] [Indexed: 01/11/2023] Open
Abstract
Background The tumor microenvironment (TME) primarily comprises cancer cells, cancer-infiltrating immune cells, and stromal cells. The tumor cells alter the TME by secreting signaling molecules to induce immune tolerance. The immune cell infiltrating the TME influences the prognosis of patients with cancers. However, immune cell infiltration (ICI) in the TME of patients with prostate cancer (PC) has not yet been studied. Methods In this study, we used Cell-type Identification by Estimating Relative Subsets of RNA Transcripts (CIBERSORT) and Estimation of Stromal and Immune cells in Malignant Tumors using Expression data (ESTIMATE) algorithms to identify three ICI clusters based on 1,099 genes associated with ICI in the TME. The patients were classified into three distinct ICI gene clusters based on overlapping differentially expressed genes in ICI clusters. Furthermore, the ICI scores were calculated using principal component analysis. Results The results revealed that patients with high ICI scores had poor prognoses and reduced expression of immune-checkpoint genes and immune-related genes. Furthermore, the transforming growth factor-beta (TGF-β) and WNT-β signaling pathways were enriched in the high ICI score subgroup, which suggests that suppression of T cells could contribute to poor prognosis of patients with PC. A positive correlation was observed between the high-ICI-score subgroup and the high tumor mutation burden (TMB) value. Patients with low ICI scores could benefit from immunotherapy, indicating that the ICI score could be used to predict the efficacy of immunotherapeutic response. Conclusions In summary, we provide a comprehensive overview of the landscape of ICI in PC, which could aid in designing the strategies for immunotherapy for patients with PC.
Collapse
Affiliation(s)
- Wei Wu
- Department of Urology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Xin’an Wang
- Department of Urology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Wei Le
- Department of Urology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Chang Lu
- Department of Urology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Haopeng Li
- Department of Urology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yaru Zhu
- Department of Urology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Xi Chen
- Department of Urology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Wenbin An
- Organ Transplantation Clinical Medical Center of Xiamen University, Department of Organ Transplantation, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China,*Correspondence: Licheng Wang, ; Qiang Wu, ; Chengdang Xu, ; Wenbin An,
| | - Chengdang Xu
- Department of Urology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China,*Correspondence: Licheng Wang, ; Qiang Wu, ; Chengdang Xu, ; Wenbin An,
| | - Qiang Wu
- Department of Urology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China,*Correspondence: Licheng Wang, ; Qiang Wu, ; Chengdang Xu, ; Wenbin An,
| | - Licheng Wang
- Department of Urology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China,*Correspondence: Licheng Wang, ; Qiang Wu, ; Chengdang Xu, ; Wenbin An,
| |
Collapse
|
106
|
Alwosaibai K, Aalmri S, Mashhour M, Ghandorah S, Alshangiti A, Azam F, Selwi W, Gharaibeh L, Alatawi Y, Alruwaii Z, Alsaab HO. PD-L1 is highly expressed in ovarian cancer and associated with cancer stem cells populations expressing CD44 and other stem cell markers. BMC Cancer 2023; 23:13. [PMID: 36604635 PMCID: PMC9814309 DOI: 10.1186/s12885-022-10404-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 12/05/2022] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Immune checkpoint inhibitors, including PD-L1 (programmed death ligand-1) inhibitors have well documented anticancer therapeutic effect in most types of cancers but its use in the treatment of ovarian cancer is not yet proven. The aim of our study is to explore the predictive biomarkers in ovarian cancer and its association with the outcomes. We have investigated the role of PD-L1 expressions in the tumor microenvironment cells including immune cells and cancer stem cells in different types of ovarian cancer. METHODS A total of 119 surgical archived ovarian cancer samples were collected from the pathology department at King Fahad Specialist Hospital, Dammam, Saudi Arabia that included serous carcinomas, clear cell carcinomas, mucinous carcinomas, endometrioid carcinomas, and granulosa cell tumors. Immunohistochemistry (IHC) staining was performed using (i) PD-L1 antibodies to detect PD-L1 expressions; (ii) CD8 and CD4 to detect Tumor Infiltrating Lymphocytes (TILs); and (iii) CD44, LGR5, and ALDH2 to detect stem cell markers. The clinicopathological data were collected from patients' medical record to investigate the association with PD-L1, TILs, and stem cells expressions. RESULTS We report high PD-L1 expressions in 47.8% of ovarian cancer samples. PD-L1 expressions were detected in different types of epithelial ovarian cancer and were not associated with poor prognosis of ovarian cancer. However, determining the expression levels of TILs in the ovarian cancer tissues found that 81% (n = 97) of ovarian cancer samples have TILs that express both of CD8 and CD4 and significantly associated with high PD-L1 expressions. Interestingly, we have found that ovarian cancer tissues with high expressions of PD-L1 were associated with high expressions of stem cells expressing CD44 and LGR5. CONCLUSIONS PD-L1 is highly expressed in the serous type of ovarian carcinomas and the overall expression of PD-L1 is not associated with poor survival rate. Furthermore, PD-L1 expressions are strongly associated with TILs and stem cell markers in ovarian cancer. Inhibiting the PD-L1 using immune checkpoint inhibitors might downregulate stem cell population that known to be associated with cancer recurrence.
Collapse
Affiliation(s)
- Kholoud Alwosaibai
- grid.415280.a0000 0004 0402 3867Research Center, Biomedical Research Department, King Fahad Specialist Hospital, Dammam, Saudi Arabia
| | - Salmah Aalmri
- grid.415280.a0000 0004 0402 3867Research Center, Biomedical Research Department, King Fahad Specialist Hospital, Dammam, Saudi Arabia
| | - Miral Mashhour
- grid.415280.a0000 0004 0402 3867Department of Pathology and Lab Medicine, King Fahad Specialist Hospital, Dammam, Saudi Arabia
| | - Salim Ghandorah
- grid.415280.a0000 0004 0402 3867Department of Pathology and Lab Medicine, King Fahad Specialist Hospital, Dammam, Saudi Arabia
| | - Abdulraheem Alshangiti
- grid.415280.a0000 0004 0402 3867Department of Medical Oncology, King Fahad Specialist Hospital-Dammam, Dammam, Saudi Arabia
| | - Faisal Azam
- grid.415280.a0000 0004 0402 3867Department of Medical Oncology, King Fahad Specialist Hospital-Dammam, Dammam, Saudi Arabia
| | - Waleed Selwi
- grid.415280.a0000 0004 0402 3867Department of Medical Oncology, King Fahad Specialist Hospital-Dammam, Dammam, Saudi Arabia
| | - Lubna Gharaibeh
- grid.116345.40000000406441915Pharmacological and Diagnostic Research Center, Faculty of Pharmacy, Al-Ahliyya Amman University, Amman, Jordan
| | - Yasser Alatawi
- grid.440760.10000 0004 0419 5685Department of Pharmacy Practice, Faculty of Pharmacy, University of Tabuk, Tabuk, Saudi Arabia
| | - Zainab Alruwaii
- Department of Anatomic Pathology, Dammam Regional Laboratory and Blood Bank, Dammam, Saudi Arabia
| | - Hashem O. Alsaab
- grid.412895.30000 0004 0419 5255Department of Pharmaceutics and Pharmaceutical Technology, Taif University, P.O BOX 11099, Taif, Saudi Arabia
| |
Collapse
|
107
|
Activated Mast Cells Combined with NRF2 Predict Prognosis for Esophageal Cancer. JOURNAL OF ONCOLOGY 2023; 2023:4211885. [PMID: 36644231 PMCID: PMC9833916 DOI: 10.1155/2023/4211885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Revised: 12/12/2022] [Accepted: 12/20/2022] [Indexed: 01/06/2023]
Abstract
Background Esophageal cancer (EC) had the sixth-highest mortality rate of all cancers due to its poor prognosis. Immune cells and mutation genes influenced the prognosis of EC, but their combined effect on predicting EC prognosis was unknown. In this study, we comprehensively analyzed the immune cell infiltration (ICI) and mutation genes and their combined effects for predicting prognosis in EC. Methods The CIBERSORT and ESTIMATE algorithms were used to analyse the ICI scape based on the TCGA and GEO databases. EC tissues and pathologic sections from Huai'an, China, were used to verify the key immune cells and mutation genes and their interactions. Results Stromal/immune score patterns and ICI/gene had no statistical significance in overall survival (OS) (p > 0.05). The combination of ICI and tumor mutation burden (TMB) showed that the high TMB and high ICI score group had the shortest OS (p = 0.004). We recognized that the key mutation gene NRF2 was significantly different in the high/low ICI score subgroups (p = 0.002) and positivity with mast cells (MCs) (p < 0.05). Through experimental validation, we found that the MCs and activated mast cells (AC-MCs) were more infiltration in stage II/III (p = 0.032; p = 0.013) of EC patients and that NRF2 expression was upregulated in EC (p = 0.045). AC-MCs combined with NRF2 had a poor prognosis, according to survival analysis (p = 0.056) and interactive analysis (p = 0.032). Conclusions We presume that NRF2 combined with AC-MCs could be a marker to predict prognosis and could influence immunotherapy through regulating PD-L1 in the EC.
Collapse
|
108
|
Li Y, Wang H, Zhao Z, Yang Y, Meng Z, Qin L. Effects of the interactions between platelets with other cells in tumor growth and progression. Front Immunol 2023; 14:1165989. [PMID: 37153586 PMCID: PMC10158495 DOI: 10.3389/fimmu.2023.1165989] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 03/31/2023] [Indexed: 05/09/2023] Open
Abstract
It has been confirmed that platelets play a key role in tumorigenesis. Tumor-activated platelets can recruit blood cells and immune cells to migrate, establish an inflammatory tumor microenvironment at the sites of primary and metastatic tumors. On the other hand, they can also promote the differentiation of mesenchymal cells, which can accelerate the proliferation, genesis and migration of blood vessels. The role of platelets in tumors has been well studied. However, a growing number of studies suggest that interactions between platelets and immune cells (e.g., dendritic cells, natural killer cells, monocytes, and red blood cells) also play an important role in tumorigenesis and tumor development. In this review, we summarize the major cells that are closely associated with platelets and discuss the essential role of the interaction between platelets with these cells in tumorigenesis and tumor development.
Collapse
|
109
|
Pourali G, Zafari N, Velayati M, Mehrabadi S, Maftooh M, Hassanian SM, Mobarhan MG, Ferns GA, Avan A, Khazaei M. Therapeutic Potential of Targeting Transforming Growth Factor-beta (TGF-β) and Programmed Death-ligand 1 (PD-L1) in Pancreatic Cancer. Curr Drug Targets 2023; 24:1335-1345. [PMID: 38053355 DOI: 10.2174/0113894501264450231129042256] [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: 07/01/2023] [Revised: 10/11/2023] [Accepted: 10/24/2023] [Indexed: 12/07/2023]
Abstract
Pancreatic cancer (PC) is one the most lethal malignancies worldwide affecting around half a million individuals each year. The treatment of PC is relatively difficult due to the difficulty in making an early diagnosis. Transforming growth factor-beta (TGF-β) is a multifunctional factor acting as both a tumor promoter in early cancer stages and a tumor suppressor in advanced disease. Programmed death-ligand 1 (PD-L1) is a ligand of programmed death-1 (PD-1), an immune checkpoint receptor, allowing tumor cells to avoid elimination by immune cells. Recently, targeting the TGF-β signaling and PD-L1 pathways has emerged as a strategy for cancer therapy. In this review, we have summarized the current knowledge regarding these pathways and their contribution to tumor development with a focus on PC. Moreover, we have reviewed the role of TGF-β and PD-L1 blockade in the treatment of various cancer types, including PC, and discussed the clinical trials evaluating TGF-β and PD-L1 antagonists in PC patients.
Collapse
Affiliation(s)
- Ghazaleh Pourali
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Medical Doctor, Mashhad University of Medical Science, Mashhad, Iran
| | - Nima Zafari
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahla Velayati
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Shima Mehrabadi
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mina Maftooh
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Basic Sciences Research Institute, Mashhad University of Medical Science, Mashhad, Iran
| | - Seyed Mahdi Hassanian
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Basic Sciences Research Institute, Mashhad University of Medical Science, Mashhad, Iran
| | - Majid Ghayour Mobarhan
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Basic Sciences Research Institute, Mashhad University of Medical Science, Mashhad, Iran
| | - Gordon A Ferns
- Brighton & Sussex Medical School, Division of Medical Education, Falmer, Brighton, Sussex, BN1 9PH, UK
| | - Amir Avan
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Basic Sciences Research Institute, Mashhad University of Medical Science, Mashhad, Iran
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- College of Medicine, University of Warith Al-Anbiyaa, Karbala, Iraq
| | - Majid Khazaei
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Basic Sciences Research Institute, Mashhad University of Medical Science, Mashhad, Iran
| |
Collapse
|
110
|
Characterization of the Immune Infiltration Landscape and Identification of Prognostic Biomarkers for Esophageal Cancer. Mol Biotechnol 2023; 65:361-383. [PMID: 35780460 PMCID: PMC9935668 DOI: 10.1007/s12033-022-00526-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 06/15/2022] [Indexed: 10/17/2022]
Abstract
Immunotherapy is an effective treatment for esophageal cancer (ESCA) patients. However, there are no dependable markers for predicting prognosis and immunotherapy responses in ESCA. Our study aims to explore immune gene prognostic models and markers in ESCA as well as predictors for immunotherapy. The expression profiles of ESCA were obtained from The Cancer Genome Atlas (TCGA), the Gene Expression Omnibus (GEO), and International Cancer Genome Consortium (ICGC) databases. Cox regression analysis was performed to construct an immune gene prognostic model. ESCA was grouped into three immune cell infiltration (ICI) clusters by CIBERSORT algorithm. The immunotherapy response of patients in different ICI score clusters was also compared. The copy number variations, somatic mutations, and single nucleotide polymorphisms were analyzed. Enrichment analyses were also performed. An immune gene prognostic model was successfully constructed. The ICI score may be used as a predictor independent of tumor mutation burden. Enrichment analyses showed that the differentially expressed genes were mostly enriched in microvillus and the KRAS and IL6/JAK/STAT3 pathways. The top eight genes with the highest mutation frequencies in ESCA were identified and all related to the prognosis of ESCA patients. Our study established an effective immune gene prognostic model and identified markers for predicting the prognosis and immunotherapy response of ESCA patients.
Collapse
|
111
|
Loss of Major Histocompatibility Complex Class I, CD8 + Tumor-infiltrating Lymphocytes, and PD-L1 Expression in Ovarian Clear Cell Carcinoma. Am J Surg Pathol 2023; 47:124-130. [PMID: 36221308 DOI: 10.1097/pas.0000000000001975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Ovarian clear cell carcinoma (OCCC), a chemoresistant ovarian cancer, shows a modest response to anti-programmed death-1/programmed death ligand-1 (PD-1/PD-L1) therapies. The effects of anti-PD-1/PD-L1 therapies rely on cytotoxic T-cell response, which is triggered by antigen presentation mediated by major histocompatibility complex (MHC) class I. The loss of MHC class I with simultaneous PD-L1 expression has been noted in several cancer types; however, these findings and their prognostic value have rarely been evaluated in OCCC. We collected data from 76 patients with OCCC for clinicopathologic analysis. Loss of MHC class I expression was seen in 44.7% of the cases including 39.3% to 47.4% of the PD-L1 + cases and was associated with fewer CD8 + tumor-infiltrating lymphocytes (TILs). PD-L1 positivity was associated with a higher number of CD8 + TILs. Cox proportional hazard models showed that high (≥50/mm 2 ) CD8 + TILs was associated with shorter disease-specific survival (hazard ratio [HR]=3.447, 95% confidence interval [CI]: 1.222-9.720, P =0.019) and overall survival (HR=3.053, 95% CI: 1.105-8.43, P =0.031). PD-L1 positivity using Combined Positive Score was associated with shorter progression-free survival (HR=3.246, 95% CI: 1.435-7.339, P =0.005), disease-specific survival (HR=4.124, 95% CI: 1.403-12.116, P =0.010), and overall survival (HR=4.489, 95% CI: 1.553-12.972, P =0.006). Loss of MHC class I may contribute to immune evasion and resistance to anti-PD-1/PD-L1 therapies in OCCC, and CD8 + TILs and PD-L1 positivity using Combined Positive Score may have a negative prognostic value.
Collapse
|
112
|
Miyamoto K, Hayabuchi H, Tokifuji Y, Ando M, Onishi N, Okamura T, Yoshimura A, Chikuma S. A protein kinase D inhibitor suppresses AKT on T cells and antagonizes cancer immunotherapy by anti-PD-1. Int Immunol 2022; 34:609-619. [PMID: 35849090 DOI: 10.1093/intimm/dxac035] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 07/15/2022] [Indexed: 02/01/2023] Open
Abstract
Antibodies that block the interaction between PD-1 and PD-1 ligands (anti-PD-1) are in clinical use for the treatment of cancer, yet their efficacy is limited. Pre-approved therapies that enhance the effect of anti-PD-1 in combination are beneficial. Small-molecule inhibitors that attenuate T cell receptor signaling are reported to prevent T cell exhaustion and induce memory T cells with stem cell potential, resulting in a durable effector T cell response in combination with anti-PD-1. In search of such targets, we focused on protein kinase D (PKD), which is suggested to be suppressive in both tumor growth and TCR signaling. We report that CRT0066101, a PKD inhibitor (PKDi), suppressed the growth of mouse tumors at a sub-micromolar concentration in vitro. Despite its inhibitory effects on tumors, a single treatment of tumor-bearing mice with PKDi did not inhibit, but rather accelerated tumor growth, and reversed the therapeutic effect of anti-PD-1. Mice treated with PKDi showed reduced T cell infiltration and defects in the generation of effector T cells, compared to those treated with anti-PD-1, suggesting that PKDi inhibited ongoing antitumor responses. Mechanistically, PKDi inhibited phosphorylation of AKT, a primary checkpoint that is reactivated by anti-PD-1. In conclusion, PKD is fundamentally required for T cell reactivation by anti-PD-1; therefore, inhibition of PKD is not appropriate for combination therapy with anti-PD-1. On the other hand, a single dose of PKDi was shown to strongly suppress experimental autoimmunity in mice, indicating that PKDi could be useful for the treatment of immune-related adverse events that are frequently reported in anti-PD-1 therapy.
Collapse
Affiliation(s)
- Kazuhide Miyamoto
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Hodaka Hayabuchi
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Yukiko Tokifuji
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Makoto Ando
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Nobuyuki Onishi
- Department of Clinical Diagnostic Oncology, Clinical Research Institute for Clinical Pharmacology and Therapeutics, Showa University, Tokyo 157-8777, Japan
| | - Tadashi Okamura
- Department of Laboratory Animal Medicine, Research Institute, National Center for Global Health and Medicine (NCGM), Tokyo 162-8655, Japan
| | - Akihiko Yoshimura
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Shunsuke Chikuma
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo 160-8582, Japan
| |
Collapse
|
113
|
Ademuyiwa FO, Gao F, Street CR, Chen I, Northfelt DW, Wesolowski R, Arora M, Brufsky A, Dees EC, Santa-Maria CA, Connolly RM, Force J, Moreno-Aspitia A, Herndon JM, Carmody M, Davies SR, Larson S, Pfaff KL, Jones SM, Weirather JL, Giobbie-Hurder A, Rodig SJ, Liu Z, Hagemann IS, Sharon E, Gillanders WE. A randomized phase 2 study of neoadjuvant carboplatin and paclitaxel with or without atezolizumab in triple negative breast cancer (TNBC) - NCI 10013. NPJ Breast Cancer 2022; 8:134. [PMID: 36585404 PMCID: PMC9803651 DOI: 10.1038/s41523-022-00500-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 12/13/2022] [Indexed: 12/31/2022] Open
Abstract
Atezolizumab with chemotherapy has shown improved progression-free and overall survival in patients with metastatic PD-L1 positive triple negative breast cancer (TNBC). Atezolizumab with anthracycline- and taxane-based neoadjuvant chemotherapy has also shown increased pathological complete response (pCR) rates in early TNBC. This trial evaluated neoadjuvant carboplatin and paclitaxel with or without atezolizumab in patients with clinical stages II-III TNBC. The co-primary objectives were to evaluate if chemotherapy and atezolizumab increase pCR rate and tumor infiltrating lymphocyte (TIL) percentage compared to chemotherapy alone in the mITT population. Sixty-seven patients (ages 25-78 years; median, 52 years) were randomly assigned - 22 patients to Arm A, and 45 to Arm B. Median follow up was 6.6 months. In the modified intent to treat population (all patients evaluable for the primary endpoints who received at least one dose of combination therapy), the pCR rate was 18.8% (95% CI 4.0-45.6%) in Arm A, and 55.6% (95% CI 40.0-70.4%) in Arm B (estimated treatment difference: 36.8%, 95% CI 8.5-56.6%; p = 0.018). Grade 3 or higher treatment-related adverse events occurred in 62.5% of patients in Arm A, and 57.8% of patients in Arm B. One patient in Arm B died from recurrent disease during the follow-up period. TIL percentage increased slightly from baseline to cycle 1 in both Arm A (mean ± SD: 0.6% ± 21.0%) and Arm B (5.7% ± 15.8%) (p = 0.36). Patients with pCR had higher median TIL percentages (24.8%) than those with non-pCR (14.2%) (p = 0.02). Although subgroup analyses were limited by the small sample size, PD-L1-positive patients treated with chemotherapy and atezolizumab had a pCR rate of 75% (12/16). The addition of atezolizumab to neoadjuvant carboplatin and paclitaxel resulted in a statistically significant and clinically relevant increased pCR rate in patients with clinical stages II and III TNBC. (Funded by National Cancer Institute).
Collapse
Affiliation(s)
| | - Feng Gao
- Washington University School of Medicine, St Louis, MO, 63110, USA
| | | | - Ina Chen
- Washington University School of Medicine, St Louis, MO, 63110, USA
| | | | - Robert Wesolowski
- Ohio State University Comprehensive Cancer Center, Columbus, OH, 43210, USA
| | - Mili Arora
- UC Davis Comprehensive Cancer Center, Sacramento, CA, 95817, USA
| | - Adam Brufsky
- University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
| | - E Claire Dees
- University of North Carolina School of Medicine, Chapel Hill, NC, 27514, USA
| | - Cesar A Santa-Maria
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD, 21287, USA
| | | | - Jeremy Force
- Duke University School of Medicine, Durham, NC, 27710, USA
| | | | - John M Herndon
- Washington University School of Medicine, St Louis, MO, 63110, USA
| | - Madelyn Carmody
- Washington University School of Medicine, St Louis, MO, 63110, USA
| | - Sherri R Davies
- Washington University School of Medicine, St Louis, MO, 63110, USA
| | - Sarah Larson
- Washington University School of Medicine, St Louis, MO, 63110, USA
| | - Kathleen L Pfaff
- Cancer Immune Monitoring and Analysis Center, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
| | - Stephanie M Jones
- Cancer Immune Monitoring and Analysis Center, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
| | - Jason L Weirather
- Cancer Immune Monitoring and Analysis Center, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
| | - Anita Giobbie-Hurder
- Cancer Immune Monitoring and Analysis Center, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
| | - Scott J Rodig
- Cancer Immune Monitoring and Analysis Center, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
| | - Zheng Liu
- Washington University School of Medicine, St Louis, MO, 63110, USA
| | - Ian S Hagemann
- Washington University School of Medicine, St Louis, MO, 63110, USA
| | - Elad Sharon
- National Cancer Institute, Bethesda, MD, 20892, USA
| | | |
Collapse
|
114
|
Qiao J, Liu J, Jacobson JC, Clark RA, Lee S, Liu L, An Z, Zhang N, Chung DH. Anti-GRP-R monoclonal antibody antitumor therapy against neuroblastoma. PLoS One 2022; 17:e0277956. [PMID: 36525420 PMCID: PMC9757561 DOI: 10.1371/journal.pone.0277956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 11/07/2022] [Indexed: 12/23/2022] Open
Abstract
Standard treatment for patients with high-risk neuroblastoma remains multimodal therapy including chemoradiation, surgical resection, and autologous stem cell rescue. Immunotherapy has demonstrated success in treating many types of cancers; however, its use in pediatric solid tumors has been limited by low tumor mutation burdens. Gastrin-releasing peptide receptor (GRP-R) is overexpressed in numerous malignancies, including poorly-differentiated neuroblastoma. Monoclonal antibodies (mAbs) to GRP-R have yet to be developed but could serve as a potential novel immunotherapy. This preclinical study aims to evaluate the efficacy of a novel GRP-R mAb immunotherapy against neuroblastoma. We established four candidate anti-GRP-R mAbs by screening a single-chain variable fragment (scFv) library. GRP-R mAb-1 demonstrated the highest efficacy with the lowest EC50 at 4.607 ng/ml against GRP-R expressing neuroblastoma cells, blocked the GRP-ligand activation of GRP-R and its downstream PI3K/AKT signaling. This resulted in functional inhibition of cell proliferation and anchorage-independent growth, indicating that mAb-1 has an antagonist inhibitory role on GRP-R. To examine the antibody-dependent cellular cytotoxicity (ADCC) of GRP-R mAb-1 on neuroblastoma, we co-cultured neuroblastoma cells with natural killer (NK) cells versus GRP-R mAb-1 treatment alone. GRP-R mAb-1 mediated ADCC effects on neuroblastoma cells and induced release of IFNγ by NK cells under co-culture conditions in vitro. The cytotoxic effects of mAb-1 were confirmed with the secretion of cytotoxic granzyme B from NK cells and the reduction of mitotic tumor cells in vivo using a murine tumor xenograft model. In summary, GRP-R mAb-1 demonstrated efficacious anti-tumor effects on neuroblastoma cells in preclinical models. Importantly, GRP-R mAb-1 may be an efficacious, novel immunotherapy in the treatment of high-risk neuroblastoma patients.
Collapse
Affiliation(s)
- Jingbo Qiao
- Department of Surgery, UT Southwestern Medical Center, Dallas, Texas, United States of America
| | - Junquan Liu
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, United States of America
| | - Jillian C. Jacobson
- Department of Surgery, UT Southwestern Medical Center, Dallas, Texas, United States of America
| | - Rachael A. Clark
- Department of Surgery, UT Southwestern Medical Center, Dallas, Texas, United States of America
| | - Sora Lee
- Department of Surgery, UT Southwestern Medical Center, Dallas, Texas, United States of America
| | - Li Liu
- Department of Radiology, UT Southwestern Medical Center, Dallas, Texas, United States of America
| | - Zhiqiang An
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, United States of America
| | - Ningyan Zhang
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, United States of America
| | - Dai H. Chung
- Department of Surgery, UT Southwestern Medical Center, Dallas, Texas, United States of America
- Department of Surgery, Children’s Health, Dallas, Texas, United States of America
| |
Collapse
|
115
|
Cioffi R, Galli F, Rabaiotti E, Candiani M, Pella F, Candotti G, Bocciolone L, De Marzi P, Mangili G, Bergamini A. Experimental drugs for fallopian cancer: promising agents in the clinical trials and key stumbling blocks for researchers. Expert Opin Investig Drugs 2022; 31:1339-1357. [PMID: 36537209 DOI: 10.1080/13543784.2022.2160313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
INTRODUCTION Fallopian tube carcinoma (FC) as a single entity is a rare disease. Although its diagnosis is increasing thanks to the widespread use of prophylactic salpingectomy, there are no clinical trials exclusively designed for FC. AREAS COVERED This review aims at identifying the most promising trials and future therapeutic pathways in the setting of FC. EXPERT OPINION Hot topics in FC treatment include the consequences of using PARP inhibitors (PARPi) as first-line therapy, ways to overcome platinum resistance, and the role of immunotherapy. Patient selection is a key point for future development of target therapies. Next-generation sequencing (NGS) is one of the most investigated technologies both for drug discovery and identification of reverse mutations, involved in resistance to PARPi and platinum. New, promising molecular targets are emerging. Notwithstanding the disappointing outcomes when used by itself, immunotherapy in FC treatment could still have a role in combination with other agents, exploiting synergistic effects at the molecular level. The development of cancer vaccines is currently hampered by the high variability of tumor neoantigens in FC. Genomic profiling could be a solution, allowing the synthesis of individualized vaccines.
Collapse
Affiliation(s)
- Raffaella Cioffi
- Obstetrics and Gynecology Unit, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy
| | - Federica Galli
- Obstetrics and Gynecology Unit, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy
| | - Emanuela Rabaiotti
- Obstetrics and Gynecology Unit, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy
| | - Massimo Candiani
- Obstetrics and Gynecology Unit, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy
| | - Francesca Pella
- Obstetrics and Gynecology Unit, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy
| | - Giorgio Candotti
- Obstetrics and Gynecology Unit, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy
| | - Luca Bocciolone
- Obstetrics and Gynecology Unit, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy
| | - Patrizia De Marzi
- Obstetrics and Gynecology Unit, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy
| | - Giorgia Mangili
- Obstetrics and Gynecology Unit, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy
| | - Alice Bergamini
- Obstetrics and Gynecology Unit, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy
| |
Collapse
|
116
|
The cross-talk of cancer-associated fibroblasts assist in prognosis and immunotherapy in patients with breast carcinoma. Cancer Gene Ther 2022; 29:2001-2012. [PMID: 35948763 DOI: 10.1038/s41417-022-00514-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Revised: 06/28/2022] [Accepted: 07/21/2022] [Indexed: 01/25/2023]
Abstract
The association between cancer-associated fibroblasts (CAFs) and tumor microenvironment (TME) is a key factor in promoting tumor progression. However, the correlation between CAFs and TME in breast carcinoma has not been elucidated. Thus, further study about the cross-effect between CAFs and TME can provide novel strategies for breast carcinoma treatment, particularly targeted immunotherapy. First, we systematically analyzed cell communication in a single-cell dataset and identified the interacted genes between CAFs and TME components. Then, a robust fibroblast-related score (FRS) model was developed using the LASSO algorithm. The FRS can be a reliable adverse prognostic factor in three cohorts with breast carcinoma. Functional enrichment analysis and single-sample Gene Set Enrichment Analysis showed that patients with a high FRS had cold tumors with active proliferation and immunosuppression. Patients with a low FRS presented with hot tumors with active immune and cell-killing functions. Genomic variation analysis revealed that patients with a low FRS had a higher somatic mutation load and copy number variation burden. Finally, patients with a low FRS were more sensitive to chemotherapy and immunotherapy, particularly anti-PD-1 therapy. In conclusion, a reliable FRS model was constructed not only reliable for predicting prognosis but also competent to estimate clinical immunotherapy and chemotherapy response for patients with BRCA, which might provide significant clinical implications for guiding clinical decision-making for patients with BRCA.
Collapse
|
117
|
Maiorano BA, Maiorano MFP, Ciardiello D, Maglione A, Orditura M, Lorusso D, Maiello E. Beyond Platinum, ICIs in Metastatic Cervical Cancer: A Systematic Review. Cancers (Basel) 2022; 14:cancers14235955. [PMID: 36497437 PMCID: PMC9737392 DOI: 10.3390/cancers14235955] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 11/27/2022] [Accepted: 11/28/2022] [Indexed: 12/03/2022] Open
Abstract
BACKGROUND Cervical cancer (CC) constitutes the fourth most common tumor among the female population. Therapeutic approaches to advanced CC are limited, with dismal results in terms of survival, mainly after progression to platinum-based regimens. Immune checkpoint inhibitors (ICIs) are remodeling the therapeutic scenario of many solid tumors. The role of ICIs in CC should be addressed. Therefore, we systematically reviewed the latest clinical trials employing ICIs in advanced CC to assess which ICIs have been employed and how ICIs might meet the need for new therapeutic options in terms of efficacy and safety. METHODS The review was conducted following the PRISMA guidelines. The following efficacy outcomes were specifically collected: overall response rate (ORR), disease control rate (DCR), progression-free survival (PFS), and overall survival (OS); for safety: type, number, and grade of adverse events (AEs). RESULTS A total of 17 studies were analyzed. Anti-PD1 (pembrolizumab, nivolumab, cemiplimab, balstilimab, and tislelizumab), anti-PD-L1 (atezolizumab), and anti-CTLA-4 (ipilimumab, zalifrelimab) agents were employed both as single agents or combinations. Overall ORR ranged from 0% to 65.9%. ORR ranged from 5.9% to 69.6% in PD-L1-positive patients and from 0% to 50% in PD-L1-negative patients. DCR was 30.6-94.1%. mPFS ranged from 2 to 10.4 months. mOS ranged from 8 months to not reached. PD-L1 status did not impact survival. A total of 33.9% to 100% of patients experienced AEs. CONCLUSION Immunotherapy represents an appealing strategy for patients with advanced CC, as 2 out of 3 patients seem to respond to ICIs. PD-L1 status might be an indicator of response without impacting survival.
Collapse
Affiliation(s)
- Brigida Anna Maiorano
- Oncology Unit, Fondazione Casa Sollievo della Sofferenza IRCCS, 71013 San Giovanni Rotondo, Italy
- Department of Translational Medicine and Surgery, Catholic University of the Sacred Heart, 00168 Rome, Italy
| | - Mauro Francesco Pio Maiorano
- Obstetrics and Gynecology Unit, Department of Biomedical Sciences and Human Oncology, University of Bari “Aldo Moro”, 70121 Bari, Italy
- Correspondence:
| | - Davide Ciardiello
- Oncology Unit, Fondazione Casa Sollievo della Sofferenza IRCCS, 71013 San Giovanni Rotondo, Italy
- Medical Oncology, Department of Precision Medicine, Luigi Vanvitelli University of Campania, 80131 Naples, Italy
| | - Annamaria Maglione
- Obstetrics and Gynecology Department, Fondazione Casa Sollievo della Sofferenza IRCCS, 71013 San Giovanni Rotondo, Italy
| | - Michele Orditura
- Medical Oncology, Department of Precision Medicine, Luigi Vanvitelli University of Campania, 80131 Naples, Italy
| | - Domenica Lorusso
- Department of Women and Child Health, Division of Gynaecologic Oncology, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy
- Scientific Directorate, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy
| | - Evaristo Maiello
- Oncology Unit, Fondazione Casa Sollievo della Sofferenza IRCCS, 71013 San Giovanni Rotondo, Italy
| |
Collapse
|
118
|
Shi W, Fijardo M, Bruce JP, Su J, Xu W, Bell R, Bissey PA, Hui ABY, Waldron J, Pugh TJ, Yip KW, Liu FF. CD8+ Tumor-Infiltrating Lymphocyte Abundance Is a Positive Prognostic Indicator in Nasopharyngeal Cancer. Clin Cancer Res 2022; 28:5202-5210. [PMID: 36129469 DOI: 10.1158/1078-0432.ccr-22-0979] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 06/17/2022] [Accepted: 09/14/2022] [Indexed: 01/24/2023]
Abstract
PURPOSE Tumor-infiltrating lymphocytes (TIL) are immune cell populations found within tumors, critical in the antigen-specific host immune response. In this study, we aimed to elucidate the prognostic significance of CD3+, CD4+, and CD8+ TILs in nasopharyngeal cancer (NPC). EXPERIMENTAL DESIGN Immune cell infiltration was quantified in NPC samples (n = 50) using RNA-sequencing (RNA-seq) data based on rearranged T-cell receptor (TCR) reads and the Estimation of Stromal and Immune cells in malignant tumors using expression data (ESTIMATE) immune score tool. The differential abundances of TIL subset populations were also characterized through IHC staining of formalin-fixed, paraffin-embedded samples from a training cohort (n = 35), which was a subset of the RNA-seq cohort (n = 50). RESULTS In the RNA-seq cohort, patients with higher rearranged TCR reads experienced superior 5- and 10-year overall survival (OS; P < 0.001), and disease-free survival (DFS; P < 0.001). Similarly, patients with higher ESTIMATE immune scores experienced superior 5- and 10-year OS (P = 0.024) and DFS (P = 0.007). In the training cohort, high abundances of CD8+ TILs were significantly associated with improved 5- and 10-year OS (P = 0.003) and DFS (P = 0.005). These findings were corroborated in an independent validation cohort (n = 84), and combined analysis of the training and validation cohorts [n = 119 (35+84)], which further demonstrated improved 5- and 10-year survival in terms of locoregional control (P < 0.001) and distant metastasis (P = 0.03). CONCLUSIONS Taken together, our study highlights the prognostic value of CD8+ TILs in NPC, and the potential of future investigations into cellular-based immunotherapies employing CD8+ lymphocytes.
Collapse
Affiliation(s)
- Wei Shi
- Research Institute, Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | - Mackenzie Fijardo
- Research Institute, Princess Margaret Cancer Centre, Toronto, Ontario, Canada.,Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario, Canada
| | - Jeff P Bruce
- Research Institute, Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | - Jie Su
- Department of Biostatistics, Princess Margaret Cancer Centre/University Health Network, Toronto, Ontario, Canada
| | - Wei Xu
- Department of Biostatistics, Princess Margaret Cancer Centre/University Health Network, Toronto, Ontario, Canada.,Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada
| | - Rachel Bell
- Research Institute, Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | | | | | - John Waldron
- Radiation Medicine Program, Princess Margaret Cancer Centre, Toronto, Ontario, Canada.,Department of Radiation Oncology, University of Toronto, Toronto, Ontario, Canada.,Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| | - Trevor J Pugh
- Research Institute, Princess Margaret Cancer Centre, Toronto, Ontario, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada.,Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | - Kenneth W Yip
- Research Institute, Princess Margaret Cancer Centre, Toronto, Ontario, Canada.,Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario, Canada
| | - Fei-Fei Liu
- Research Institute, Princess Margaret Cancer Centre, Toronto, Ontario, Canada.,Radiation Medicine Program, Princess Margaret Cancer Centre, Toronto, Ontario, Canada.,Department of Radiation Oncology, University of Toronto, Toronto, Ontario, Canada.,Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
119
|
Jin H, Chen Y, Ren J, Huang J, Zhao Y, Liu H. TERC suppresses PD-L1 expression by downregulating RNA binding protein HuR. SCIENCE CHINA. LIFE SCIENCES 2022; 65:2505-2516. [PMID: 35661964 DOI: 10.1007/s11427-021-2085-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 03/02/2022] [Indexed: 06/15/2023]
Abstract
TERC is the RNA component of telomerase, and provides a template for TERT to synthesize telomere repeats at chromosome ends. Increasing evidence has revealed that TERC is involved in other biological processes beyond telomerase. Here, we found that the expression level of TERC is negatively correlated with PD-L1 and that ectopic expression of TERC but not TERT in ALT cells significantly inhibits PD-L1, suggesting that TERC suppresses PD-L1 expression in a telomerase-independent manner. Mechanistically, instead of regulating PD-L1 mRNA directly, TERC accelerates PD-L1 mRNA degradation by inhibiting the expression of HuR, which binds to the 3'UTR of PD-L1 mRNA and maintains its stability. We also found that the small molecule AS1842856, a FoxO1 inhibitor, promotes TERC expression and reverses the PD-L1 upregulation caused by chemotherapy, providing a potential combination cancer therapy that avoids cancer immune escape during chemotherapy.
Collapse
Affiliation(s)
- Heping Jin
- MOE Key Laboratory of Gene Function and Regulation, School of Life Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Yanlian Chen
- MOE Key Laboratory of Gene Function and Regulation, School of Life Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Jian Ren
- MOE Key Laboratory of Gene Function and Regulation, School of Life Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Junjiu Huang
- MOE Key Laboratory of Gene Function and Regulation, School of Life Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Yong Zhao
- MOE Key Laboratory of Gene Function and Regulation, School of Life Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Haiying Liu
- MOE Key Laboratory of Gene Function and Regulation, School of Life Sciences, Sun Yat-sen University, Guangzhou, 510006, China.
| |
Collapse
|
120
|
Gertych A, Walts AE, Cheng K, Liu M, John J, Lester J, Karlan BY, Orsulic S. Dynamic Changes in the Extracellular Matrix in Primary, Metastatic, and Recurrent Ovarian Cancers. Cells 2022; 11:3769. [PMID: 36497028 PMCID: PMC9736731 DOI: 10.3390/cells11233769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 11/21/2022] [Accepted: 11/24/2022] [Indexed: 11/29/2022] Open
Abstract
Cancer-associated fibroblasts (CAFs) and their extracellular matrix are active participants in cancer progression. While it is known that functionally different subpopulations of CAFs co-exist in ovarian cancer, it is unclear whether certain CAF subsets are enriched during metastatic progression and/or chemotherapy. Using computational image analyses of patient-matched primary high-grade serous ovarian carcinomas, synchronous pre-chemotherapy metastases, and metachronous post-chemotherapy metastases from 42 patients, we documented the dynamic spatiotemporal changes in the extracellular matrix, fibroblasts, epithelial cells, immune cells, and CAF subsets expressing different extracellular matrix components. Among the different CAF subsets, COL11A1+ CAFs were associated with linearized collagen fibers and exhibited the greatest enrichment in pre- and post-chemotherapy metastases compared to matched primary tumors. Although pre- and post-chemotherapy metastases were associated with increased CD8+ T cell infiltration, the infiltrate was not always evenly distributed between the stroma and cancer cells, leading to an increased frequency of the immune-excluded phenotype where the majority of CD8+ T cells are present in the tumor stroma but absent from the tumor parenchyma. Overall, most of the differences in the tumor microenvironment were observed between primary tumors and metastases, while fewer differences were observed between pre- and post-treatment metastases. These data suggest that the tumor microenvironment is largely determined by the primary vs. metastatic location of the tumor while chemotherapy does not have a significant impact on the host microenvironment.
Collapse
Affiliation(s)
- Arkadiusz Gertych
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Faculty of Biomedical Engineering, Silesian University of Technology, 44-100 Zabrze, Poland
| | - Ann E. Walts
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Keyi Cheng
- Department of Mathematics, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Manyun Liu
- Jiann-Ping Hsu College of Public Health, Georgia Southern University, Statesboro, GA 30458, USA
| | - Joshi John
- Department of Veterans Affairs, Greater Los Angeles Healthcare System, Los Angeles, CA 90095, USA
| | - Jenny Lester
- Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Beth Y. Karlan
- Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
- Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Sandra Orsulic
- Department of Veterans Affairs, Greater Los Angeles Healthcare System, Los Angeles, CA 90095, USA
- Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
- Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, CA 90095, USA
| |
Collapse
|
121
|
Mechanisms of Resistance and Strategies to Combat Resistance in PD-(L)1 Blockade. IMMUNO 2022. [DOI: 10.3390/immuno2040041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Prolonged survival and durable responses in several late-stage cancers such as melanoma and lung cancer have been made possible with the use of immune checkpoint inhibitors targeting the programmed cell-death protein 1 (PD-1) or its ligand PD-L1. While it is prudent to focus on the unprecedented and durable clinical responses, there are subsets of cancer patients that do not respond to immunotherapies or respond early and then relapse later. Many pathways of resistance have been characterized, and more continue to be uncovered. To overcome the development of resistance, an in-depth investigation is necessary to identify alternative immune receptors and signals with the overarching goal of expanding treatment options for those with demonstrated resistance to PD1 checkpoint immunotherapy. In this mini-review, we will discuss the mechanisms by which tumors exhibit resistance to anti-PD-1/PD-L1 immunotherapy and explore strategies to overcome such resistances.
Collapse
|
122
|
Suarez Mora A, Strange M, Fang Y, Uygun I, Zhang L, Tseng GC, Kalinski P, Edwards RP, Vlad AM. Longitudinal Modulation of Loco-Regional Immunity in Ovarian Cancer Patients Receiving Intraperitoneal Chemotherapy. Cancers (Basel) 2022; 14:cancers14225647. [PMID: 36428740 PMCID: PMC9688312 DOI: 10.3390/cancers14225647] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 11/15/2022] [Accepted: 11/16/2022] [Indexed: 11/19/2022] Open
Abstract
The immune tumor microenvironment (TME) of epithelial ovarian cancer (EOC) carries both effector and suppressive functions. To define immune correlates of chemotherapy-induced tumor involution, we performed longitudinal evaluation of biomarker expression on serial biological specimens collected during intraperitoneal (IP) platinum-based chemotherapy. Serial biological samples were collected at several time points during IP chemotherapy. RNA from IP fluid cells and tumor tissue was analyzed via NanoString. Meso Scale Discovery (MSD) multiplex assay and ELISA for MUC1 antibodies were performed on plasma and IP fluid. Differentially expressed genes in IP fluid demonstrate an upregulation of B cell function and activation of Th2 immune response along with dampening of Th1 immunity during chemotherapy. MSD analysis of IP fluid and gene expression analysis of tumor tissue revealed activation of Th2 immunity and the complement system. Anti-MUC1 antibodies were detected in IP fluid samples. IP fluid analysis in a secondary cohort also identified chemotherapy-induced B cell function genes. This study shows that serial IP fluid sampling is an effective method to capture changes in the immune TME during chemotherapy and reveals treatment induced changes in B cell function and Th2 immunity.
Collapse
Affiliation(s)
- Adria Suarez Mora
- Department of Obstetrics and Gynecology and Reproductive Sciences, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Magee-Womens Research Institute, Pittsburgh, PA 15213, USA
- Magee-Womens Hospital of UPMC, Pittsburgh, PA 15213, USA
| | - Mary Strange
- Magee-Womens Research Institute, Pittsburgh, PA 15213, USA
| | - Yusi Fang
- Department of Biostatistics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Ibrahim Uygun
- Department of Obstetrics and Gynecology and Reproductive Sciences, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Magee-Womens Research Institute, Pittsburgh, PA 15213, USA
| | - Lixin Zhang
- Department of Obstetrics and Gynecology and Reproductive Sciences, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Magee-Womens Research Institute, Pittsburgh, PA 15213, USA
| | - George C. Tseng
- Department of Biostatistics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Pawel Kalinski
- Roswell Park Comprehensive Cancer Center, Buffalo, NY 14203, USA
| | - Robert P. Edwards
- Department of Obstetrics and Gynecology and Reproductive Sciences, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Magee-Womens Research Institute, Pittsburgh, PA 15213, USA
- Magee-Womens Hospital of UPMC, Pittsburgh, PA 15213, USA
- Correspondence: (R.P.E.); (A.M.V.)
| | - Anda M. Vlad
- Department of Obstetrics and Gynecology and Reproductive Sciences, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Magee-Womens Research Institute, Pittsburgh, PA 15213, USA
- Correspondence: (R.P.E.); (A.M.V.)
| |
Collapse
|
123
|
Sun X, Liu L, Wan T, Huang Q, Chen J, Luo R, Liu J. The prognostic impact of the immune microenvironment in small-cell neuroendocrine carcinoma of the uterine cervix: PD-L1 and immune cell subtypes. Cancer Cell Int 2022; 22:348. [PMCID: PMC9664608 DOI: 10.1186/s12935-022-02716-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2022] [Accepted: 09/09/2022] [Indexed: 11/16/2022] Open
Abstract
Abstract
Background
We investigate the correlation between programmed cell death-ligand 1 (PD-L1) and tumor-associated immune cell (TAIC) density in small-cell neuroendocrine carcinoma of the uterine cervix (SCNEC) and their correlation with clinicopathologic features.
Methods
PD-L1 and mismatch repair protein (MMR) expression in cancer cells and the density of TAIC were evaluated by immunohistochemistry in 89 SCNEC patients. The combined positive score (CPS), tumor proportion score (TPS), and immune cell score (ICS) of PD-L1 were measured, along with their correlation with clinicopathologic features in SCNEC patients using statistical analyses.
Results
CPS of PD-L1 ≥ 1 was seen in 68.5% of patients, positive TPS and ICS of PD-L1 were detected in 59.6% and 33.7% of patients, respectively. PD-L1CPS was higher in tumor-infiltrating immune cells (r = 0.387, p = 0.001) and positively correlated with programmed cell death-1 and forkhead box P3 + regulatory T cell (FOXP3 + Treg) infiltration (r = 0.443, p < 0.001; r = 0.532, p < 0.001). There was no statistical correlation between PD-L1 and MMR status. PD-L1CPS and PD-L1ICS positivity were independent prognostic factors, correlating with a favorable survival (HR (95%CI) = 0.363(0.139–0.950), p = 0.039 and HR (95% CI) = 0.199(0.050–0.802), p = 0.023, respectively). PD-L1ICS positivity was an independent indicator of recurrence in SCNEC patients and associated with better disease-free survival (HR (95% CI) = 0.124(0.036–0425), p = 0.001). TAIC and MMR levels had no statistical impact on survival results.
Conclusions
PD-L1 positivity was seen in over half of SCNEC tumors. It may work synergistically with FOXP3 + Treg and other infiltrating immune cells to support an adaptive immune response. PD-L1 positivity may be a favorable prognostic factor in SCNEC.
Collapse
|
124
|
Chen C, Tang D, Gu C, Wang B, Yao Y, Wang R, Zhang H, Gao W. Characterization of the Immune Microenvironmental Landscape of Lung Squamous Cell Carcinoma with Immune Cell Infiltration. DISEASE MARKERS 2022; 2022:2361507. [PMID: 36411824 PMCID: PMC9674995 DOI: 10.1155/2022/2361507] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 09/13/2022] [Indexed: 08/22/2023]
Abstract
BACKGROUND Increasing evidence supports that immune cell infiltration (ICI) patterns play a key role in the tumor progression of lung squamous cell carcinoma (LUSC). However, to date, the immune infiltration picture of LUSC has not been elucidated. METHOD TCGA was used to download multiomics data from LUSC samples. At the same time, we included two datasets on lung squamous cell carcinoma, GSE17710 and GSE157010. To reveal the landscape of tumor immune microenvironment (TIME), the ESTIMATE algorithm, ssGSEA approach, and CIBERSORT analysis are used. To quantify the ICI pattern in a single tumor, consistent clustering is used to determine the LUSC subtype based on the ICI pattern, and principal component analysis (PCA) is used to obtain the ICI score. The prognostic value of the Kaplan-Meier curves is confirmed. GSEA (Gene Set Enrichment Analysis) was used to perform functional annotation. To investigate the immunotherapeutic effects of the ICI score, the immunophenotyping score (IPS) is used. Finally, analyze the mutation data with the "maftools" R package. RESULTS We identified four different immune infiltration patterns with different prognosis and biological characteristics in 792 LUSC samples. The identification of ICI patterns in individual tumors developed under ICI-related characteristic genes based on the ICI score helps to analyze the biological process, clinical results, immune cell infiltration, immunotherapy effects, and genetic variation. Immune failure is indicated by a high ICI score subtype marked by immunosuppression. Patients with low ICI scores have an abundance of efficient immune cells, which corresponds to the immunological activation phenotype and may have therapeutic benefits. The immunophenotypic score was used as a surrogate indicator of immunotherapy results, and samples with low ICI scores obtained significantly higher immunophenotypic scores. Finally, the relationship between the ICI score and tumor mutation burden (TMB) was proven. CONCLUSION This study fully clarified the indispensable role of the ICI model in the complexity and diversity of TIME. The quantitative identification of ICI patterns in a single tumor will help draw the picture of TIME and further optimize precision immunotherapy.
Collapse
Affiliation(s)
- Chunji Chen
- Shanghai Key Laboratory of Clinical Geriatric Medicine, Huadong Hospital Affiliated to Fudan University, No. 221 West Yanan Road, Shanghai, China
- Department of Thoracic Surgery, Huadong Hospital Affiliated to Fudan University, No. 221 West Yanan Road, Shanghai 200040, China
- Department of Thoracic Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University, No. 241 West Huaihai Road, Shanghai 200030, China
| | - Dongfang Tang
- Shanghai Key Laboratory of Clinical Geriatric Medicine, Huadong Hospital Affiliated to Fudan University, No. 221 West Yanan Road, Shanghai, China
- Department of Thoracic Surgery, Huadong Hospital Affiliated to Fudan University, No. 221 West Yanan Road, Shanghai 200040, China
| | - Chang Gu
- Department of Cardiothoracic Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai 200092, China
| | - Bin Wang
- Shanghai Key Laboratory of Clinical Geriatric Medicine, Huadong Hospital Affiliated to Fudan University, No. 221 West Yanan Road, Shanghai, China
- Department of Thoracic Surgery, Huadong Hospital Affiliated to Fudan University, No. 221 West Yanan Road, Shanghai 200040, China
| | - Yuanshan Yao
- Shanghai Key Laboratory of Clinical Geriatric Medicine, Huadong Hospital Affiliated to Fudan University, No. 221 West Yanan Road, Shanghai, China
- Department of Thoracic Surgery, Huadong Hospital Affiliated to Fudan University, No. 221 West Yanan Road, Shanghai 200040, China
| | - Rui Wang
- Department of Thoracic Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University, No. 241 West Huaihai Road, Shanghai 200030, China
| | - Huibiao Zhang
- Shanghai Key Laboratory of Clinical Geriatric Medicine, Huadong Hospital Affiliated to Fudan University, No. 221 West Yanan Road, Shanghai, China
- Department of Thoracic Surgery, Huadong Hospital Affiliated to Fudan University, No. 221 West Yanan Road, Shanghai 200040, China
| | - Wen Gao
- Shanghai Key Laboratory of Clinical Geriatric Medicine, Huadong Hospital Affiliated to Fudan University, No. 221 West Yanan Road, Shanghai, China
- Department of Thoracic Surgery, Huadong Hospital Affiliated to Fudan University, No. 221 West Yanan Road, Shanghai 200040, China
| |
Collapse
|
125
|
Rajtak A, Ostrowska-Leśko M, Żak K, Tarkowski R, Kotarski J, Okła K. Integration of local and systemic immunity in ovarian cancer: Implications for immunotherapy. Front Immunol 2022; 13:1018256. [PMID: 36439144 PMCID: PMC9684707 DOI: 10.3389/fimmu.2022.1018256] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Accepted: 10/18/2022] [Indexed: 08/21/2023] Open
Abstract
Cancer is a disease that induces many local and systemic changes in immunity. The difficult nature of ovarian cancer stems from the lack of characteristic symptoms that contributes to a delayed diagnosis and treatment. Despite the enormous progress in immunotherapy, its efficacy remains limited. The heterogeneity of tumors, lack of diagnostic biomarkers, and complex immune landscape are the main challenges in the treatment of ovarian cancer. Integrative approaches that combine the tumor microenvironment - local immunity - together with periphery - systemic immunity - are urgently needed to improve the understanding of the disease and the efficacy of treatment. In fact, multiparametric analyses are poised to improve our understanding of ovarian tumor immunology. We outline an integrative approach including local and systemic immunity in ovarian cancer. Understanding the nature of both localized and systemic immune responses will be crucial to boosting the efficacy of immunotherapies in ovarian cancer patients.
Collapse
Affiliation(s)
- Alicja Rajtak
- 1st Chair and Department of Oncological Gynecology and Gynecology, Medical University of Lublin, Lublin, Poland
| | - Marta Ostrowska-Leśko
- 1st Chair and Department of Oncological Gynecology and Gynecology, Medical University of Lublin, Lublin, Poland
- Chair and Department of Toxicology, Medical University of Lublin, Lublin, Poland
| | - Klaudia Żak
- 1st Chair and Department of Oncological Gynaecology and Gynaecology, Student Scientific Association, Medical University of Lublin, Lublin, Poland
| | - Rafał Tarkowski
- 1st Chair and Department of Oncological Gynecology and Gynecology, Medical University of Lublin, Lublin, Poland
| | - Jan Kotarski
- 1st Chair and Department of Oncological Gynecology and Gynecology, Medical University of Lublin, Lublin, Poland
| | - Karolina Okła
- 1st Chair and Department of Oncological Gynecology and Gynecology, Medical University of Lublin, Lublin, Poland
- Department of Surgery, University of Michigan Rogel Cancer Center, Ann Arbor, MI, United States
| |
Collapse
|
126
|
Dang Q, Sun Z, Wang Y, Wang L, Liu Z, Han X. Ferroptosis: a double-edged sword mediating immune tolerance of cancer. Cell Death Dis 2022; 13:925. [PMID: 36335094 PMCID: PMC9637147 DOI: 10.1038/s41419-022-05384-6] [Citation(s) in RCA: 60] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 10/24/2022] [Accepted: 10/26/2022] [Indexed: 11/08/2022]
Abstract
The term ferroptosis was put forward in 2012 and has been researched exponentially over the past few years. Ferroptosis is an unconventional pattern of iron-dependent programmed cell death, which belongs to a type of necrosis and is distinguished from apoptosis and autophagy. Actuated by iron-dependent phospholipid peroxidation, ferroptosis is modulated by various cellular metabolic and signaling pathways, including amino acid, lipid, iron, and mitochondrial metabolism. Notably, ferroptosis is associated with numerous diseases and plays a double-edged sword role. Particularly, metastasis-prone or highly-mutated tumor cells are sensitive to ferroptosis. Hence, inducing or prohibiting ferroptosis in tumor cells has vastly promising potential in treating drug-resistant cancers. Immunotolerant cancer cells are not sensitive to the traditional cell death pathway such as apoptosis and necroptosis, while ferroptosis plays a crucial role in mediating tumor and immune cells to antagonize immune tolerance, which has broad prospects in the clinical setting. Herein, we summarized the mechanisms and delineated the regulatory network of ferroptosis, emphasized its dual role in mediating immune tolerance, proposed its significant clinical benefits in the tumor immune microenvironment, and ultimately presented some provocative doubts. This review aims to provide practical guidelines and research directions for the clinical practice of ferroptosis in treating immune-resistant tumors.
Collapse
Affiliation(s)
- Qin Dang
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Ziqi Sun
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Yang Wang
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Libo Wang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Zaoqu Liu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China.
| | - Xinwei Han
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China.
| |
Collapse
|
127
|
Intratumoral PD-1 +CD8 + T cells associate poor clinical outcomes and adjuvant chemotherapeutic benefit in gastric cancer. Br J Cancer 2022; 127:1709-1717. [PMID: 36002752 PMCID: PMC9596411 DOI: 10.1038/s41416-022-01939-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 07/16/2022] [Accepted: 07/28/2022] [Indexed: 02/01/2023] Open
Abstract
BACKGROUND Although PD-1 has been reported to be a marker of T-cell exhaustion in several malignancies, the biological role of PD-1+CD8+ T cells in gastric cancer (GC) remains unclear. Herein, we aimed to investigate the role of PD-1+CD8+ T cells in the tumour microenvironment and its clinical significance in GC. DESIGNS This study included 441 tumour microarray specimens and 60 Flow cytometry specimens of GC patients from Zhongshan Hospital, and 250 GC patients from the Asian Cancer Research Group. RESULTS Here, we demonstrated that PD-1+CD8+ T cells functioned as an independent adverse prognosticator in GC. In addition, an abundance of intratumoral PD-1+CD8+ T cells indicated worse chemotherapeutic responsiveness to fluorouracil in Stage III GC patients. Mechanistically, PD-1+CD8+ T cell high infiltration indicated an exhausted phenotype of global CD8+ T cells in GC tissues, which was characterised by elevated immune checkpoint expression including CTLA-4 and TIM-3, whereas decreased expression of perforin. Furthermore, PD-1+CD8+ T cell high-infiltration patients with Stage III GC held elevated activity of several therapeutic signal pathways. CONCLUSIONS Our study highlighted that PD-1+CD8+ T cell abundance predicts inferior prognosis in GC, and may serve as a novel predictive biomarker to guide therapeutic option.
Collapse
|
128
|
Hudry D, Le Guellec S, Meignan S, Bécourt S, Pasquesoone C, El Hajj H, Martínez-Gómez C, Leblanc É, Narducci F, Ladoire S. Tumor-Infiltrating Lymphocytes (TILs) in Epithelial Ovarian Cancer: Heterogeneity, Prognostic Impact, and Relationship with Immune Checkpoints. Cancers (Basel) 2022; 14:5332. [PMID: 36358750 PMCID: PMC9656626 DOI: 10.3390/cancers14215332] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 10/24/2022] [Accepted: 10/26/2022] [Indexed: 08/13/2023] Open
Abstract
Epithelial ovarian cancers (EOC) are often diagnosed at an advanced stage with carcinomatosis and a poor prognosis. First-line treatment is based on a chemotherapy regimen combining a platinum-based drug and a taxane-based drug along with surgery. More than half of the patients will have concern about a recurrence. To improve the outcomes, new therapeutics are needed, and diverse strategies, such as immunotherapy, are currently being tested in EOC. To better understand the global immune contexture in EOC, several studies have been performed to decipher the landscape of tumor-infiltrating lymphocytes (TILs). CD8+ TILs are usually considered effective antitumor immune effectors that immune checkpoint inhibitors can potentially activate to reject tumor cells. To synthesize the knowledge of TILs in EOC, we conducted a review of studies published in MEDLINE or EMBASE in the last 10 years according to the PRISMA guidelines. The description and role of TILs in EOC prognosis are reviewed from the published data. The links between TILs, DNA repair deficiency, and ICs have been studied. Finally, this review describes the role of TILs in future immunotherapy for EOC.
Collapse
Affiliation(s)
- Delphine Hudry
- Inserm, U1192–Protéomique Réponse Inflammatoire Spectrométrie de Masse–PRISM, Lille University, F-59000 Lille, France
- Department of Gynecologic Oncology, Oscar Lambret Center, F-59000 Lille, France
| | - Solenn Le Guellec
- Department of Gynecologic Oncology, Oscar Lambret Center, F-59000 Lille, France
| | - Samuel Meignan
- Tumorigenesis and Resistance to Treatment Unit, Centre Oscar Lambret, F-59000 Lille, France
- CNRS, Inserm, CHU Lille, UMR9020-U1277-CANTHER-Cancer Heterogeneity Plasticity and Resistance to Therapies, Lille University, F-59000 Lille, France
| | - Stéphanie Bécourt
- Department of Gynecologic Oncology, Oscar Lambret Center, F-59000 Lille, France
| | - Camille Pasquesoone
- Department of Gynecologic Oncology, Oscar Lambret Center, F-59000 Lille, France
| | - Houssein El Hajj
- Department of Gynecologic Oncology, Oscar Lambret Center, F-59000 Lille, France
| | | | - Éric Leblanc
- Inserm, U1192–Protéomique Réponse Inflammatoire Spectrométrie de Masse–PRISM, Lille University, F-59000 Lille, France
- Department of Gynecologic Oncology, Oscar Lambret Center, F-59000 Lille, France
| | - Fabrice Narducci
- Inserm, U1192–Protéomique Réponse Inflammatoire Spectrométrie de Masse–PRISM, Lille University, F-59000 Lille, France
- Department of Gynecologic Oncology, Oscar Lambret Center, F-59000 Lille, France
| | - Sylvain Ladoire
- Department of Medical Oncology, Centre Georges-François Leclerc, F-21000 Dijon, France
- INSERM, CRI-866 Faculty of Medicine, F-21000 Dijon, France
| |
Collapse
|
129
|
Ruiz M, Zhang N, Sood AK, An Z. Antibody therapeutics for epithelial ovarian cancer. Expert Opin Biol Ther 2022; 22:1379-1391. [PMID: 36302510 PMCID: PMC10375545 DOI: 10.1080/14712598.2022.2141565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION High-grade serous ovarian carcinoma (HGSC) is an aggressive subtype of epithelial ovarian carcinoma (EOC) and remains the most lethal gynecologic cancer. A lack of effective and tolerable therapeutic options and nonspecific symptoms at presentation with advanced stage of disease are among the challenges in the management of the disease. AREAS COVERED An overview of ovarian cancer, followed by a discussion of the current therapeutic regimes and challenges that arise during and after the treatment of EOC. We discuss different formats of antibody therapeutics and their usage in targeting validated targets implicated in ovarian cancer, as well as three emerging novel proteins as examples recently implicated in their contribution to adaptive resistance in ovarian cancer. EXPERT OPINION Antibody therapeutics allow for a unique and effective way to target proteins implicated in cancer and other diseases, and have the potential to radically change the outcomes of patients suffering from ovarian cancer. The vast array of targets that have been implicated in ovarian cancer and yet the lack of effective therapeutic options for patients further stresses the importance of discovering novel proteins that can be targeted, as well as predictive biomarkers that can inform the stratification of patients into treatment-specific populations.
Collapse
Affiliation(s)
- Mason Ruiz
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, Texas 77030, USA
| | - Ningyan Zhang
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, Texas 77030, USA
| | - Anil K Sood
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Zhiqiang An
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, Texas 77030, USA
| |
Collapse
|
130
|
Recouvreux MS, Miao J, Gozo MC, Wu J, Walts AE, Karlan BY, Orsulic S. FOXC2 Promotes Vasculogenic Mimicry in Ovarian Cancer. Cancers (Basel) 2022; 14:4851. [PMID: 36230774 PMCID: PMC9564305 DOI: 10.3390/cancers14194851] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 09/27/2022] [Accepted: 09/29/2022] [Indexed: 11/16/2022] Open
Abstract
FOXC2 is a forkhead family transcription factor that plays a critical role in specifying mesenchymal cell fate during embryogenesis. FOXC2 expression is associated with increased metastasis and poor survival in various solid malignancies. Using in vitro and in vivo assays in mouse ovarian cancer cell lines, we confirmed the previously reported mechanisms by which FOXC2 could promote cancer growth, metastasis, and drug resistance, including epithelial-mesenchymal transition, stem cell-like differentiation, and resistance to anoikis. In addition, we showed that FOXC2 expression is associated with vasculogenic mimicry in mouse and human ovarian cancers. FOXC2 overexpression increased the ability of human ovarian cancer cells to form vascular-like structures in vitro, while inhibition of FOXC2 had the opposite effect. Thus, we present a novel mechanism by which FOXC2 might contribute to cancer aggressiveness and poor patient survival.
Collapse
Affiliation(s)
- Maria Sol Recouvreux
- Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Jiangyong Miao
- Department of Pathology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Maricel C. Gozo
- Women’s Cancer Program, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Jingni Wu
- Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Ann E. Walts
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Beth Y. Karlan
- Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
- Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Sandra Orsulic
- Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
- Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, CA 90095, USA
- Veterans Administration Greater Los Angeles Healthcare System, Los Angeles, CA 90095, USA
| |
Collapse
|
131
|
Suszczyk D, Skiba W, Zardzewiały W, Pawłowska A, Włodarczyk K, Polak G, Tarkowski R, Wertel I. Clinical Value of the PD-1/PD-L1/PD-L2 Pathway in Patients Suffering from Endometriosis. Int J Mol Sci 2022; 23:ijms231911607. [PMID: 36232911 PMCID: PMC9570092 DOI: 10.3390/ijms231911607] [Citation(s) in RCA: 6] [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: 08/29/2022] [Revised: 09/23/2022] [Accepted: 09/29/2022] [Indexed: 11/09/2022] Open
Abstract
The interaction between dendritic cells (DCs) and T cells mediated by the programmed cell death 1 (PD-1)/programmed cell death ligand 1 (PD-L1)/programmed cell death ligand 2 (PD-L2) pathway is the most important point in regulating immunological tolerance and autoimmunity. Disturbances in the quantity, maturity, and activity of DCs may be involved in the implantation and growth of endometrial tissue outside the uterus in endometriosis (EMS). However, little is known about the role of the immune checkpoint pathways in EMS. In our study, we examined the expression of PD-L1/PD-L2 on myeloid DCs (mDCs) and plasmacytoid DCs (pDCs) in the peripheral blood (PB) and peritoneal fluid (PF) of both EMS patients (n = 72) and healthy subjects (n = 20) via flow cytometry. The concentration of soluble PD-L1 and PD-L2 in the plasma and PF of EMS patients and the control group were determined using ELISA. We demonstrated an elevated percentage of mDCs, mDCs and pDCs with the PD-L1or PD-L2 expression, and a higher concentration of the soluble forms of PD-L1 and PD-L2 in the PF than in the plasma of EMS patients. We conclude that the peritoneal cavity environment and the PD-1/PD-L1/PD-L2 axis may play an important role in the modulation of immune response and the development and/or progression of EMS.
Collapse
Affiliation(s)
- Dorota Suszczyk
- Independent Laboratory of Cancer Diagnostics and Immunology, Medical University of Lublin, Chodźki 4a, 20-093 Lublin, Poland
- Correspondence:
| | - Wiktoria Skiba
- Independent Laboratory of Cancer Diagnostics and Immunology, Medical University of Lublin, Chodźki 4a, 20-093 Lublin, Poland
| | - Witold Zardzewiały
- Students’ Scientific Association, Independent Laboratory of Cancer Diagnostics and Immunology, Medical University of Lublin, Chodźki 4a, 20-093 Lublin, Poland
| | - Anna Pawłowska
- Independent Laboratory of Cancer Diagnostics and Immunology, Medical University of Lublin, Chodźki 4a, 20-093 Lublin, Poland
| | - Karolina Włodarczyk
- Independent Laboratory of Cancer Diagnostics and Immunology, Medical University of Lublin, Chodźki 4a, 20-093 Lublin, Poland
| | - Grzegorz Polak
- I Chair and Department of Gynaecologic Oncology and Gynaecology, Medical University of Lublin, Staszica 16, 20-081 Lublin, Poland
| | - Rafał Tarkowski
- I Chair and Department of Gynaecologic Oncology and Gynaecology, Medical University of Lublin, Staszica 16, 20-081 Lublin, Poland
| | - Iwona Wertel
- Independent Laboratory of Cancer Diagnostics and Immunology, Medical University of Lublin, Chodźki 4a, 20-093 Lublin, Poland
| |
Collapse
|
132
|
Fanale D, Corsini LR, Brando C, Cutaia S, Di Donna MC, Filorizzo C, Lisanti MC, Randazzo U, Magrin L, Romano R, Bazan Russo TD, Olive D, Vieni S, Pantuso G, Chiantera V, Russo A, Bazan V, Iovanna JL. Can circulating PD-1, PD-L1, BTN3A1, pan-BTN3As, BTN2A1 and BTLA levels enhance prognostic power of CA125 in patients with advanced high-grade serous ovarian cancer? Front Oncol 2022; 12:946319. [PMID: 36212445 PMCID: PMC9532861 DOI: 10.3389/fonc.2022.946319] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Accepted: 08/29/2022] [Indexed: 11/13/2022] Open
Abstract
The most common subtype of ovarian cancer (OC) is the high-grade serous ovarian carcinoma (HGSOC), accounting for 70%–80% of all OC deaths. Although HGSOC is a potentially immunogenic tumor, clinical studies assessing the effectiveness of inhibitors of programmed death protein and its ligand (PD-1/PD-L1) in OC patients so far showed only response rates <15%. However, recent studies revealed an interesting prognostic role of plasma PD-1/PD-L1 and other circulating immunoregulatory molecules, such as the B- and T-lymphocyte attenuator (BTLA), butyrophilin sub-family 3A/CD277 receptors (BTN3A), and butyrophilin sub-family 2 member A1 (BTN2A1), in several solid tumors. Since evidence showed the prognostic relevance of pretreatment serum CA125 levels in OC, the aim of our study was to investigate if soluble forms of inhibitory immune checkpoints can enhance prognostic power of CA125 in advanced HGSOC women. Using specific ELISA tests, we examined the circulating PD-1, PD-L1, pan-BTN3As, BTN3A1, BTN2A1, and BTLA levels in 100 advanced HGSOC patients before treatment, correlating them with baseline serum CA125, age at diagnosis, body mass index (BMI), and peritoneal carcinomatosis. A multivariate analysis revealed that plasma BTN3A1 ≤4.75 ng/ml (HR, 1.94; 95% CI, 1.23–3.07; p=0.004), age at diagnosis ≤60 years (HR, 1.65; 95% CI, 1.05–2.59; p=0.03) and absence of peritoneal carcinomatosis (HR, 2.65; 95% CI, 1.66–4.22; p<0.0001) were independent prognostic factors for a longer progression-free survival (PFS) (≥30 months) in advanced HGSOC women. However, further two-factor multivariate analyses highlighted that baseline serum CA125 levels >401 U/ml and each soluble protein above respective concentration cutoff were covariates associated with shorter PFS (<30 months) and unfavorable clinical outcome, suggesting that contemporary measurement of both biomarkers than CA125 only could strengthen prognostic power of serum CA125 in predicting PFS of advanced HGSOC women. Plasma PD-L1, PD-1, BTN3A1, pan-sBTN3As, BTN2A1, or BTLA levels could be helpful biomarkers to increase prognostic value of CA125.
Collapse
Affiliation(s)
- Daniele Fanale
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, Palermo, Italy
| | - Lidia Rita Corsini
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, Palermo, Italy
| | - Chiara Brando
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, Palermo, Italy
| | - Sofia Cutaia
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, Palermo, Italy
| | | | - Clarissa Filorizzo
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, Palermo, Italy
| | - Maria Chiara Lisanti
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, Palermo, Italy
| | - Ugo Randazzo
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, Palermo, Italy
| | - Luigi Magrin
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, Palermo, Italy
| | - Raffaella Romano
- Department of Gynecologic Oncology, University of Palermo, Palermo, Italy
| | - Tancredi Didier Bazan Russo
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, Palermo, Italy
| | - Daniel Olive
- Team Immunity and Cancer, Centre de Recherche en Cancérologie de Marseille (CRCM), Institut National de la Santé et de la Recherche Médicale (INSERM) U1068, Centre National de la Recherche Scientifique Unité Mixte de Recherche (CNRS UMR) 7258 Aix-Marseille Université and Institut Paoli-Calmettes, Marseille, France
| | - Salvatore Vieni
- Division of General and Oncological Surgery, Department of Surgical, Oncological and Oral Sciences, University of Palermo, Palermo, Italy
| | - Gianni Pantuso
- Division of General and Oncological Surgery, Department of Surgical, Oncological and Oral Sciences, University of Palermo, Palermo, Italy
| | - Vito Chiantera
- Department of Gynecologic Oncology, University of Palermo, Palermo, Italy
| | - Antonio Russo
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, Palermo, Italy
- *Correspondence: Viviana Bazan, ; Antonio Russo,
| | - Viviana Bazan
- Department of Biomedicine, Neuroscience and Advanced Diagnostics, University of Palermo, Palermo, Italy
- *Correspondence: Viviana Bazan, ; Antonio Russo,
| | - Juan Lucio Iovanna
- Centre de Recherche en Cancérologie de Marseille (CRCM), Institut National de la Santé et de la Recherche Médicale (INSERM) U1068, Centre National de la Recherche Scientifique Unité Mixte de Recherche (CNRS UMR) 7258, Aix-Marseille Université and Institut Paoli-Calmettes, Parc Scientifique et Technologique de Luminy, Marseille, France
| |
Collapse
|
133
|
Kepenekian V, Bhatt A, Péron J, Alyami M, Benzerdjeb N, Bakrin N, Falandry C, Passot G, Rousset P, Glehen O. Advances in the management of peritoneal malignancies. Nat Rev Clin Oncol 2022; 19:698-718. [PMID: 36071285 DOI: 10.1038/s41571-022-00675-5] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/29/2022] [Indexed: 11/09/2022]
Abstract
Peritoneal surface malignancies (PSMs) are usually associated with a poor prognosis. Nonetheless, in line with advances in the management of most abdominopelvic metastatic diseases, considerable progress has been made over the past decade. An improved understanding of disease biology has led to the more accurate prediction of neoplasia aggressiveness and the treatment response and has been reflected in the proposal of new classification systems. Achieving complete cytoreductive surgery remains the cornerstone of curative-intent treatment of PSMs. Alongside centralization in expert centres, enabling the delivery of multimodal and multidisciplinary strategies, preoperative management is a crucial step in order to select patients who are most likely to benefit from surgery. Depending on the specific PSM, the role of intraperitoneal chemotherapy and of perioperative systemic chemotherapy, in particular, in the neoadjuvant setting, is established in certain scenarios but questioned in several others, although more prospective data are required. In this Review, we describe advances in all aspects of the management of PSMs including disease biology, assessment and improvement of disease resectability, perioperative management, systemic therapy and pre-emptive management, and we speculate on future research directions.
Collapse
Affiliation(s)
- Vahan Kepenekian
- Surgical Oncology Department, Hôpital Lyon Sud, Hospices Civils de Lyon, Pierre Bénite, France.,CICLY - EA3738, Université Claude Bernard Lyon I (UCBL1), Lyon, France
| | - Aditi Bhatt
- Department of Surgical Oncology, Zydus hospital, Ahmedabad, Gujarat, India
| | - Julien Péron
- Medical Oncology Department, Hôpital Lyon Sud, Hospices Civils de Lyon, Pierre Bénite, France.,Laboratoire de Biométrie et Biologie Evolutive, Equipe Biostatistique-Santé, UCBL1, Lyon, France
| | - Mohammad Alyami
- Department of General Surgery and Surgical Oncology, Oncology Center, King Khalid Hospital, Najran, Saudi Arabia
| | - Nazim Benzerdjeb
- CICLY - EA3738, Université Claude Bernard Lyon I (UCBL1), Lyon, France.,Department of Pathology, Institut de Pathologie Multisite, Hospices Civils de Lyon, UCBL1, Lyon, France
| | - Naoual Bakrin
- Surgical Oncology Department, Hôpital Lyon Sud, Hospices Civils de Lyon, Pierre Bénite, France.,CICLY - EA3738, Université Claude Bernard Lyon I (UCBL1), Lyon, France
| | - Claire Falandry
- Department of Onco-Geriatry, Hôpital Lyon Sud, Hospices Civils de Lyon, Lyon, France
| | - Guillaume Passot
- Surgical Oncology Department, Hôpital Lyon Sud, Hospices Civils de Lyon, Pierre Bénite, France.,CICLY - EA3738, Université Claude Bernard Lyon I (UCBL1), Lyon, France
| | - Pascal Rousset
- CICLY - EA3738, Université Claude Bernard Lyon I (UCBL1), Lyon, France.,Department of Radiology, Hôpital Lyon Sud, Hospices Civils de Lyon, UCBL1, Lyon, France
| | - Olivier Glehen
- Surgical Oncology Department, Hôpital Lyon Sud, Hospices Civils de Lyon, Pierre Bénite, France. .,CICLY - EA3738, Université Claude Bernard Lyon I (UCBL1), Lyon, France.
| |
Collapse
|
134
|
Fanale D, Dimino A, Pedone E, Brando C, Corsini LR, Filorizzo C, Fiorino A, Lisanti MC, Magrin L, Randazzo U, Bazan Russo TD, Russo A, Bazan V. Prognostic and Predictive Role of Tumor-Infiltrating Lymphocytes (TILs) in Ovarian Cancer. Cancers (Basel) 2022; 14:4344. [PMID: 36139508 PMCID: PMC9497073 DOI: 10.3390/cancers14184344] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 09/02/2022] [Accepted: 09/02/2022] [Indexed: 12/12/2022] Open
Abstract
In the last decade, tumor-infiltrating lymphocytes (TILs) have been recognized as clinically relevant prognostic markers for improved survival, providing the immunological basis for the development of new therapeutic strategies and showing a significant prognostic and predictive role in several malignancies, including ovarian cancer (OC). In fact, many OCs show TILs whose typology and degree of infiltration have been shown to be strongly correlated with prognosis and survival. The OC histological subtype with the higher presence of TILs is the high-grade serous carcinoma (HGSC) followed by the endometrioid subtype, whereas mucinous and clear cell OCs seem to contain a lower percentage of TILs. The abundant presence of TILs in OC suggests an immunogenic potential for this tumor. Despite the high immunogenic potential, OC has been described as a highly immunosuppressive tumor with a high expression of PD1 by TILs. Although further studies are needed to better define their role in prognostic stratification and the therapeutic implication, intraepithelial TILs represent a relevant prognostic factor to take into account in OC. In this review, we will discuss the promising role of TILs as markers which are able to reflect the anticancer immune response, describing their potential capability to predict prognosis and therapy response in OC.
Collapse
Affiliation(s)
- Daniele Fanale
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, 90127 Palermo, Italy
| | - Alessandra Dimino
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, 90127 Palermo, Italy
| | - Erika Pedone
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, 90127 Palermo, Italy
| | - Chiara Brando
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, 90127 Palermo, Italy
| | - Lidia Rita Corsini
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, 90127 Palermo, Italy
| | - Clarissa Filorizzo
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, 90127 Palermo, Italy
| | - Alessia Fiorino
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, 90127 Palermo, Italy
| | - Maria Chiara Lisanti
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, 90127 Palermo, Italy
| | - Luigi Magrin
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, 90127 Palermo, Italy
| | - Ugo Randazzo
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, 90127 Palermo, Italy
| | - Tancredi Didier Bazan Russo
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, 90127 Palermo, Italy
| | - Antonio Russo
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, 90127 Palermo, Italy
| | - Viviana Bazan
- Department of Biomedicine, Neuroscience and Advanced Diagnostics, University of Palermo, 90127 Palermo, Italy
| |
Collapse
|
135
|
Li T, Liu T, Zhao Z, Pan Y, Xu X, Zhang Y, Zhan S, Zhou S, Zhu W, Guo H, Yang R. Antifungal immunity mediated by C-type lectin receptors may be a novel target in immunotherapy for urothelial bladder cancer. Front Immunol 2022; 13:911325. [PMID: 36131933 PMCID: PMC9483128 DOI: 10.3389/fimmu.2022.911325] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Accepted: 07/18/2022] [Indexed: 11/29/2022] Open
Abstract
Immunotherapies, such as immune-checkpoint blockade and adoptive T-cell therapy, offer novel treatment options with good efficacy for patients with urothelial bladder cancer. However, heterogeneity and therapeutic resistance have limited the use of immunotherapy. Further research into immune-regulatory mechanisms in bladder cancer is urgently required. Emerging evidence demonstrates that the commensal microbiota and its interactions with host immunity play pivotal roles in a variety of physiological and pathological processes, including in cancer. The gut microbiota has been identified as a potentially effective target of treatment that can be synergized with immunotherapy. The urothelial tract is also a key site for multiple microbes, although the immune-regulatory role of the urinary microbiome in the process of carcinogenesis of bladder cancer remains to be elucidated. We performed a comprehensive analysis of the expression and biological functions of C-type lectin receptors (CLRs), which have been recognized as innate pathogen-associated receptors for fungal microbiota, in bladder cancer. In line with previous research on fungal colonization of the urothelial tract, we found that CLRs, including Dectin-1, Dectin-2, Dectin-3, and macrophage-inducible Ca2+-dependent lectin receptor (Mincle), had a significant association with immune infiltration in bladder cancer. Multiple innate and adaptive pathways are positively correlated with the upregulation of CLRs. In addition, we found a significant correlation between the expression of CLRs and a range of immune-checkpoint proteins in bladder cancer. Based on previous studies and our findings, we hypothesize that the urinary mycobiome plays a key role in the pathogenesis of bladder cancer and call for more research on CLR-mediated anti-fungal immunity against bladder cancer as a novel target for immunotherapy in urothelial bladder cancer.
Collapse
Affiliation(s)
- Tianhang Li
- Department of Urology, Affiliated Nanjing Drum Tower Hospital, Medical School, Nanjing University, Nanjing, China
| | - Tianyao Liu
- Department of Urology, Affiliated Nanjing Drum Tower Hospital, Medical School, Nanjing University, Nanjing, China
| | - Zihan Zhao
- Department of Urology, Affiliated Nanjing Drum Tower Hospital, Medical School, Nanjing University, Nanjing, China
| | - Yuchen Pan
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing, China
| | - Xinyan Xu
- Department of Urology, Affiliated Nanjing Drum Tower Hospital, Medical School, Nanjing University, Nanjing, China
| | - Yulin Zhang
- Department of Urology, Affiliated Nanjing Drum Tower Hospital, Medical School, Nanjing University, Nanjing, China
| | - Shoubin Zhan
- Jiangsu Engineering Research Center for microRNA Biology and Biotechnology, State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Shengkai Zhou
- Jiangsu Engineering Research Center for microRNA Biology and Biotechnology, State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Wenjie Zhu
- Department of Urology, Affiliated Nanjing Drum Tower Hospital, Medical School, Nanjing University, Nanjing, China
| | - Hongqian Guo
- Department of Urology, Affiliated Nanjing Drum Tower Hospital, Medical School, Nanjing University, Nanjing, China
- *Correspondence: Rong Yang, ; Hongqian Guo,
| | - Rong Yang
- Department of Urology, Affiliated Nanjing Drum Tower Hospital, Medical School, Nanjing University, Nanjing, China
- *Correspondence: Rong Yang, ; Hongqian Guo,
| |
Collapse
|
136
|
Padmanabhan S, Gaire B, Zou Y, Uddin MM, Vancurova I. IFNγ-induced PD-L1 expression in ovarian cancer cells is regulated by JAK1, STAT1 and IRF1 signaling. Cell Signal 2022; 97:110400. [PMID: 35820543 PMCID: PMC9357219 DOI: 10.1016/j.cellsig.2022.110400] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 07/04/2022] [Accepted: 07/06/2022] [Indexed: 11/26/2022]
Abstract
Expression of the immune checkpoint programmed death ligand-1 (PD-L1) is increased in ovarian cancer (OC) and correlates with poor prognosis. Interferon-γ (IFNγ) induces PD-L1 expression in OC cells, resulting in their increased proliferation and tumor growth, but the mechanisms that regulate the PD-L1 expression in OC remain unclear. Here, we show that the IFNγ-induced PD-L1 expression in OC cells is associated with increased levels of STAT1, Tyr-701 pSTAT1 and Ser-727 pSTAT1. Suppression of JAK1 and STAT1 significantly decreases the IFNγ-induced PD-L1 expression in OC cells, and STAT1 overexpression increases the IFNγ-induced PD-L1 expression. In addition, IFNγ induces expression of the transcription factor interferon regulatory factor 1 (IRF1) and IRF1 suppression attenuates the IFNγ-induced gene and protein levels of PD-L1. Chromatin immunoprecipitation results show that IFNγ induces PD-L1 promoter acetylation and recruitment of STAT1, Ser-727 pSTAT1 and IRF1 in OC cells. Together, these findings demonstrate that the IFNγ-induced PD-L1 expression in OC cells is regulated by JAK1, STAT1, and IRF1 signaling, and suggest that targeting the JAK1/ STAT1/IRF1 pathway may provide a leverage to regulate the PD-L1 levels in ovarian cancer.
Collapse
Affiliation(s)
- Sveta Padmanabhan
- Department of Biological Sciences, St. John's University, New York 11439, USA
| | - Bijaya Gaire
- Department of Biological Sciences, St. John's University, New York 11439, USA
| | - Yue Zou
- Department of Biological Sciences, St. John's University, New York 11439, USA
| | - Mohammad M Uddin
- Department of Biological Sciences, St. John's University, New York 11439, USA
| | - Ivana Vancurova
- Department of Biological Sciences, St. John's University, New York 11439, USA.
| |
Collapse
|
137
|
Liu T, Liu Q, Wang Y, Yang R, Tian F. Cuproptosis scoring model predicts overall survival and assists in immunotherapeutic decision making in pancreatic carcinoma. Front Genet 2022; 13:938488. [PMID: 36118866 PMCID: PMC9472214 DOI: 10.3389/fgene.2022.938488] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Accepted: 07/11/2022] [Indexed: 11/16/2022] Open
Abstract
Background: Cuproptosis is a newly identified form of non-apoptotic cell death that is associated with the progression and treatment responses in pancreatic adenocarcinoma (PAAD). However, its impact on oncology and tumor microenvironment (TME) remains unclear. Methods: Hub genes were identified using least absolute shrinkage and selection operator (LASSO) Cox regression for 25 newly reported cuproptosis-related regulators and subjected to stepwise regression to obtain cuproptosis-related score (CuRS). Additionally, the clinical significance, functional status, role on TME, and genomic variation of CuRS were further examined systematically. Results: A CuRS model incorporating TRAF2, TRADD, USP21, FAS, MLKL, TNFRSF10B, MAPK8, TRAF5, and RIPK3 was developed. The stability and accuracy of this risk model as an independent prognostic factor for PAAD were confirmed in the training and external validation cohorts. Patients in the high-CuRS group had “cold” tumors with active tumor proliferation and immunosuppression, whereas those in the low-CuRS group comprised “hot” tumors with active immune function and cell killing capacity. Additionally, patients in the high-CuRS group carried fewer genomic copy number variations (CNVs) and greater somatic mutations. Furthermore, patients in the low- and high-CuRS groups exhibited increased sensitivity to immunotherapy and chemotherapy, respectively. Conclusion: We developed and validated a robust CuRS model based on cuproptosis to assess patients’ prognoses and guide clinical decision-making. Overall, the findings of this study are expected to contribute to the comprehensive understanding of cuproptosis and facilitate precise treatment of PAAD.
Collapse
Affiliation(s)
- Tijun Liu
- Department of Rehabilitation Medicine, Xiantao First People’s Hospital Affiliated to Yangtze University, Xiantao, China
| | - Qing Liu
- Department of Rehabilitation Medicine, Xiantao First People’s Hospital Affiliated to Yangtze University, Xiantao, China
| | - Yongju Wang
- Department of Rehabilitation Medicine, Xiantao First People’s Hospital Affiliated to Yangtze University, Xiantao, China
| | - Rong Yang
- Department of Oncology, Xiantao First People’s Hospital Affiliated to Yangtze University, Xiantao, China
| | - Fang Tian
- Department of Rehabilitation Medicine, Xiantao First People’s Hospital Affiliated to Yangtze University, Xiantao, China
- *Correspondence: Fang Tian,
| |
Collapse
|
138
|
Yang X, Yang F, Lan L, Wen N, Li H, Sun X. Diagnostic and prognostic value of m5C regulatory genes in hepatocellular carcinoma. Front Genet 2022; 13:972043. [PMID: 36105093 PMCID: PMC9465290 DOI: 10.3389/fgene.2022.972043] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 08/05/2022] [Indexed: 12/20/2022] Open
Abstract
Background: A high mortality rate makes hepatocellular carcinoma (HCC) one of the most common types of cancer globally. 5-methylcytosine (m5C) is an epigenetic modification that contributes to the prognosis of several cancers, but its relevance to HCC remains unknown. We sought to determine if the m5C-related regulators had any diagnostic or prognostic value in HCC. Methods: M5C regulatory genes were screened and compared between HCC and normal tissue from The Cancer Genome Atlas (TCGA)and Gene Expression Omnibus (GEO) databases. Least absolute shrinkage and selection operator method (LASSO) and univariate Cox regression analysis of differentially expressed genes were then performed to identify diagnostic markers. A LASSO prognostic model was constructed using M5C regulatory genes with prognostic values screened by TCGA expression data. HCC patients were stratified based on risk score, then clinical characteristics analysis and immune correlation analysis were performed for each subgroup, and the molecular functions of different subgroups were analyzed using both Gene Set Enrichment Analysis (GSEA) and Gene Set Variation Analysis (GSVA). The prognostic model was evaluated using univariate and multivariate Cox analyses as well as a nomogram. Molecular typing was performed according to m5C regulatory genes and immune checkpoint genes expression respectively, and clinical characterization and immune correlation analysis were performed for each subgroup. Results: M5C regulatory genes are expressed differently in HCC patients with different clinical and pathological characteristics, and mutations in these genes are frequent. Based on five m5C regulators (NOP2, NSUN2, TET1, YBX1, and DNMT3B), we constructed a prognostic model with high predictive ability. The risk score was found to be an independent prognostic indicator. Additionally, risk scores can also be applied in subgroups with different clinical characteristics as prognostic indicators. Conclusion: The study combined data from TCGA and GEO for the first time to reveal the genetic and prognostic significance of m5C-related regulators in HCC, which provides new directions for identifying predictive biomarkers and developing molecularly targeted therapies for HCC.
Collapse
Affiliation(s)
- Xiawei Yang
- Graduate School, Guangxi Medical University, Nanning, China
| | - Feng Yang
- Department of Gynocology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Liugen Lan
- Transplant Medical Center, The Second Affiliated Hospital of Guangxi Medical University, Nanning, China
- Guangxi Key Laboratory of Organ Donation and Transplantation, Nanning, China
- Guangxi Key Laboratory for Transplantation Medicine, Nanning, China
- Guangxi Transplantation Medicine Research Center of Engineering Technology, Nanning, China
| | - Ning Wen
- Transplant Medical Center, The Second Affiliated Hospital of Guangxi Medical University, Nanning, China
- Guangxi Key Laboratory of Organ Donation and Transplantation, Nanning, China
- Guangxi Key Laboratory for Transplantation Medicine, Nanning, China
- Guangxi Transplantation Medicine Research Center of Engineering Technology, Nanning, China
| | - Haibin Li
- Transplant Medical Center, The Second Affiliated Hospital of Guangxi Medical University, Nanning, China
- Guangxi Key Laboratory of Organ Donation and Transplantation, Nanning, China
- Guangxi Key Laboratory for Transplantation Medicine, Nanning, China
- Guangxi Transplantation Medicine Research Center of Engineering Technology, Nanning, China
| | - Xuyong Sun
- Graduate School, Guangxi Medical University, Nanning, China
- Transplant Medical Center, The Second Affiliated Hospital of Guangxi Medical University, Nanning, China
- Guangxi Key Laboratory of Organ Donation and Transplantation, Nanning, China
- Guangxi Key Laboratory for Transplantation Medicine, Nanning, China
- Guangxi Transplantation Medicine Research Center of Engineering Technology, Nanning, China
- *Correspondence: Xuyong Sun,
| |
Collapse
|
139
|
Wei S, Gu X, Zhang W. Development and validation of a novel fibroblast scoring model for lung adenocarcinoma. Front Oncol 2022; 12:905212. [PMID: 36072807 PMCID: PMC9444064 DOI: 10.3389/fonc.2022.905212] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Accepted: 07/22/2022] [Indexed: 11/23/2022] Open
Abstract
The interaction between cancer-associated fibroblasts (CAFs) and the tumor microenvironment (TME) is a key factor for promoting tumor progression. In lung cancer, the crosstalk between CAFs and malignant and immune cells is expected to provide new directions for the development of immunotherapy. In this study, we have systematically analyzed a single-cell dataset and identified interacting genes between CAFs and other cells. Subsequently, a robust fibroblast-related score (FRS) was developed. Kaplan-Meier (KM) and ROC analyses showed its good predictive power for patient prognoses in the training set comprising of specimens from the cancer genome atlas (TCGA) and in three external validation sets from the Gene Expression Omnibus (GEO). Univariate and multivariate Cox regression analyses suggested that FRS was a significant prognostic factor independent of multiple clinical characteristics. Functional enrichment and ssGSEA analyses indicated that patients with a high FRS developed “cold” tumors with active tumor proliferation and immunosuppression capacities. In contrast, those with a low FRS developed “hot” tumors with active immune function and cell killing abilities. Genomic variation analysis showed that the patients with a high FRS possessed a higher somatic mutation burden and copy number alterations and were more sensitive to chemotherapy; patients with a low FRS were more sensitive to immunotherapy, particularly anti-PD1 therapy. Overall, these findings advance the understanding of CAFs in tumor progression and we generated a reliable FRS-based model to assess patient prognoses and guide clinical decision-making.
Collapse
Affiliation(s)
- Shiyou Wei
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Xuyu Gu
- School of Medicine, Southeast University, Nanjing, China
| | - Wentian Zhang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
- *Correspondence: Wentian Zhang,
| |
Collapse
|
140
|
Caro AA, Deschoemaeker S, Allonsius L, Coosemans A, Laoui D. Dendritic Cell Vaccines: A Promising Approach in the Fight against Ovarian Cancer. Cancers (Basel) 2022; 14:cancers14164037. [PMID: 36011029 PMCID: PMC9406463 DOI: 10.3390/cancers14164037] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 08/16/2022] [Accepted: 08/19/2022] [Indexed: 11/22/2022] Open
Abstract
Simple Summary With an overall 5-year survival of only 20% for advanced-stage ovarian cancer patients, enduring and effective therapies are a highly unmet clinical need. Current standard-of-care therapies are able to improve progression-free survival; however, patients still relapse. Moreover, immunotherapy has not resulted in clear patient benefits so far. In this situation, dendritic cell vaccines can serve as a potential therapeutic addition against ovarian cancer. In the current review, we provide an overview of the different dendritic cell subsets and the roles they play in ovarian cancer. We focus on the advancements in dendritic cell vaccination against ovarian cancer and highlight the key outcomes and pitfalls associated with currently used strategies. Finally, we address future directions that could be taken to improve the dendritic cell vaccination outcomes in ovarian cancer. Abstract Ovarian cancer (OC) is the deadliest gynecological malignancy in developed countries and is the seventh-highest cause of death in women diagnosed with cancer worldwide. Currently, several therapies are in use against OC, including debulking surgery, chemotherapy, as well as targeted therapies. Even though the current standard-of-care therapies improve survival, a vast majority of OC patients relapse. Additionally, immunotherapies have only resulted in meager patient outcomes, potentially owing to the intricate immunosuppressive nexus within the tumor microenvironment. In this scenario, dendritic cell (DC) vaccination could serve as a potential addition to the therapeutic options available against OC. In this review, we provide an overview of current therapies in OC, focusing on immunotherapies. Next, we highlight the potential of using DC vaccines in OC by underscoring the different DC subsets and their functions in OC. Finally, we provide an overview of the advances and pitfalls of current DC vaccine strategies in OC while providing future perspectives that could improve patient outcomes.
Collapse
Affiliation(s)
- Aarushi Audhut Caro
- Laboratory of Myeloid Cell Immunology, VIB Center for Inflammation Research, 1050 Brussels, Belgium
- Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel, 1050 Brussels, Belgium
- Laboratory of Tumor Immunology and Immunotherapy, Department of Oncology, Leuven Cancer Institute, KU Leuven, 3000 Leuven, Belgium
| | - Sofie Deschoemaeker
- Laboratory of Myeloid Cell Immunology, VIB Center for Inflammation Research, 1050 Brussels, Belgium
- Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel, 1050 Brussels, Belgium
| | - Lize Allonsius
- Laboratory of Myeloid Cell Immunology, VIB Center for Inflammation Research, 1050 Brussels, Belgium
- Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel, 1050 Brussels, Belgium
| | - An Coosemans
- Laboratory of Tumor Immunology and Immunotherapy, Department of Oncology, Leuven Cancer Institute, KU Leuven, 3000 Leuven, Belgium
| | - Damya Laoui
- Laboratory of Myeloid Cell Immunology, VIB Center for Inflammation Research, 1050 Brussels, Belgium
- Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel, 1050 Brussels, Belgium
- Correspondence: ; Tel.: +32-2-6291969
| |
Collapse
|
141
|
Phosphofructokinase 1 platelet isoform induces PD-L1 expression to promote glioblastoma immune evasion. Genes Genomics 2022; 44:1509-1517. [PMID: 35917090 DOI: 10.1007/s13258-022-01291-4] [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: 04/01/2022] [Accepted: 07/18/2022] [Indexed: 11/04/2022]
Abstract
BACKGROUND Overexpression of PD-L1 is observed in many types of human cancer, including glioblastoma (GBM) and contributes to tumor immune evasion. In addition, GBM shows highly-activated aerobic glycolysis due to overexpression of phosphofructokinase 1 platelet isoform (PFKP), which the key enzyme in the glycolysis. However, it remains unclear whether the metabolic enzyme PFKP plays a role in the regulation of PD-L1 expression and GBM immune evasion. OBJECTIVE We aimed to investigate the non-metabolic role of PFKP in PD-L1 expression-induced GBM immune evasion. METHODS The mechanisms of PFKP-induced PD-L1 expression were studied by several experiments, including real-time PCR, immunoblot analysis, and ATP production. The coculture experiments using GBM cell and T cells were performed to evaluate the effect of PFKP on T cell activation. The clinical relationship between PFKP and PD-L1 was analyzed in The Cancer Genome Atlas (TCGA) database and in human GBM specimens. RESULTS We showed that PFKP promotes EGFR activation-induced PD-L1 expression in human GBM cells. Importantly, we demonstrated that EGFR-phosphorylated PFKP Y64 plays an important role in AKT-mediated β-catenin transactivation and subsequent PD-L1 transcriptional expression, thereby enhancing the GBM immune evasion. In addition, based on our findings, the levels of PFKP Y64 phosphorylation are positively correlated with PD-L1 expression in human GBM specimens, highlighting the clinical significance of PFKP Y64 phosphorylation in the GBM immune evasion. CONCLUSION These findings provide new mechanistic insight into the regulation of PD-L1 expression by a non-metabolic function of PFKP on tumor cells.
Collapse
|
142
|
Damato A, Rotolo M, Caputo F, Borghi E, Iachetta F, Pinto C. New Potential Immune Biomarkers in the Era of Precision Medicine: Lights and Shadows in Colorectal Cancer. Life (Basel) 2022; 12:1137. [PMID: 36013315 PMCID: PMC9410155 DOI: 10.3390/life12081137] [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: 07/02/2022] [Revised: 07/21/2022] [Accepted: 07/26/2022] [Indexed: 11/16/2022] Open
Abstract
Genetic alterations in CRC have shown a negative predictive and prognostic role in specific target therapies. The onset of immunotherapy has also undergone remarkable therapeutic innovation, although limited to a small subgroup of patients, the MSI-H/dMMR, which represents only 5% of CRC. Research is moving forward to identify whether other biomarkers can predict response to ICIs, despite various limitations regarding expression and identification methods. For this purpose, TMB, LAG3, and PD-L1 expression have been retrospectively evaluated in several solid tumors establishing the rationale to design clinical trials with concurrent inhibition of LAG3 and PD-1 results in a significant advantage in PFS and OS in advanced melanoma patients. Based on these data, there are clinical trials ongoing in the CRC as well. This review aims to highlight what is already known about genetic mutations and genomic alterations in CRC, their inhibition with targeted therapies and immune checkpoints inhibitors, and new findings useful to future treatment strategies.
Collapse
Affiliation(s)
- Angela Damato
- Medical Oncology, Comprehensive Cancer Center, Azienda USL-IRCCS Reggio Emilia, 42122 Reggio Emilia, Italy; (M.R.); (F.C.); (E.B.); (F.I.); (C.P.)
| | | | | | | | | | | |
Collapse
|
143
|
Khatoon E, Parama D, Kumar A, Alqahtani MS, Abbas M, Girisa S, Sethi G, Kunnumakkara AB. Targeting PD-1/PD-L1 axis as new horizon for ovarian cancer therapy. Life Sci 2022; 306:120827. [PMID: 35907493 DOI: 10.1016/j.lfs.2022.120827] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Revised: 07/08/2022] [Accepted: 07/16/2022] [Indexed: 02/08/2023]
Abstract
Ovarian cancer is among the deadliest gynecological cancers and the 7th most commonly occurring cancer in women globally. The 5 year survival rate is estimated to be less than 25 %, as in most cases, diagnosis occurs at an advanced stage. Despite recent advancements in treatment, clinical outcomes still remain poor, thus implicating the need for urgent identification of novel therapeutics for the treatment of this cancer. Ovarian cancer is considered a low immune reactive cancer as the tumor cells express insufficient neoantigens to be recognized by the immune cells and thus tend to escape from immune surveillance. Thus, in the recent decade, immunotherapy has gained significant attention and has rejuvenated the understanding of immune regulation in tumor biology. One of the critical immune checkpoints is programmed cell death-1 (PD-1)/programmed cell death ligand-1 (PD-L1) axis. Engagement of PD-1 to PD-L1 promotes immunologic tolerance and suppresses the effector T cells and maintains tumor Tregs, thus playing a crucial role in enhancing tumor survival. Recent studies are targeted to develop inhibitors that block this signal to augment the anti-tumor activity of immune cells. Also, compared to monotherapy, the combinatorial treatment of immune checkpoint inhibitors with small molecule inhibitors have shown promising results with improved efficacy and acceptable adverse events. The present review provides an overview of the PD-1/PD-L1 axis and role of non-coding RNAs in regulating this axis. Moreover, we have highlighted the various preclinical and clinical investigations on PD-1/PD-L1 immune checkpoint inhibitors and have discussed the limitations of immunotherapies in ovarian cancer.
Collapse
Affiliation(s)
- Elina Khatoon
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology (IIT) Guwahati, Guwahati 781 039, Assam, India
| | - Dey Parama
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology (IIT) Guwahati, Guwahati 781 039, Assam, India
| | - Aviral Kumar
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology (IIT) Guwahati, Guwahati 781 039, Assam, India
| | - Mohammed S Alqahtani
- Radiological Sciences Department, College of Applied Medical Sciences, King Khalid University, Abha 61421, Saudi Arabia; BioImaging Unit, Space Research Centre, Michael Atiyah Building, University of Leicester, Leicester LE1 7RH, UK
| | - Mohamed Abbas
- Electrical Engineering Department, College of Engineering, King Khalid University, Abha 61421, Saudi Arabia; Computers and communications Department, College of Engineering, Delta University for Science and Technology, Gamasa 35712, Egypt
| | - Sosmitha Girisa
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology (IIT) Guwahati, Guwahati 781 039, Assam, India
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore.
| | - Ajaikumar B Kunnumakkara
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology (IIT) Guwahati, Guwahati 781 039, Assam, India.
| |
Collapse
|
144
|
Garg T, Weiss CR, Sheth RA. Techniques for Profiling the Cellular Immune Response and Their Implications for Interventional Oncology. Cancers (Basel) 2022; 14:3628. [PMID: 35892890 PMCID: PMC9332307 DOI: 10.3390/cancers14153628] [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: 07/08/2022] [Revised: 07/19/2022] [Accepted: 07/20/2022] [Indexed: 12/07/2022] Open
Abstract
In recent years there has been increased interest in using the immune contexture of the primary tumors to predict the patient's prognosis. The tumor microenvironment of patients with cancers consists of different types of lymphocytes, tumor-infiltrating leukocytes, dendritic cells, and others. Different technologies can be used for the evaluation of the tumor microenvironment, all of which require a tissue or cell sample. Image-guided tissue sampling is a cornerstone in the diagnosis, stratification, and longitudinal evaluation of therapeutic efficacy for cancer patients receiving immunotherapies. Therefore, interventional radiologists (IRs) play an essential role in the evaluation of patients treated with systemically administered immunotherapies. This review provides a detailed description of different technologies used for immune assessment and analysis of the data collected from the use of these technologies. The detailed approach provided herein is intended to provide the reader with the knowledge necessary to not only interpret studies containing such data but also design and apply these tools for clinical practice and future research studies.
Collapse
Affiliation(s)
- Tushar Garg
- Division of Vascular and Interventional Radiology, Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; (T.G.); (C.R.W.)
| | - Clifford R. Weiss
- Division of Vascular and Interventional Radiology, Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; (T.G.); (C.R.W.)
| | - Rahul A. Sheth
- Department of Interventional Radiology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
145
|
Li L. Tumor Microenvironment Characterization in Breast Cancer and an Immune Cell Infiltration Score Development, Validation, and Application. Front Oncol 2022; 12:844082. [PMID: 35832543 PMCID: PMC9273207 DOI: 10.3389/fonc.2022.844082] [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: 01/12/2022] [Accepted: 05/19/2022] [Indexed: 11/23/2022] Open
Abstract
The tumor microenvironment (TME) refers to the cellular environment in which tumors exist. An increasing number of reports have emphasized its role in tumor progression, prognosis, relapse, metastasis, and therapeutic response with breast cancer (BRCA). Few studies have revealed a systematic landscape of immune cell infiltration (ICI) in BRCA. In this study, we comprehensively analyzed the immune cells infiltrating TME in BRCA. Three ICI patterns were identified through an unsupervised clustering method and an ICI score was developed by a principal component analysis (PCA). A Kaplan-Meier survival with log-rank test revealed a significant overall survival (OS) difference of BRCA patients with these three ICI patterns. We also found that a high ICI score was characterized by an elevated tumor mutation burden (TMB), effector T-cell infiltration, INF-γ-related cytotoxicity, and cytolytic activity score. An independent cohort validated that this ICI score could be a prognostic indicator for BRCA. Two immunotherapeutic cohorts and two chemotherapeutic cohorts confirmed that patients with higher ICI scores showed significant chemotherapeutic and immunotherapeutic advantages. In summary, these results suggest that the ICI patterns could act as a prognostic indicator and that the ICI score could precisely predict the clinical outcome for BRCA patients.
Collapse
Affiliation(s)
- Lang Li
- Department of Hematology, Jinhua Hospital of Traditional Chinese Medicine, Jinhua, China
| |
Collapse
|
146
|
Hamade DF, Espinal A, Yu J, Leibowitz BJ, Fisher R, Hou W, Shields D, van Pijkeren JP, Mukherjee A, Epperly MW, Vlad A, Coffman L, Wang H, Huq MS, Patel R, Huang J, Greenberger JS. Lactobacillus reuteri Releasing IL-22 (LR-IL-22) Facilitates Intestinal Radioprotection for Whole-Abdomen Irradiation (WAI) of Ovarian Cancer. Radiat Res 2022; 198:89-105. [PMID: 35446961 PMCID: PMC9278541 DOI: 10.1667/rade-21-00224.1] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 03/28/2022] [Indexed: 12/13/2022]
Abstract
Oral administration (gavage) of a second-generation probiotic, Lactobacillus reuteri (L. reuteri), that releases interleukin-22 (LR-IL-22) at 24 h after total-body irradiation (TBI) mitigates damage to the intestine. We determined that LR-IL-22 also mitigates partial-body irradiation (PBI) and whole-abdomen irradiation (WAI). Irradiation can be an effective treatment for ovarian cancer, but its use is limited by intestinal toxicity. Strategies to mitigate toxicity are important and can revitalize this modality to treat ovarian cancer. In the present studies, we evaluated whether LR-IL-22 facilitates fractionated WAI in female C57BL/6 mice with disseminated ovarian cancer given a single fraction of either 15.75 Gy or 19.75 Gy or 4 daily fractions of 6 Gy or 6.5 Gy. Mice receiving single or multiple administrations of LR-IL-22 during WAI showed improved intestinal barrier integrity (P = 0.0167), reduced levels of radiation-induced intestinal cytokines including KC/CXCL1 (P = 0.002) and IFN-γ (P = 0.0024), and reduced levels of plasma, Eotaxin/CCL11 (P = 0.0088). LR-IL-22 significantly preserved the numbers of Lgr5+GFP+ intestinal stem cells (P = 0.0010) and improved survival (P < 0.0343). Female C57BL/6MUC-1 mice with widespread abdominal syngeneic 2F8cis ovarian cancer that received LR-IL-22 during 6.5 Gy WAI in 4 fractions had reduced tumor burden, less intestinal toxicity, and improved 30-day survival. Furthermore, LR-IL-22 facilitated WAI when added to Paclitaxel and Carboplatin chemotherapy and further increased survival. Oral administration (gavage) of LR-IL-22 is a potentially valuable intestinal radioprotector, which can facilitate therapeutic WAI for widespread intra-abdominal ovarian cancer.
Collapse
Affiliation(s)
- Diala F. Hamade
- Department of Radiation Oncology, UPMC Hillman Cancer Center, Pittsburgh, PA 15232
| | - Alexis Espinal
- Department of Radiation Oncology, UPMC Hillman Cancer Center, Pittsburgh, PA 15232
| | - Jian Yu
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA 15260
| | | | - Renee Fisher
- Department of Radiation Oncology, UPMC Hillman Cancer Center, Pittsburgh, PA 15232
| | - Wen Hou
- Department of Radiation Oncology, UPMC Hillman Cancer Center, Pittsburgh, PA 15232
| | - Donna Shields
- Department of Radiation Oncology, UPMC Hillman Cancer Center, Pittsburgh, PA 15232
| | | | - Amitava Mukherjee
- Department of Radiation Oncology, UPMC Hillman Cancer Center, Pittsburgh, PA 15232
| | - Michael W. Epperly
- Department of Radiation Oncology, UPMC Hillman Cancer Center, Pittsburgh, PA 15232
| | - Anda Vlad
- Department of OB/Gyn and Reproductive Sciences, University of Pittsburgh, Pittsburgh, PA 15260
| | - Lan Coffman
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA 15260
| | - Hong Wang
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, PA 15260
| | - M. Saiful Huq
- Department of Radiation Oncology, UPMC Hillman Cancer Center, Pittsburgh, PA 15232
| | - Ravi Patel
- Department of Radiation Oncology, UPMC Hillman Cancer Center, Pittsburgh, PA 15232
| | - Jason Huang
- Department of Radiation Oncology, UPMC Hillman Cancer Center, Pittsburgh, PA 15232
| | - Joel S. Greenberger
- Department of Radiation Oncology, UPMC Hillman Cancer Center, Pittsburgh, PA 15232
| |
Collapse
|
147
|
Bayrak AF, Eliyatkın NÖ, İşlek A, Özkul Y, Kılıç HS, Aktas S. Association of immune response with overall and disease-free survival in laryngeal squamous cell carcinomas. Am J Otolaryngol 2022; 43:103477. [PMID: 35537231 DOI: 10.1016/j.amjoto.2022.103477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 04/20/2022] [Accepted: 05/01/2022] [Indexed: 11/01/2022]
Abstract
OBJECTIVES This study aimed to examine the relationship between checkpoint receptors (PD-1, PD-L1, PD-L2, CTLA-4) and lymphoid infiltration level (TILs) with prognostic features of patients with laryngeal squamous cell carcinoma (LSCC). METHODS A retrospective study was designed at a tertiary referential university hospital between April 2008 and December 2020. The surgical specimen of the patients who met the eligibility criteria were re-examined histopathological, sociodemographic, clinical, pathological, and follow-up findings of patients were determined. The impact of PD-1, PD-L1, PD-L2, CTLA4, and TILs levels for the presence of cancer recurrence, disease-specific mortality, overall survival (OS), disease-free survival (DFS) was investigated. RESULTS Forty-five patients with LSCC were included in the study. The mean follow-up period was 48.3 ± 14.3 months (min: 36, max 84). TILs scores were detected significantly lower in patients with distant metastasis and recurrence (p = 0.046 and 0.010). Also, only TILs was a significant risk factor for recurrence and survival among the PD-1, PD-L1, PD-L2, CTLA-4, and TILs (HR = 0.217 CI: 0.070-0.679, p = 0.009 and HR = 0.566, CI: 0,321-980, p = 0.048). Similarly, for the TILs score: > 1 was significant for DFS. (Long-Rank = 0.009). The examined markers and TILs scores were not a significant predictive factor for OS. CONCLUSION An increase in TILs density in LSCCs is associated with a better prognosis. However, PD-1, PD-L1, PD-L2, CTLA-4 could not be associated with prognosis. Controlled studies combined with immunotherapy treatment results are needed to reveal their role as a marker and prognostic factor of the anti-tumor immune response.
Collapse
|
148
|
TNFRSF9 Suppressed the Progression of Breast Cancer via the p38MAPK/PAX6 Signaling Pathway. JOURNAL OF ONCOLOGY 2022; 2022:8549781. [PMID: 35799609 PMCID: PMC9256432 DOI: 10.1155/2022/8549781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 06/08/2022] [Accepted: 06/09/2022] [Indexed: 11/18/2022]
Abstract
Worldwide, breast cancer is the most common cancer in females. Endocrine therapy can effectively treat 85% of breast cancer patients, but 15% of patients could only be treated with chemotherapy and surgery, and the prognosis is much worse. Immunotherapy is the novel treatment for breast cancer, where PD-1 and CTLA-4 antibodies have shown evidence of immune modulation in breast cancer drug trials. In this study, we report that TNFRSF9 regulates the cell proliferation, invasion, and apoptosis of breast cancer cells through regulating the phosphorylation of p38, thus further regulating the expression of PAX6. In both breast cancer tissues and cell lines, the levels of TNFRSF9 are significantly decreased, and breast cancer cell development will be promoted with knockdown of TNFRSF9. Moreover, we identify that downregulation of TNFRSF9 can upregulate the phosphorylated p38 (p-p38) and PAX6. We further elucidate that p-p38 is essential for PAX6 expression as p38 phosphorylation inhibitor can reverse the upregulation of PAX6 and suppress cell proliferation and invasion and promote apoptosis in breast cancer cells. In summary, this study proposed a novel TNFRSF9/p38/PAX6 axis that contributes to tumor suppression, which suggests a potential immunotherapy target for breast cancer.
Collapse
|
149
|
Sun H, Li Y, Si W, Hua T, Chen J, Kang S. Genetic Variation of PD-L1 Gene Affects its Expression and Is Related to Clinical Outcome in Epithelial Ovarian Cancer. Front Oncol 2022; 12:763134. [PMID: 35719980 PMCID: PMC9204247 DOI: 10.3389/fonc.2022.763134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 04/26/2022] [Indexed: 11/21/2022] Open
Abstract
Objective This study aims to investigate the effect of polymorphisms of programmed cell death-ligand 1 (PD-L1) on the risk and patient’s outcomes of epithelial ovarian cancer (EOC). Methods Totally, 568 patients and 532 healthy women were included. Three polymorphisms in the PD-L1 gene, rs2297136, rs4143815 and rs4742098, were genotyped by the polymerase chain reaction/ligase detection reaction (PCR-LDR). Survival analysis was performed in 234 patients (received primary debulking surgery followed by platinum-based chemotherapy). Results Patients with the rs2297136 AG + GG genotypes had shorter progression-free survival (PFS) (hazard ratio (HR)=1.44, 95% CI=1.03-2.01) and overall survival (OS) (HR=1.55, 95% CI=1.06-2.27) than those with the AA genotype. Moreover, the mRNA and protein expression levels of PD-L1 in EOC tissues with the rs2297136 AG + GG genotypes were remarkably higher than those with the AA genotype (P=0.032 and P=0.047, respectively). Survival analysis showed that high expression of PD-L1 mRNA was remarkably associated with worse 10-year PFS (HR=1.55, 95% CI=1.28-1.88) and OS (HR=1.51, 95% CI=1.00-2.28) in EOC patients. Conclusions The rs2297136 may not only effectively influence the expression of PD-L1, but also is significantly associated with EOC patients’ outcomes.
Collapse
Affiliation(s)
- Haiyan Sun
- Department of Obstetrics and Gynecology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Yan Li
- Department of Molecular Biology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Wengang Si
- Department of Obstetrics and Gynecology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Tian Hua
- Department of Obstetrics and Gynecology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Juan Chen
- Department of Obstetrics and Gynecology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Shan Kang
- Department of Obstetrics and Gynecology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
150
|
Yung MMH, Siu MKY, Ngan HYS, Chan DW, Chan KKL. Orchestrated Action of AMPK Activation and Combined VEGF/PD-1 Blockade with Lipid Metabolic Tunning as Multi-Target Therapeutics against Ovarian Cancers. Int J Mol Sci 2022; 23:ijms23126857. [PMID: 35743298 PMCID: PMC9224484 DOI: 10.3390/ijms23126857] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 06/06/2022] [Accepted: 06/17/2022] [Indexed: 02/06/2023] Open
Abstract
Ovarian cancer is one of the most lethal gynecological malignancies worldwide, and chemoresistance is a critical obstacle in the clinical management of the disease. Recent studies have suggested that exploiting cancer cell metabolism by applying AMP-activated protein kinase (AMPK)-activating agents and distinctive adjuvant targeted therapies can be a plausible alternative approach in cancer treatment. Therefore, the perspectives about the combination of AMPK activators together with VEGF/PD-1 blockade as a dual-targeted therapy against ovarian cancer were discussed herein. Additionally, ferroptosis, a non-apoptotic regulated cell death triggered by the availability of redox-active iron, have been proposed to be governed by multiple layers of metabolic signalings and can be synergized with immunotherapies. To this end, ferroptosis initiating therapies (FITs) and metabolic rewiring and immunotherapeutic approaches may have substantial clinical potential in combating ovarian cancer development and progression. It is hoped that the viewpoints deliberated in this review would accelerate the translation of remedial concepts into clinical trials and improve the effectiveness of ovarian cancer treatment.
Collapse
Affiliation(s)
- Mingo M. H. Yung
- Department of Obstetrics & Gynaecology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China; (M.M.H.Y.); (M.K.Y.S.); (H.Y.S.N.)
| | - Michelle K. Y. Siu
- Department of Obstetrics & Gynaecology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China; (M.M.H.Y.); (M.K.Y.S.); (H.Y.S.N.)
| | - Hextan Y. S. Ngan
- Department of Obstetrics & Gynaecology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China; (M.M.H.Y.); (M.K.Y.S.); (H.Y.S.N.)
| | - David W. Chan
- Department of Obstetrics & Gynaecology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China; (M.M.H.Y.); (M.K.Y.S.); (H.Y.S.N.)
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
- Correspondence: or (D.W.C.); (K.K.L.C.); Tel.: +852-3917-9367 or +852-3943-6053 (D.W.C.); +852-2255-4260 (K.K.L.C.); Fax: +852-2816-1947 or +852-2603-5123 (D.W.C.); +852-2255-0947 (K.K.L.C.)
| | - Karen K. L. Chan
- Department of Obstetrics & Gynaecology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China; (M.M.H.Y.); (M.K.Y.S.); (H.Y.S.N.)
- Correspondence: or (D.W.C.); (K.K.L.C.); Tel.: +852-3917-9367 or +852-3943-6053 (D.W.C.); +852-2255-4260 (K.K.L.C.); Fax: +852-2816-1947 or +852-2603-5123 (D.W.C.); +852-2255-0947 (K.K.L.C.)
| |
Collapse
|