1
|
Kang M, Du W, Ding L, Wu M, Pei D. HIC1 suppresses Tumor Progression and Enhances CD8 + T Cells Infiltration Through Promoting GSDMD-induced Pyroptosis in Gastric Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2412083. [PMID: 40279559 DOI: 10.1002/advs.202412083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Revised: 02/25/2025] [Indexed: 04/27/2025]
Abstract
Recently, immune checkpoint blockade treatment has made remarkable strides in combatting malignancies, including gastric cancer (GC). Nonetheless, the efficacy of immunotherapy in GC patients remains constrained, warranting further exploration of the underlying molecular mechanisms to improve therapeutic outcomes. Hypermethylated in cancer 1 (HIC1) is acknowledged as a transcriptional regulator crucial for multiple aspects of cell development, yet its role in antitumor immune responses remains incompletely understood. This investigation reveals a significant downregulation of HIC1 in gastric cancer, correlating with a less favorable prognosis. Overexpression of HIC1 promotes the initiation of cell pyroptosis. Mechanistically, gasdermin D (GSDMD), a pivotal executor of pyroptosis, is identified as a downstream target of HIC1 and activated by HIC1 at the transcriptional level. Subsequent cleavage of the GSDMD N-terminal region punctures the cell membrane, instigating pyroptosis and releasing inflammatory factors. Furthermore, HIC1 augments the infiltration of CD8+ T cells to counteract immune evasion. The combinatorial approach of HIC1 overexpression with PD-L1 antibody demonstrates a synergistic therapeutic impact in treating GC. Additionally, c-Jun activation domain-binding protein 1 (Jab1) mediates the ubiquitylation and proteasomal degradation of HIC1 at Lys517. Ultimately, these findings underscore the potential of HIC1 as a promising immunotherapeutic target for the treatment of GC.
Collapse
Affiliation(s)
- Mengjie Kang
- Department of Pathology, School of Basic Medical Sciences, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu, 221004, China
| | - Wenqi Du
- Department of Pathology, School of Basic Medical Sciences, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu, 221004, China
- Department of Anatomy, School of Basic Medical Sciences, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu, 221004, China
| | - Lina Ding
- Department of Pathology, School of Basic Medical Sciences, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu, 221004, China
| | - Mengdi Wu
- Department of Pathology, School of Basic Medical Sciences, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu, 221004, China
| | - Dongsheng Pei
- Department of Pathology, School of Basic Medical Sciences, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu, 221004, China
| |
Collapse
|
2
|
Wang X, Li X, Niu L, Lv F, Guo T, Gao Y, Ran Y, Huang W, Wang B. FAK-LINC01089 negative regulatory loop controls chemoresistance and progression of small cell lung cancer. Oncogene 2024; 43:1669-1687. [PMID: 38594505 DOI: 10.1038/s41388-024-03027-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Revised: 03/28/2024] [Accepted: 04/03/2024] [Indexed: 04/11/2024]
Abstract
The focal adhesion kinase (FAK) tyrosine kinase is activated and upregulated in multiple cancer types including small cell lung cancer (SCLC). However, FAK inhibitors have shown limited efficacy in clinical trials for cancer treatment. With the aim of identifying potential therapeutic strategies to inhibit FAK for cancer treatment, we investigated long non-coding RNAs (lncRNAs) that potentially regulate FAK in SCLC. In this study, we identified a long non-coding RNA LINC01089 that binds and inhibits FAK phosphorylation (activation). Expression analysis revealed that LINC01089 was downregulated in SCLC tissues and negatively correlated with chemoresistance and survival in SCLC patients. Functionally, LINC01089 inhibited chemoresistance and progression of SCLC in vitro and in vivo. Mechanistically, LINC01089 inhibits FAK activation by blocking binding with Src and talin kinases, while FAK negatively regulates LINC01089 transcription by activating the ERK signaling pathway to recruit the REST transcription factor. Furthermore, LINC01089-FAK axis mediates the expression of drug resist-related genes by modulating YBX1 phosphorylation, leading to drug resistance in SCLC. Intriguingly, the FAK-LINC01089 interaction depends on the co-occurrence of the novel FAK variant and the non-conserved region of LINC01089 in primates. In Conclusion, our results indicated that LINC01089 may serve as a novel high-efficiency FAK inhibitor and the FAK-LINC01089 axis represents a valuable prognostic biomarker and potential therapeutic target in SCLC.
Collapse
Affiliation(s)
- Xianteng Wang
- Department of Urology, Shenzhen Institute of Translational Medicine, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical school, Shenzhen, 518060, China
- Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- Guangdong Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China
- School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, 518107, China
| | - Xingkai Li
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Liman Niu
- Chongqing Key Laboratory of Sichuan-Chongging Co-construction for Diagnosis and Treatment of Infectious Diseases Integrated Traditional Chinese and Western Medicine College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Fang Lv
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ting Guo
- Graduate School, Guangxi University of Chinese Medicine, Nanning, China
| | - Yushun Gao
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yuliang Ran
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| | - Weiren Huang
- Department of Urology, Shenzhen Institute of Translational Medicine, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical school, Shenzhen, 518060, China.
- Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China.
- Guangdong Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China.
| | - Bing Wang
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
3
|
Zhang B, Chen J, Yu H, Li M, Cai M, Chen L. Prognostic Nutritional Index Predicts Efficacy and Immune-Related Adverse Events of First-Line Chemoimmunotherapy in Patients with Extensive-Stage Small-Cell Lung Cancer. J Inflamm Res 2024; 17:1777-1788. [PMID: 38523686 PMCID: PMC10959246 DOI: 10.2147/jir.s450804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Accepted: 03/12/2024] [Indexed: 03/26/2024] Open
Abstract
Background Currently, there is a lack of well-established markers to predict the efficacy of chemoimmunotherapy in small-cell lung cancer (SCLC). Neutrophil-to-lymphocyte ratio (NLR), platelet-to-lymphocyte ratio (PLR), advanced lung cancer inflammation index (ALI) and prognostic nutritional index (PNI) are associated with prognosis in several tumors, whereas their predictive role in SCLC remains unclear. Methods A retrospective study was conducted at Sun Yat-sen University Cancer Center, involving extensive-stage SCLC (ES-SCLC) patients who received first-line chemoimmunotherapy between January 2020 and December 2021. Peripheral blood biomarkers were extracted from medical records and their correlation with prognosis and immune-related adverse events (IRAEs) was analyzed. Results A total of 114 patients were included. Patients with a low PLR, high ALI and high PNI had prolonged progression-free survival (PFS) compared to those with a high PLR, low ALI and low PNI. Patients with a low NLR, low PLR, high ALI and high PNI had prolonged overall survival (OS) compared to those with a high NLR, high PLR, low ALI and low PNI. Cox regression model showed that PNI was an independent risk factor for both PFS and OS. ROC curve showed that PNI outperforms NLR, PLR and ALI in predicting both PFS and OS. The PNI-based nomogram demonstrated strong predictive capability for both PFS and OS. In addition, there was a significant correlation between PNI and IRAEs. Conclusion A high baseline PNI might be associated with improved prognosis and the occurrence of IRAEs in ES-SCLC patients treated with first-line chemoimmunotherapy.
Collapse
Affiliation(s)
- Baishen Zhang
- Sun Yat-Sen University Cancer Center; State Key Laboratory of Oncology in South China; Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, People’s Republic of China
| | - Jing Chen
- Department of Medical Oncology, Sun Yat-Sen University Cancer Center; State Key Laboratory of Oncology in South China; Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, People’s Republic of China
| | - Hui Yu
- Department of Medical Oncology, Sun Yat-Sen University Cancer Center; State Key Laboratory of Oncology in South China; Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, People’s Republic of China
| | - Meichen Li
- Department of Medical Oncology, Sun Yat-Sen University Cancer Center; State Key Laboratory of Oncology in South China; Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, People’s Republic of China
| | - Muyan Cai
- Department of Pathology, Sun Yat-Sen University Cancer Center; State Key Laboratory of Oncology in South China; Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, People’s Republic of China
| | - Likun Chen
- Department of Medical Oncology, Sun Yat-Sen University Cancer Center; State Key Laboratory of Oncology in South China; Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, People’s Republic of China
| |
Collapse
|
4
|
Xagara A, Roumeliotou A, Kokkalis A, Tsapakidis K, Papakonstantinou D, Papadopoulos V, Samaras I, Chantzara E, Kallergi G, Kotsakis A. ES-SCLC Patients with PD-L1 + CTCs and High Percentages of CD8 +PD-1 +T Cells in Circulation Benefit from Front-Line Immunotherapy Treatment. Biomedicines 2024; 12:146. [PMID: 38255251 PMCID: PMC10813758 DOI: 10.3390/biomedicines12010146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 12/29/2023] [Accepted: 01/08/2024] [Indexed: 01/24/2024] Open
Abstract
SCLC is an aggressive cancer type with high metastatic potential and bad prognosis. CTCs are a valuable source of tumor cells in blood circulation and are among the major contributors to metastasis. In this study we evaluated the number of CTCs that express PD-L1 in treatment-naïve ES-SCLC patients receiving ICI in a front-line setting. Moreover, we explored the percentages of different immune T-cell subsets in circulation to assess their potential role in predicting responses. A total of 43 patients were enrolled-6 of them with LS-SCLC, and 37 with ES-SCLC disease. In addition, PBMCs from 10 healthy donors were used as a control group. Different T-cell subtypes were examined through multicolor FACS analysis and patients' CTCs were detected using immunofluorescence staining. SCLC patients had higher percentages of PD-1-expressing CD3+CD4+ and CD3+CD8+ T-cells, as well as elevated PD-1 protein expression compared to healthy individuals. Additionally, in ES-SCLC patients, a positive correlation between CD3+CD8+PD-1+ T-cells and PD-L1+ CTCs was detected. Importantly, patients harboring higher numbers of CD3+CD8+PD-1+ T-cells together with PD-L1+CTCs had a survival advantage when receiving front-line immunotherapy. Thus, this study proposes, for first time possible, immune cell-CTCs interaction, as well as a potential novel clinical biomarker for ICI responses in ES-SCLC patients.
Collapse
Affiliation(s)
- Anastasia Xagara
- Laboratory of Oncology, Faculty of Medicine, School of Health Sciences, University of Thessaly, GR-41110 Larissa, Greece; (A.X.); (A.K.); (K.T.); (V.P.); (I.S.); (E.C.)
| | - Argyro Roumeliotou
- Laboratory of Biochemistry/Metastatic Signaling, Section of Genetics, Cell Biology and Development, Department of Biology, University of Patras, GR-26504 Patras, Greece; (A.R.); (D.P.); (G.K.)
| | - Alexandros Kokkalis
- Laboratory of Oncology, Faculty of Medicine, School of Health Sciences, University of Thessaly, GR-41110 Larissa, Greece; (A.X.); (A.K.); (K.T.); (V.P.); (I.S.); (E.C.)
- Department of Medical Oncology, University General Hospital of Larissa, GR-41110 Larissa, Greece
| | - Konstantinos Tsapakidis
- Laboratory of Oncology, Faculty of Medicine, School of Health Sciences, University of Thessaly, GR-41110 Larissa, Greece; (A.X.); (A.K.); (K.T.); (V.P.); (I.S.); (E.C.)
- Department of Medical Oncology, University General Hospital of Larissa, GR-41110 Larissa, Greece
| | - Dimitris Papakonstantinou
- Laboratory of Biochemistry/Metastatic Signaling, Section of Genetics, Cell Biology and Development, Department of Biology, University of Patras, GR-26504 Patras, Greece; (A.R.); (D.P.); (G.K.)
| | - Vassilis Papadopoulos
- Laboratory of Oncology, Faculty of Medicine, School of Health Sciences, University of Thessaly, GR-41110 Larissa, Greece; (A.X.); (A.K.); (K.T.); (V.P.); (I.S.); (E.C.)
- Department of Medical Oncology, University General Hospital of Larissa, GR-41110 Larissa, Greece
| | - Ioannis Samaras
- Laboratory of Oncology, Faculty of Medicine, School of Health Sciences, University of Thessaly, GR-41110 Larissa, Greece; (A.X.); (A.K.); (K.T.); (V.P.); (I.S.); (E.C.)
- Department of Medical Oncology, University General Hospital of Larissa, GR-41110 Larissa, Greece
| | - Evagelia Chantzara
- Laboratory of Oncology, Faculty of Medicine, School of Health Sciences, University of Thessaly, GR-41110 Larissa, Greece; (A.X.); (A.K.); (K.T.); (V.P.); (I.S.); (E.C.)
- Department of Medical Oncology, University General Hospital of Larissa, GR-41110 Larissa, Greece
| | - Galatea Kallergi
- Laboratory of Biochemistry/Metastatic Signaling, Section of Genetics, Cell Biology and Development, Department of Biology, University of Patras, GR-26504 Patras, Greece; (A.R.); (D.P.); (G.K.)
| | - Athanasios Kotsakis
- Laboratory of Oncology, Faculty of Medicine, School of Health Sciences, University of Thessaly, GR-41110 Larissa, Greece; (A.X.); (A.K.); (K.T.); (V.P.); (I.S.); (E.C.)
- Department of Medical Oncology, University General Hospital of Larissa, GR-41110 Larissa, Greece
| |
Collapse
|
5
|
Kapagan T, Aksu F, Yuzkan S, Bulut N, Erdem GU. Atezolizumab-induced cerebellar ataxia in a patient with metastatic small cell lung cancer: A case report and literature review. J Oncol Pharm Pract 2024; 30:201-205. [PMID: 37321205 DOI: 10.1177/10781552231180594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
INTRODUCTION The use of immune checkpoint inhibitors, which have an important role in the treatment of malignant tumors, is increasing. Although rarely observed, neurological immune-related adverse events associated with immune checkpoint inhibitors result in high morbidity and mortality. Small cell lung cancer is a common cause of neurological paraneoplastic syndromes. The differentiation between paraneoplastic syndromes and neurological immune-related adverse events is important in patients using immune checkpoint inhibitors. Cerebellar ataxia caused by atezolizumab is a rare immune-related adverse event. CASE REPORT In this context, we present a 66-year-old man with small cell lung cancer who developed immune-mediated cerebellar ataxia after three cycles of atezolizumab, a programmed cell death ligand-1 inhibitor. The admission of brain and spinal gadolinium-based contrast-enhanced magnetic resonance imaging supported the preliminary diagnosis and indicated leptomeningeal involvement. However, the blood tests and a lumbar puncture did not reveal any structural, biochemical, paraneoplastic, or infectious cause. MANAGEMENT AND OUTCOME High-dose steroid treatment resulted in an improvement in the radiological involvement, as evidenced both clinically and on follow-up whole spine magnetic resonance imaging. Therefore, the immunotherapy was discontinued. The patient was discharged on day 20 without neurological sequelae. DISCUSSION In light of this, we present this case to emphasize the differential diagnosis of neurological immune-related adverse events originating from immune checkpoint inhibitors, which require rapid diagnosis and treatment, and clinically similar paraneoplastic syndromes and radiologically similar leptomeningeal involvement, in a case of small cell lung cancer.
Collapse
Affiliation(s)
- Tanju Kapagan
- Department of Internal Medicine, Division of Medical Oncology, Başakşehir Çam and Sakura City Hospıtal, Istanbul, Turkey
| | - Faruk Aksu
- Department of Internal Medicine, Division of Medical Oncology, Başakşehir Çam and Sakura City Hospıtal, Istanbul, Turkey
| | - Sabahattin Yuzkan
- Department of Radiology, Başakşehir Çam and Sakura City Hospıtal, Istanbul, Turkey
| | - Nilufer Bulut
- Department of Internal Medicine, Division of Medical Oncology, Başakşehir Çam and Sakura City Hospıtal, Istanbul, Turkey
| | - Gokmen Umut Erdem
- Department of Internal Medicine, Division of Medical Oncology, Başakşehir Çam and Sakura City Hospıtal, Istanbul, Turkey
| |
Collapse
|
6
|
Yao S, Han Y, Yang M, Jin K, Lan H. Integration of liquid biopsy and immunotherapy: opening a new era in colorectal cancer treatment. Front Immunol 2023; 14:1292861. [PMID: 38077354 PMCID: PMC10702507 DOI: 10.3389/fimmu.2023.1292861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Accepted: 11/03/2023] [Indexed: 12/18/2023] Open
Abstract
Immunotherapy has revolutionized the conventional treatment approaches for colorectal cancer (CRC), offering new therapeutic prospects for patients. Liquid biopsy has shown significant potential in early screening, diagnosis, and postoperative monitoring by analyzing circulating tumor cells (CTC) and circulating tumor DNA (ctDNA). In the era of immunotherapy, liquid biopsy provides additional possibilities for guiding immune-based treatments. Emerging technologies such as mass spectrometry-based detection of neoantigens and flow cytometry-based T cell sorting offer new tools for liquid biopsy, aiming to optimize immune therapy strategies. The integration of liquid biopsy with immunotherapy holds promise for improving treatment outcomes in colorectal cancer patients, enabling breakthroughs in early diagnosis and treatment, and providing patients with more personalized, precise, and effective treatment strategies.
Collapse
Affiliation(s)
- Shiya Yao
- Department of Colorectal Surgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang, China
| | - Yuejun Han
- Department of Colorectal Surgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang, China
| | - Mengxiang Yang
- Department of Colorectal Surgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang, China
| | - Ketao Jin
- Department of Colorectal Surgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang, China
| | - Huanrong Lan
- Department of Surgical Oncology, Hangzhou Cancer Hospital, Hangzhou, Zhejiang, China
| |
Collapse
|
7
|
Hou W, Zhao Y, Zhu H. Predictive Biomarkers for Immunotherapy in Gastric Cancer: Current Status and Emerging Prospects. Int J Mol Sci 2023; 24:15321. [PMID: 37895000 PMCID: PMC10607383 DOI: 10.3390/ijms242015321] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 10/12/2023] [Accepted: 10/16/2023] [Indexed: 10/29/2023] Open
Abstract
Gastric cancer presents substantial management challenges, and the advent of immunotherapy has ignited renewed hope among patients. Nevertheless, a significant proportion of patients do not respond to immunotherapy, and adverse events associated with immunotherapy also occur on occasion, underscoring the imperative to identify suitable candidates for treatment. Several biomarkers, including programmed death ligand-1 expression, tumor mutation burden, mismatch repair status, Epstein-Barr Virus infection, circulating tumor DNA, and tumor-infiltrating lymphocytes, have demonstrated potential in predicting the effectiveness of immunotherapy in gastric cancer. However, the quest for the optimal predictive biomarker for gastric cancer immunotherapy remains challenging, as each biomarker carries its own limitations. Recently, multi-omics technologies have emerged as promising platforms for discovering novel biomarkers that may help in selecting gastric cancer patients likely to respond to immunotherapy. The identification of reliable predictive biomarkers for immunotherapy in gastric cancer holds the promise of enhancing patient selection and improving treatment outcomes. In this review, we aim to provide an overview of clinically established biomarkers of immunotherapy in gastric cancer. Additionally, we introduce newly reported biomarkers based on multi-omics studies in the context of gastric cancer immunotherapy, thereby contributing to the ongoing efforts to refine patient stratification and treatment strategies.
Collapse
Affiliation(s)
- Wanting Hou
- Division of Abdominal Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu 610065, China; (W.H.); (Y.Z.)
- Department of Radiation Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu 610065, China
| | - Yaqin Zhao
- Division of Abdominal Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu 610065, China; (W.H.); (Y.Z.)
- Department of Radiation Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu 610065, China
| | - Hong Zhu
- Division of Abdominal Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu 610065, China; (W.H.); (Y.Z.)
| |
Collapse
|
8
|
Antigene MYCN Silencing by BGA002 Inhibits SCLC Progression Blocking mTOR Pathway and Overcomes Multidrug Resistance. Cancers (Basel) 2023; 15:cancers15030990. [PMID: 36765949 PMCID: PMC9913109 DOI: 10.3390/cancers15030990] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 01/27/2023] [Accepted: 01/30/2023] [Indexed: 02/08/2023] Open
Abstract
Small-cell lung cancer (SCLC) is the most aggressive lung cancer type, and is associated with smoking, low survival rate due to high vascularization, metastasis and drug resistance. Alterations in MYC family members are biomarkers of poor prognosis for a large number of SCLC. In particular, MYCN alterations define SCLC cases with immunotherapy failure. MYCN has a highly restricted pattern of expression in normal cells and is an ideal target for cancer therapy but is undruggable by traditional approaches. We propose an innovative approach to MYCN inhibition by an MYCN-specific antigene-PNA oligonucleotide (BGA002)-as a new precision medicine for MYCN-related SCLC. We found that BGA002 profoundly and specifically inhibited MYCN expression in SCLC cells, leading to cell-growth inhibition and apoptosis, while also overcoming multidrug resistance. These effects are driven by mTOR pathway block in concomitance with autophagy reactivation, thus avoiding the side effects of targeting mTOR in healthy cells. Moreover, we identified an MYCN-related SCLC gene signature comprehending CNTFR, DLX5 and TNFAIP3, that was reverted by BGA002. Finally, systemic treatment with BGA002 significantly increased survival in MYCN-amplified SCLC mouse models, including in a multidrug-resistant model in which tumor vascularization was also eliminated. These findings warrant the clinical testing of BGA002 in MYCN-related SCLC.
Collapse
|
9
|
Hou W, Yi C, Zhu H. Predictive biomarkers of colon cancer immunotherapy: Present and future. Front Immunol 2022; 13:1032314. [PMID: 36483562 PMCID: PMC9722772 DOI: 10.3389/fimmu.2022.1032314] [Citation(s) in RCA: 76] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 11/08/2022] [Indexed: 11/23/2022] Open
Abstract
Immunotherapy has revolutionized colon cancer treatment. Immune checkpoint inhibitors (ICIs) have shown clinical benefits for colon cancer patients, especially those with high microsatellite instability (MSI-H). In 2020, the US Food and Drug Administration (FDA)-approved ICI pembrolizumab as the first-line treatment for metastatic MSI-H colon cancer patients. Additionally, neoadjuvant immunotherapy has presented efficacy in treating early-stage colon cancer patients. Although MSI has been thought of as an effective predictive biomarker for colon cancer immunotherapy, only a small proportion of colon cancer patients were MSI-H, and certain colon cancer patients with MSI-H presented intrinsic or acquired resistance to immunotherapy. Thus, further search for predictive biomarkers to stratify patients is meaningful in colon cancer immunotherapy. Except for MSI, other biomarkers, such as PD-L1 expression level, tumor mutation burden (TMB), tumor-infiltrating lymphocytes (TILs), certain gut microbiota, ctDNA, and circulating immune cells were also proposed to be correlated with patient survival and ICI efficacy in some colon cancer clinical studies. Moreover, developing new diagnostic techniques helps identify accurate predictive biomarkers for colon cancer immunotherapy. In this review, we outline the reported predictive biomarkers in colon cancer immunotherapy and further discuss the prospects of technological changes for biomarker development in colon cancer immunotherapy.
Collapse
Affiliation(s)
- Wanting Hou
- Department of Medical Oncology Cancer Center, West China Hospital, Sichuan University, Sichuan, China
| | - Cheng Yi
- Department of Medical Oncology Cancer Center, West China Hospital, Sichuan University, Sichuan, China
| | - Hong Zhu
- Department of Medical Oncology Cancer Center, West China Hospital, Sichuan University, Sichuan, China
| |
Collapse
|
10
|
Ma J, Song P, Liu X, Ma C, Zheng M, Ren X, Wang R, Liu W, Lu Z, Li J. Insights into the roles and driving forces of CCT3 in human tumors. Front Pharmacol 2022; 13:1005855. [PMID: 36313331 PMCID: PMC9596777 DOI: 10.3389/fphar.2022.1005855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 09/28/2022] [Indexed: 11/22/2022] Open
Abstract
CCT3 played a key role in many cancers. This study aimed to further explore the characteristics of CCT3 from a pan-cancer perspective and reveal the driving forces for CCT3. By bioinformatic analysis, we found that the mRNA and protein levels of CCT3 were abnormally elevated in most tumor types and were correlated with poor prognosis. Single-cell sequencing data indicated an abnormal increase of CCT3 expression in both malignant cells and multiple immune cells. In the tumor microenvironment, CCT3 expression was negatively relevant with immune cell infiltration and immune checkpoint genes expression. In colon cancer, knockdown of CCT3 inhibited cell proliferation. Gene set enrichment analysis showed that CCT3 may be oncogenic by regulating amino acid metabolism. Furthermore, we predicted sensitive drugs for CCT3 by virtual screening and sensitivity analysis. Many driver genes such as TP53 and KRAS were essential for CCT3 overexpression. Epigenetic factors, enhancers in particular, were also critical for CCT3 expression. Additionally, we constructed the lncRNA/circRNA-miRNA-CCT3 regulatory network. Collectively, CCT3 had the potential to be a diagnostic and prognostic biomarker for multiple tumor types. CCT3 expression was relevant with an immunosuppressive tumor microenvironment. CCT3 could be a new molecular target for colon cancer. Both genetic and epigenetic factors were responsible for CCT3 expression in tumors.
Collapse
Affiliation(s)
- Jingang Ma
- Department of Oncology, Affiliated Hospital of Weifang Medical University, School of Clinical Medicine, Weifang Medical University, Weifang, China
| | - Ping Song
- Department of Gastroenterology, Affiliated Hangzhou First People’s Hospital, Medical School of Zhejiang University, Hangzhou, China
| | - Xinling Liu
- Department of Hematology, Laboratory for Stem Cell and Regenerative Medicine, Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Changgeng Ma
- Department of Radiotherapy, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Mingzhu Zheng
- Department of Oncology, Affiliated Hospital of Weifang Medical University, School of Clinical Medicine, Weifang Medical University, Weifang, China
| | - Xiaomin Ren
- Department of Oncology, Affiliated Hospital of Weifang Medical University, School of Clinical Medicine, Weifang Medical University, Weifang, China
| | - Rui Wang
- Department of Oncology, Affiliated Hospital of Weifang Medical University, School of Clinical Medicine, Weifang Medical University, Weifang, China
| | - Wenshan Liu
- Shandong Key Laboratory of Clinical Applied Pharmacology, Department of Pharmacy, Affiliated Hospital of Weifang Medical University, Weifang, China
- *Correspondence: Wenshan Liu, ; Zhong Lu, ; Jiaqiu Li,
| | - Zhong Lu
- Department of Oncology, Affiliated Hospital of Weifang Medical University, School of Clinical Medicine, Weifang Medical University, Weifang, China
- *Correspondence: Wenshan Liu, ; Zhong Lu, ; Jiaqiu Li,
| | - Jiaqiu Li
- Department of Oncology, Affiliated Hospital of Weifang Medical University, School of Clinical Medicine, Weifang Medical University, Weifang, China
- *Correspondence: Wenshan Liu, ; Zhong Lu, ; Jiaqiu Li,
| |
Collapse
|
11
|
Chen X, Yu D, Zhou H, Zhang X, Hu Y, Zhang R, Gao X, Lin M, Guo T, Zhang K. The role of EphA7 in different tumors. Clin Transl Oncol 2022; 24:1274-1289. [PMID: 35112312 DOI: 10.1007/s12094-022-02783-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 01/18/2022] [Indexed: 12/06/2022]
Abstract
Ephrin receptor A7 (EphA7) is a member of the Eph receptor family. It is widely involved in signal transduction between cells, regulates cell proliferation and differentiation, and participates in developing neural tubes and brain. In addition, EphA7 also has a dual role of tumor promoter and tumor suppressor. It can participate in cell proliferation, migration and apoptosis through various mechanisms, and affect tumor differentiation, staging and prognosis. EphA7 may be a potential diagnostic marker and tumor treatment target. This article reviews the effects of EphA7 on a variety of tumor biological processes and pathological characteristics, as well as specific effects and regulatory mechanisms.
Collapse
Affiliation(s)
- Xiangyi Chen
- Department of Orthopedics, Lanzhou University Second Hospital, 82 Cuiying Men, Lanzhou, 730030, China.,Gansu Key Laboratory of Bone and Joint Diseases, Lanzhou, 730030, China.,Xigu District People's Hospital, Lanzhou, 730030, China
| | - Dechen Yu
- Department of Orthopedics, Lanzhou University Second Hospital, 82 Cuiying Men, Lanzhou, 730030, China.,Gansu Key Laboratory of Bone and Joint Diseases, Lanzhou, 730030, China.,Xigu District People's Hospital, Lanzhou, 730030, China
| | - Haiyu Zhou
- Department of Orthopedics, Lanzhou University Second Hospital, 82 Cuiying Men, Lanzhou, 730030, China. .,Gansu Key Laboratory of Bone and Joint Diseases, Lanzhou, 730030, China. .,Xigu District People's Hospital, Lanzhou, 730030, China.
| | - Xiaobo Zhang
- Department of Orthopedics, Lanzhou University Second Hospital, 82 Cuiying Men, Lanzhou, 730030, China.,Gansu Key Laboratory of Bone and Joint Diseases, Lanzhou, 730030, China.,Xigu District People's Hospital, Lanzhou, 730030, China
| | - Yicun Hu
- Department of Orthopedics, Lanzhou University Second Hospital, 82 Cuiying Men, Lanzhou, 730030, China.,Gansu Key Laboratory of Bone and Joint Diseases, Lanzhou, 730030, China.,Xigu District People's Hospital, Lanzhou, 730030, China
| | - Ruihao Zhang
- Department of Orthopedics, Lanzhou University Second Hospital, 82 Cuiying Men, Lanzhou, 730030, China.,Gansu Key Laboratory of Bone and Joint Diseases, Lanzhou, 730030, China.,Xigu District People's Hospital, Lanzhou, 730030, China
| | - Xidan Gao
- Department of Orthopedics, Lanzhou University Second Hospital, 82 Cuiying Men, Lanzhou, 730030, China.,Gansu Key Laboratory of Bone and Joint Diseases, Lanzhou, 730030, China.,Xigu District People's Hospital, Lanzhou, 730030, China
| | - Maoqiang Lin
- Department of Orthopedics, Lanzhou University Second Hospital, 82 Cuiying Men, Lanzhou, 730030, China.,Gansu Key Laboratory of Bone and Joint Diseases, Lanzhou, 730030, China.,Xigu District People's Hospital, Lanzhou, 730030, China
| | - Taowen Guo
- Department of Orthopedics, Lanzhou University Second Hospital, 82 Cuiying Men, Lanzhou, 730030, China.,Gansu Key Laboratory of Bone and Joint Diseases, Lanzhou, 730030, China.,Xigu District People's Hospital, Lanzhou, 730030, China
| | - Kun Zhang
- Department of Orthopedics, Lanzhou University Second Hospital, 82 Cuiying Men, Lanzhou, 730030, China.,Gansu Key Laboratory of Bone and Joint Diseases, Lanzhou, 730030, China.,Xigu District People's Hospital, Lanzhou, 730030, China
| |
Collapse
|
12
|
Buch SA, Baba MR. Immune-Related Adverse Events (irAEs) in Cancer, with Inputs from a Nursing Expert: A Review. Indian J Med Paediatr Oncol 2022. [DOI: 10.1055/s-0042-1742442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
Abstract
AbstractImmune checkpoint inhibitors (ICPis) belong to a group of immunotherapeutic agents that act on different immune cells and tumor cells and reactivate the suppressed immune system of the host. The emergence of immunotherapy has resulted in the successful management of many malignancies. High success rates with certain advanced cancers have attributed wide importance and relevance to the use of immunotherapy. Although ICPis have gained huge popularity, their use often leads to side effects that can affect almost any system; immune-related adverse events (irAEs). These adverse events occur due to unrestrained T cell activity that unsettles the immune homeostasis of the host. Although close monitoring for toxicities controls the events on most of the occasions, the inability to diagnose them early may prove fatal on some occasions due to their subtle and nonspecific symptoms. This review summarizes in brief the usual irAEs and their management, besides a very important nursing perspective, from a nursing expert about an overall insight into the routine irAEs.
Collapse
Affiliation(s)
- Sajad Ahmad Buch
- Department of Oral Medicine and Radiology, Yenepoya Dental College, Yenepoya (Deemed to be University), Mangalore, Karnataka, India
| | - Mudasir Rashid Baba
- Yenepoya Physiotherapy College, Yenepoya (Deemed to be University), Mangalore, Karnataka, India
| |
Collapse
|
13
|
Zeng R, Liu F, Fang C, Yang J, Luo L, Yue P, Gao B, Dong Y, Xiang Y. PIV and PILE Score at Baseline Predict Clinical Outcome of Anti-PD-1/PD-L1 Inhibitor Combined With Chemotherapy in Extensive-Stage Small Cell Lung Cancer Patients. Front Immunol 2021; 12:724443. [PMID: 34777341 PMCID: PMC8586214 DOI: 10.3389/fimmu.2021.724443] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2021] [Accepted: 10/04/2021] [Indexed: 01/22/2023] Open
Abstract
Objectives The objective of this study is to evaluate whether PIV (Pan-Immune-Inflammation Value) and PILE [a score derived from PIV, lactate dehydrogenase (LDH), and Eastern Cooperative Oncology Group Performance Status (ECOG PS)] can predict clinical outcome of anti-PD-1/PD-L1 inhibitor combined with chemotherapy in patients with extensive-stage (ES) small cell lung cancer (SCLC). Methods A total of 53 patients with ES-SCLC in the control group of clinical trial (NCT03041311) were included in this study. PIV was calculated as follows: (neutrophil count × platelet count × monocyte count)/lymphocyte count. The PILE scores were composited based on PIV, LDH levels, and ECOG PS. The Kaplan–Meier method and Cox hazards regression models were used for survival analyses. Moreover, the predictive ability of PIV and PILE was validated in an independent real-world group consisting of 84 patients. Results Patients in the low PIV group (PIV < median) had longer progression-free survival (PFS) and overall survival (OS) than those in the high PIV group (PIV ≥ median), along with the HR, which was 2.157 and 2.359, respectively (PFS HR 95% CI: 1.181–3.940, p = 0.012; OS HR 95% CI: 1.168–4.762, p = 0.020). High PILE score was observed relating to worse treatment efficacy (disease control rate (DCR): 84.21% vs. 100%, p = 0.047; durable clinical benefit (DCB) rate: 10% vs. 48.5%, p = 0.060) and poor clinical outcome (median PFS: 4.75 vs. 5.53 m, p = 0.043; median OS: 7.13 vs. 15.93 m, p = 0.002). Similar results were obtained about the predictive and prognostic abilities of PIV and PILE scores in the validation group. Conclusions High PIV and high PILE were correlated with worse clinical outcomes in ES-SCLC patients treated with anti-PD-1/PD-L1 inhibitor combined with chemotherapy, reflecting that PIV and PILE might be useful to identify patients unlikely to benefit from anti-PD-1/PD-L1 therapy.
Collapse
Affiliation(s)
- Ran Zeng
- Department of Respiratory and Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fang Liu
- Department of Respiratory and Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chen Fang
- Respiratory and Critical Care Medicine Department, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Jin Yang
- Department of Respiratory and Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lifeng Luo
- Department of Respiratory and Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ping Yue
- Department of Respiratory and Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Beili Gao
- Department of Respiratory and Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuchao Dong
- Respiratory and Critical Care Medicine Department, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Yi Xiang
- Department of Respiratory and Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Institute of Respiratory Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|