1
|
Qiu Z, Li Z, Liu X, Zhang R, Li Y, Gao C, Mao X, Bao Y, Zhang M, Guo C. From tumor microenvironment to emerging biomarkers: the reshaping of the esophageal squamous cell carcinoma tumor microenvironment by neoadjuvant chemotherapy combined with immunotherapy. Front Immunol 2024; 15:1478922. [PMID: 39703499 PMCID: PMC11655454 DOI: 10.3389/fimmu.2024.1478922] [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: 08/11/2024] [Accepted: 11/15/2024] [Indexed: 12/21/2024] Open
Abstract
Esophageal squamous cell carcinoma is a cancer with high morbidity and mortality. The advent of immune checkpoint inhibitors has significantly increased complete response rates and postoperative R0 resection rates after neoadjuvant therapy. These drugs can largely reverse the suppression of the immune system caused by the tumor microenvironment, allowing the reactivation of anti-tumor immune infiltrating cells, significantly improving the patient's tumor microenvironment, and thus preventing tumor development. However, there are still some patients who respond poorly to neoadjuvant combined immunotherapy and cannot achieve the expected results. It is now found that exploring changes in the tumor microenvironment not only elucidates patient responsiveness to immunotherapy and identifies more reliable biomarkers, but also addresses the limitations of prediction with imaging examination such as CT and the instability of existing biomarkers. In light of these considerations, this review aims to delve into the alterations within the tumor microenvironment and identify potential predictive biomarkers ensuing from neoadjuvant immunotherapy in the context of esophageal squamous cell carcinoma.
Collapse
Affiliation(s)
- Zhengzhou Qiu
- Jiangxi Medical College, Nanchang University, NanChang, China
- Department of Thoracic Surgery, Jiangxi Cancer Hospital, The Second Affiliated Hospital of Nanchang Medical College, Jiangxi Cancer Institute, Nanchang, China
- Jiangxi Key Laboratory of Oncology, Jiangxi Cancer Hospital, Nanchang, Jiangxi, China
| | - Zhao Li
- Department of Thoracic Surgery, Jiangxi Cancer Hospital, The Second Affiliated Hospital of Nanchang Medical College, Jiangxi Cancer Institute, Nanchang, China
| | - Xingfei Liu
- Jiangxi Medical College, Nanchang University, NanChang, China
- Department of Thoracic Surgery, Jiangxi Cancer Hospital, The Second Affiliated Hospital of Nanchang Medical College, Jiangxi Cancer Institute, Nanchang, China
| | - Ruilin Zhang
- Jiangxi Medical College, Nanchang University, NanChang, China
- Department of Thoracic Surgery, Jiangxi Cancer Hospital, The Second Affiliated Hospital of Nanchang Medical College, Jiangxi Cancer Institute, Nanchang, China
| | - Yongxuan Li
- Jiangxi Medical College, Nanchang University, NanChang, China
- Department of Thoracic Surgery, Jiangxi Cancer Hospital, The Second Affiliated Hospital of Nanchang Medical College, Jiangxi Cancer Institute, Nanchang, China
| | - Chenggen Gao
- Jiangxi Medical College, Nanchang University, NanChang, China
- Department of Thoracic Surgery, Jiangxi Cancer Hospital, The Second Affiliated Hospital of Nanchang Medical College, Jiangxi Cancer Institute, Nanchang, China
- Jiangxi Key Laboratory of Oncology, Jiangxi Cancer Hospital, Nanchang, Jiangxi, China
| | - Xiaoling Mao
- Jiangxi Key Laboratory of Oncology, Jiangxi Cancer Hospital, Nanchang, Jiangxi, China
- Medical College, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, China
| | - Yin Bao
- Jiangxi Key Laboratory of Oncology, Jiangxi Cancer Hospital, Nanchang, Jiangxi, China
- Medical College, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, China
| | - Mingyue Zhang
- Jiangxi Key Laboratory of Oncology, Jiangxi Cancer Hospital, Nanchang, Jiangxi, China
- Medical College, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, China
| | - Changying Guo
- Department of Thoracic Surgery, Jiangxi Cancer Hospital, The Second Affiliated Hospital of Nanchang Medical College, Jiangxi Cancer Institute, Nanchang, China
- Zhejiang-Jiangxi Joint Thoracic Oncology Research Laboratory, Jiangxi Cancer Hospital, Nanchang, Jiangxi, China
| |
Collapse
|
2
|
Wang J, Wang B, Guo H. Research progress and prospect of immunotherapy in locally advanced esophageal cancer. Asian J Surg 2024; 47:3096-3097. [PMID: 38438291 DOI: 10.1016/j.asjsur.2024.02.131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Accepted: 02/22/2024] [Indexed: 03/06/2024] Open
Affiliation(s)
- Jianbo Wang
- Department of Thoracic and Cardiac Surgery, Sichuan Tianfu New Area People's Hospital, Chengdu, Sichuan Province, China
| | - Bo Wang
- Department of Thoracic and Cardiac Surgery, Sichuan Tianfu New Area People's Hospital, Chengdu, Sichuan Province, China
| | - Hai Guo
- Department of Thoracic and Cardiac Surgery, Sichuan Tianfu New Area People's Hospital, Chengdu, Sichuan Province, China.
| |
Collapse
|
3
|
Sweeney R, Omstead AN, Fitzpatrick JT, Zheng P, Gorbunova A, Grayhack EE, Goel A, Khan AF, Kosovec JE, Wagner PL, Jobe BA, Kelly RJ, Zaidi AH. Sitravatinib combined with PD-1 blockade enhances cytotoxic T-cell infiltration by M2 to M1 tumor macrophage repolarization in esophageal adenocarcinoma. Carcinogenesis 2024; 45:210-219. [PMID: 38019590 DOI: 10.1093/carcin/bgad087] [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: 03/09/2023] [Revised: 11/06/2023] [Accepted: 11/27/2023] [Indexed: 12/01/2023] Open
Abstract
Esophageal adenocarcinoma (EAC) is a leading cause of cancer-related mortality. Sitravatinib is a novel multi-gene tyrosine kinase inhibitor (TKI) that targets tumor-associated macrophage (TAM) receptors, VEGF, PDGF and c-Kit. Currently, sitravatinib is actively being studied in clinical trials across solid tumors and other TKIs have shown efficacy in combination with immune checkpoint inhibitors (ICI) in cancer models. In this study, we investigated the anti-tumor activity of sitravatinib alone and in combination with PD-1 blockade in an EAC rat model. Treatment response was evaluated by mortality, pre- and post-treatment MRI, gene expression, immunofluorescence and immunohistochemistry. Our results demonstrated adequate safety and significant tumor shrinkage in animals treated with sitravatinib, and more profoundly, sitravatinib and PD-1 inhibitor, AUNP-12 (P < 0.01). Suppression of TAM receptors resulted in increased gene expression of pro-inflammatory cytokines and decreased expression of anti-inflammatory cytokines, enhanced infiltration of CD8+ T cells, and M2 to M1 macrophage phenotype repolarization in the tumor microenvironment of treated animals (P < 0.01). Moreover, endpoint immunohistochemistry staining corroborated the anti-tumor activity by downregulation of Ki67 and upregulation of Caspase-3 in the treated animals. Additionally, pretreatment gene expression of TAM receptors and PD-L1 were significantly higher in major responders compared with the non-responders, in animals that received sitravatinib and AUNP-12 (P < 0.02), confirming that TAM suppression enhances the efficacy of PD-1 blockade. In conclusion, this study proposes a promising immunomodulatory strategy using a multi-gene TKI to overcome developed resistance to an ICI in EAC, establishing rationale for future clinical development.
Collapse
Affiliation(s)
- Ryan Sweeney
- Allegheny Health Network, Allegheny Health Network Cancer Institute, Pittsburgh, PA, USA
| | - Ashten N Omstead
- Allegheny Health Network, Allegheny Health Network Cancer Institute, Pittsburgh, PA, USA
| | - John T Fitzpatrick
- Allegheny Health Network, Allegheny Health Network Cancer Institute, Pittsburgh, PA, USA
| | - Ping Zheng
- Allegheny Health Network, Allegheny Health Network Cancer Institute, Pittsburgh, PA, USA
| | - Anastasia Gorbunova
- Allegheny Health Network, Allegheny Health Network Cancer Institute, Pittsburgh, PA, USA
| | - Erin E Grayhack
- Allegheny Health Network, Allegheny Health Network Cancer Institute, Pittsburgh, PA, USA
| | - Arul Goel
- University of California Santa Barbara, Santa Barbara, CA, USA
| | - Alisha F Khan
- Allegheny Health Network, Allegheny Health Network Cancer Institute, Pittsburgh, PA, USA
| | | | - Patrick L Wagner
- Allegheny Health Network, Allegheny Health Network Cancer Institute, Pittsburgh, PA, USA
| | - Blair A Jobe
- Allegheny Health Network, Esophageal Institute, Pittsburgh, PA, USA
| | - Ronan J Kelly
- Baylor University Medical Center at Dallas, Charles A. Sammons Cancer Center, Dallas, TX, USA
| | - Ali H Zaidi
- Allegheny Health Network, Allegheny Health Network Cancer Institute, Pittsburgh, PA, USA
| |
Collapse
|
4
|
Khateeb S, Cavalcante L, Alnairat N, Singh M, Sahin IH, Saeed A, Saeed A. Who Should Receive Immunotherapy for Advanced Gastroesophageal Cancer? Curr Treat Options Oncol 2024; 25:496-509. [PMID: 38372852 DOI: 10.1007/s11864-024-01189-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/05/2024] [Indexed: 02/20/2024]
Abstract
OPINION STATEMENT This paper shines a light on the exciting progress being made in using immunotherapy to treat advanced gastroesophageal cancers. The positive results from trials using drugs like Pembrolizumab and Nivolumab are certainly encouraging and open new possibilities for treating this challenging disease. However, it is clear that we still have a lot to learn about how to predict which patients will benefit most from these treatments. The exploration of combining therapies and using machine learning to guide treatment shows promise. Moving forward, it is crucial that researchers and healthcare professionals continue to work together, sharing knowledge and findings to continue the advancements in this important area.
Collapse
Affiliation(s)
- Suhaib Khateeb
- Department of Medicine, Division of Hematology & Oncology, University of Pittsburgh Medical Center (UPMC), Pittsburgh, PA, USA
| | | | - Noor Alnairat
- Department of Internal Medicine, AL Abdali Hospital, Amman, Jordan
| | - Meghana Singh
- Department of Medicine, Division of Hematology & Oncology, University of Pittsburgh Medical Center (UPMC), Pittsburgh, PA, USA
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| | - Ibrahim Halil Sahin
- Department of Medicine, Division of Hematology & Oncology, University of Pittsburgh Medical Center (UPMC), Pittsburgh, PA, USA
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| | - Azhar Saeed
- Department of Pathology and Laboratory Medicine, University of Vermont Medical Center, Burlington, VT, USA
| | - Anwaar Saeed
- Department of Medicine, Division of Hematology & Oncology, University of Pittsburgh Medical Center (UPMC), Pittsburgh, PA, USA.
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA.
| |
Collapse
|
5
|
Li F, Wang W, Lai G, Lan S, Lv L, Wang S, Liu X, Zheng J. Development and validation of a novel lysosome-related LncRNA signature for predicting prognosis and the immune landscape features in colon cancer. Sci Rep 2024; 14:622. [PMID: 38182713 PMCID: PMC10770065 DOI: 10.1038/s41598-023-51126-9] [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: 10/11/2023] [Accepted: 12/31/2023] [Indexed: 01/07/2024] Open
Abstract
Lysosomes are essential components for managing tumor microenvironment and regulating tumor growth. Moreover, recent studies have also demonstrated that long non-coding RNAs could be used as a clinical biomarker for diagnosis and treatment of colorectal cancer. However, the influence of lysosome-related lncRNA (LRLs) on the progression of colon cancer is still unclear. This study aimed to identify a prognostic LRL signature in colon cancer and elucidated potential biological function. Herein, 10 differential expressed lysosome-related genes were obtained by the TCGA database and ultimately 4 prognostic LRLs for conducting a risk model were identified by the co-expression, univariate cox, least absolute shrinkage and selection operator analyses. Kaplan-Meier analysis, principal-component analysis, functional enrichment annotation, and nomogram were used to verify the risk model. Besides, the association between the prognostic model and immune infiltration, chemotherapeutic drugs sensitivity were also discussed in this study. This risk model based on the LRLs may be promising for potential clinical prognosis and immunotherapeutic responses related indicator in colon cancer patients.
Collapse
Affiliation(s)
- Fengming Li
- Center of Digestive Endoscopy, Longyan First Affiliated Hospital of Fujian Medical University, Longyan, China
| | - Wenyi Wang
- Department of Medical Oncology, Xiamen Key Laboratory of Antitumor Drug Transformation Research, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen, China
| | - Guanbiao Lai
- Center of Digestive Endoscopy, Longyan First Affiliated Hospital of Fujian Medical University, Longyan, China
| | - Shiqian Lan
- Center of Digestive Endoscopy, Longyan First Affiliated Hospital of Fujian Medical University, Longyan, China
| | - Liyan Lv
- Center of Digestive Endoscopy, Longyan First Affiliated Hospital of Fujian Medical University, Longyan, China
| | - Shengjie Wang
- Department of Thyroid and Breast Surgery, Xiamen Humanity Hospital Fujian Medical University, Xiamen, Fujian, China.
| | - Xinli Liu
- Department of Medical Oncology, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China.
| | - Juqin Zheng
- Center of Digestive Endoscopy, Longyan First Affiliated Hospital of Fujian Medical University, Longyan, China.
| |
Collapse
|
6
|
Wu L, Gao Y, Xie S, Ye W, Uemura Y, Zhang R, Yu Y, Li J, Chen M, Wu Q, Cui P, Liu H, Mu S, Li Y, Wang L, Liu C, Li J, Zhang L, Jiao S, Zhang G, Liu T. The level of macrophage migration inhibitory factor is negatively correlated with the efficacy of PD-1 blockade immunotherapy combined with chemotherapy as a neoadjuvant therapy for esophageal squamous cell carcinoma. Transl Oncol 2023; 37:101775. [PMID: 37678132 PMCID: PMC10492201 DOI: 10.1016/j.tranon.2023.101775] [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: 02/14/2023] [Revised: 08/10/2023] [Accepted: 08/28/2023] [Indexed: 09/09/2023] Open
Abstract
PURPOSE This study aimed to screen biomarkers to predict the efficacy of programmed cell death 1 (PD-1) blockade immunotherapy combined with chemotherapy as neoadjuvant therapy for esophageal squamous cell carcinoma (ESCC). METHODS In the first stage of the study, the baseline concentrations of 40 tumor-related chemokines in the serum samples of 50 patients were measured to screen for possible biomarkers. We investigated whether the baseline concentration of the selected chemokine was related to the therapeutic outcomes and tumor microenvironment states of patients treated with the therapy. In the second stage, the reliability of the selected biomarkers was retested in 34 patients. RESULTS The baseline concentration of macrophage migration inhibitory factor (MIF) was negatively correlated with disease-free survival (DFS) and overall survival (OS) in patients treated with the therapy. In addition, a low baseline expression level of MIF is related to a better tumor microenvironment for the treatment of ESCC. A secondary finding was that effective treatment decreased the serum concentration of MIF. CONCLUSION Baseline MIF levels were negatively correlated with neoadjuvant therapy efficacy. Thus, MIF may serve as a predictive biomarker for this therapy. The accuracy of the prediction could be improved if the serum concentration of MIF is measured again after the patient received several weeks of treatment.
Collapse
Affiliation(s)
- Liangliang Wu
- Laboratory of Oncology, The First Medical Center of Chinese PLA General Hospital, Beijing, China; Institute of Oncology, Senior Department of Oncology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Yiming Gao
- Department of Oncology, The First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Shengzhi Xie
- Department of Oncology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Wan Ye
- Institute of Oncology, Senior Department of Oncology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Yasushi Uemura
- Division of Cancer Immunotherapy, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa 277-8577, Japan
| | - Rong Zhang
- Division of Cancer Immunotherapy, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa 277-8577, Japan
| | - Yanju Yu
- Institute of Oncology, Senior Department of Oncology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Jinfeng Li
- Institute of Oncology, Senior Department of Oncology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Man Chen
- Department of Laboratory Medicine, Hebei Yanda Lu Daopei Hospital, Lang Fang, China
| | - Qiyan Wu
- Laboratory of Oncology, The First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Pengfei Cui
- Department of Oncology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Hongyu Liu
- Department of Neurosurgery, Hainan Hospital of Chinese PLA General Hospital, Sanya, Hainan, China
| | - Shuai Mu
- Department of Oncology, The First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Yilan Li
- Institute of Oncology, Senior Department of Oncology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Lingxiong Wang
- Laboratory of Oncology, The First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Chunxi Liu
- Laboratory of Oncology, The First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Jiahui Li
- Institute of Oncology, Senior Department of Oncology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Lijun Zhang
- Laboratory of Oncology, The First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Shunchang Jiao
- Department of Oncology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China.
| | - Guoqing Zhang
- Department of Oncology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China.
| | - Tianyi Liu
- Institute of Oncology, Senior Department of Oncology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China.
| |
Collapse
|
7
|
Uchida S, Sugino T. ERBB2-Mutant Gastrointestinal Tumors Represent Heterogeneous Molecular Biology, Particularly in Microsatellite Instability, Tumor Mutation Burden, and Co-Mutated Genes: An In Silico Study. Curr Issues Mol Biol 2023; 45:7404-7416. [PMID: 37754252 PMCID: PMC10528499 DOI: 10.3390/cimb45090468] [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: 08/11/2023] [Revised: 09/09/2023] [Accepted: 09/10/2023] [Indexed: 09/28/2023] Open
Abstract
During recent years, activating mutations in ERBB2 have been reported in solid tumors of various organs, and clinical trials targeting ERBB2-mutant tumors have been conducted. However, no effective treatment has been established for gastrointestinal tumors targeting ERBB2 mutations. ERBB2-mutant tumors have a higher tumor mutation burden (TMB) and microsatellite instability (MSI) than ERBB2 non-mutant tumors, but not all ERBB2-mutant tumors are TMB- and MSI-high. Thus, a more detailed classification of ERBB2-mutant tumors based on the underlying molecular mechanisms is required. Herein, we classified ERBB2 mutations into three groups-group 1: both ERBB2 mutations and amplifications; group 2: ERBB2 mutations annotated as putative driver mutations but without amplifications; group 3: ERBB2 mutations annotated as non-driver mutations (passenger mutations or unknown significance) and those that were not amplified in gastrointestinal tumors. Esophageal adenocarcinoma, gastric cancer, and colorectal cancer presented significantly higher MSI and TMB in the ERBB2-mutant group than in the ERBB2-wild-type group. The proportions of TMB- and MSI-high tumors and frequency of co-mutated downstream genes differed among the groups. We identified TMB- and MSI-high groups; this classification is considered important for guiding the selection of drugs for ERBB2-mutant tumors with downstream genetic mutations.
Collapse
Affiliation(s)
- Shiro Uchida
- Division of Diagnostic Pathology, Kikuna Memorial Hospital, 4-4-27, Kikuna, Kohoku-ku, Yokohama 222-0011, Japan
- Division of Pathology, Shizuoka Cancer Center, Shizuoka 411-8777, Japan;
- Department of Human Pathology, Juntendo University School of Medicine, Tokyo 113-8421, Japan
| | - Takashi Sugino
- Division of Pathology, Shizuoka Cancer Center, Shizuoka 411-8777, Japan;
| |
Collapse
|
8
|
Jiang C, Fan F, Xu W, Jiang X. POLD4 Promotes Glioma Cell Proliferation and Suppressive Immune Microenvironment: A Pan-Cancer Analysis Integrated with Experimental Validation. Int J Mol Sci 2023; 24:13919. [PMID: 37762224 PMCID: PMC10530695 DOI: 10.3390/ijms241813919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Revised: 09/05/2023] [Accepted: 09/07/2023] [Indexed: 09/29/2023] Open
Abstract
POLD4 plays a crucial part in the complex machinery of DNA replication and repair as a vital component of the DNA polymerase delta complex. In this research, we obtained original information from various publicly available databases. Using a blend of R programming and internet resources, we initiated an extensive examination into the correlation between POLD4 expression and the various elements of cancers. In addition, we performed knockdown experiments in glioma cell lines to authenticate its significant impact. We discovered that POLD4 is upregulated in various malignant tumors, demonstrating a significant correlation with poor patient survival prognosis. Using function analysis, it was uncovered that POLD4 exhibited intricate associations with signaling pathways spanning multiple tumor types. Subsequent investigations unveiled the close association of POLD4 with the immune microenvironment and the effectiveness of immunotherapy. Drugs like trametinib, saracatinib, and dasatinib may be used in patients with high POLD4. Using experimental analysis, we further confirmed the overexpression of POLD4 in gliomas, as well as its correlation with glioma recurrence, proliferation, and the suppressive immune microenvironment. Our research findings indicate that the expression pattern of POLD4 not only serves as a robust indicator of prognosis in cancer patients but also holds promising potential as a new focus for treatment.
Collapse
Affiliation(s)
| | | | | | - Xiaobing Jiang
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China (W.X.)
| |
Collapse
|
9
|
Dadgar N, Edlukudige Keshava V, Raj MS, Wagner PL. The Influence of the Microbiome on Immunotherapy for Gastroesophageal Cancer. Cancers (Basel) 2023; 15:4426. [PMID: 37760397 PMCID: PMC10526145 DOI: 10.3390/cancers15184426] [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: 08/08/2023] [Revised: 08/27/2023] [Accepted: 08/30/2023] [Indexed: 09/29/2023] Open
Abstract
Immunotherapy has shown promise as a treatment option for gastroesophageal cancer, but its effectiveness is limited in many patients due to the immunosuppressive tumor microenvironment (TME) commonly found in gastrointestinal tumors. This paper explores the impact of the microbiome on the TME and immunotherapy outcomes in gastroesophageal cancer. The microbiome, comprising microorganisms within the gastrointestinal tract, as well as within malignant tissue, plays a crucial role in modulating immune responses and tumor development. Dysbiosis and reduced microbial diversity are associated with poor response rates and treatment resistance, while specific microbial profiles correlate with improved outcomes. Understanding the complex interactions between the microbiome, tumor biology, and immunotherapy is crucial for developing targeted interventions. Microbiome-based biomarkers may enable personalized treatment approaches and prediction of patient response. Interventions targeting the microbiome, such as microbiota-based therapeutics and dietary modifications, offer the potential for reshaping the gut microbiota and creating a favorable TME that enhances immunotherapy efficacy. Further research is needed to reveal the underlying mechanisms, and large-scale clinical trials will be required to validate the efficacy of microbiome-targeted interventions.
Collapse
Affiliation(s)
- Neda Dadgar
- Cole Eye Institute, Cleveland Clinic, Cleveland, OH 44106, USA;
| | | | - Moses S. Raj
- Allegheny Health Network Cancer Institute, Pittsburgh, PA 15224, USA; (V.E.K.); (M.S.R.)
| | - Patrick L. Wagner
- Allegheny Health Network Cancer Institute, Pittsburgh, PA 15224, USA; (V.E.K.); (M.S.R.)
| |
Collapse
|
10
|
Puhr HC, Reiter TJ, Preusser M, Prager GW, Ilhan-Mutlu A. Recent Advances in the Systemic Treatment of Localized Gastroesophageal Cancer. Cancers (Basel) 2023; 15:1900. [PMID: 36980786 PMCID: PMC10047169 DOI: 10.3390/cancers15061900] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 01/30/2023] [Accepted: 03/20/2023] [Indexed: 03/30/2023] Open
Abstract
The overall survival expectancy of localized gastroesophageal cancer patients still remains under 5 years despite advances in neoadjuvant and adjuvant treatment strategies in recent years. For almost a decade, immunotherapy has been successfully implemented as a first-line treatment for various oncological diseases in advanced stages. In the case of advanced gastroesophageal cancer, 2021 witnessed several approvals of immune checkpoint inhibitor therapies by different authorities. Although it is still a debate whether this treatment should be restricted to a certain subgroup of patients based on biomarker selection, immunotherapy agents are making remarkable steps in resectable settings as well. The Checkmate-577 study demonstrated significant benefits of nivolumab as an adjuvant treatment for resectable esophageal and gastroesophageal junction tumors and thereby obtained approvals both from U.S. American and European authorities. First results of further potential practice-changing clinical trials are expected in 2023, which might change the treatment armamentarium for resectable gastroesophageal cancers significantly. This review aims to demonstrate the advances of immunotherapy and targeted therapies in treatment of localized gastric, gastroesophageal junction and esophageal tumors and gives a short summary on promising ongoing clinical trials.
Collapse
Affiliation(s)
| | | | | | | | - Aysegül Ilhan-Mutlu
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| |
Collapse
|
11
|
Li N, Wan Z, Lu D, Chen R, Ye X. Long-term benefit of immunotherapy in a patient with squamous lung cancer exhibiting mismatch repair deficient/high microsatellite instability/high tumor mutational burden: A case report and literature review. Front Immunol 2023; 13:1088683. [PMID: 36703977 PMCID: PMC9871463 DOI: 10.3389/fimmu.2022.1088683] [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: 11/03/2022] [Accepted: 12/20/2022] [Indexed: 01/12/2023] Open
Abstract
Genetic mutations that render mismatch repair defective may result in microsatellite instability, which is common in colorectal carcinomas and gastric cancers as well as Lynch syndrome. Mismatch repair deficiency/high microsatellite instability (dMMR/MSI-H) predicts the tumor response to immune checkpoint inhibitors. However, few studies have evaluated the efficacy of immune checkpoint inhibitors in non-small cell lung cancer (NSCLC) patients with dMMR/MSI-H. In this work, we present a patient with advanced squamous lung cancer with dMMR/MSI-H and a high tumor mutational burden (TMB-H) who obtained a long-term benefit from immunotherapy. NSCLC patients with dMMR/MSI-H/TMB-H may thus benefit from immune checkpoint inhibitors.
Collapse
Affiliation(s)
- Na Li
- First Clinical Medical College, Guangzhou University of Traditional Chinese, Guangzhou, China
| | - Zixuan Wan
- First Clinical Medical College, Guangzhou University of Traditional Chinese, Guangzhou, China
| | - Dongyan Lu
- Department of Oncology, The First Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine, Guangzhou, China
| | - Ruilian Chen
- Department of Oncology, The First Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine, Guangzhou, China,*Correspondence: Ruilian Chen, ; Xiaowei Ye,
| | - Xiaowei Ye
- Department of Oncology, The First Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine, Guangzhou, China,*Correspondence: Ruilian Chen, ; Xiaowei Ye,
| |
Collapse
|
12
|
Feng J, Tang X, Song L, Zhou Z, Jiang Y, Huang Y. A telomerase regulation-related lncRNA signature predicts prognosis and immunotherapy response for gastric cancer. J Cancer Res Clin Oncol 2023; 149:135-146. [PMID: 36333566 DOI: 10.1007/s00432-022-04456-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Accepted: 10/27/2022] [Indexed: 11/07/2022]
Abstract
BACKGROUND Telomeres are involved in the development and progression of gastric cancer (GC). However, the association of telomerase regulation-related lncRNAs with prognosis and immunotherapy responsiveness in gastric cancer is unclear. METHODS This study systematically evaluated the relationship between lncRNAs co-expressed with 67 telomerase regulatory genes and gastric cancer prognosis. The risk scores of the samples were calculated based on telomerase regulation-related lncRNAs with prognostic value, and the samples were classified into high-/low-risk groups. The prognostic value of risk groups was then evaluated, a GC prognostic prediction model based on risk groups and clinical characteristics was established, and the prediction accuracy of the model was clarified by receiving operating characteristic (ROC) curves and calibration curves. Finally, the value of risk grouping in GC immunotherapy sensitivity was predicted by comparing MSI status and tumor mutation load between the high- and low-risk groups. RESULTS We identified 13 lncRNAs with prognostic value co-expressed with telomerase regulatory genes and observed that the prognosis of the low-risk group was significantly better than that of the high-risk group. Meanwhile, a GC overall survival (OS) prediction model based on risk grouping and clinical characteristics was developed, and ROC curves and calibration curves confirmed the good predictive ability of the model. In addition, the low-risk group exhibited a higher tumor mutation load and MSI-H, suggesting a possible benefit of immunotherapy. CONCLUSION We found that telomerase regulation-related lncRNAs have prognostic value in GC patients and contribute to the exploration of more effective immunotherapeutic strategies.
Collapse
Affiliation(s)
- Jinggao Feng
- Department of Gastrointestinal and Anorectal Surgery, The Central Hospital of Yongzhou, No. 151, Xiaoshui West Road, Lingling District, Yongzhou, 425100, Hunan Province, China.
| | - Xiayu Tang
- Department of Gastrointestinal and Anorectal Surgery, The Central Hospital of Yongzhou, No. 151, Xiaoshui West Road, Lingling District, Yongzhou, 425100, Hunan Province, China
| | - Liusong Song
- Department of Gastrointestinal and Anorectal Surgery, The Central Hospital of Yongzhou, No. 151, Xiaoshui West Road, Lingling District, Yongzhou, 425100, Hunan Province, China
| | - Zhipeng Zhou
- Department of Gastrointestinal and Anorectal Surgery, The Central Hospital of Yongzhou, No. 151, Xiaoshui West Road, Lingling District, Yongzhou, 425100, Hunan Province, China
| | - Yuan Jiang
- Department of Gastrointestinal and Anorectal Surgery, The Central Hospital of Yongzhou, No. 151, Xiaoshui West Road, Lingling District, Yongzhou, 425100, Hunan Province, China
| | - Yao Huang
- Department of Gastrointestinal and Anorectal Surgery, The Central Hospital of Yongzhou, No. 151, Xiaoshui West Road, Lingling District, Yongzhou, 425100, Hunan Province, China
| |
Collapse
|
13
|
Bai Z, Bai Y, Fang C, Chen W. Oxidative stress-related patterns determination for establishment of prognostic models, and characteristics of tumor microenvironment infiltration. Front Surg 2022; 9:1013794. [PMID: 36386530 PMCID: PMC9665876 DOI: 10.3389/fsurg.2022.1013794] [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: 08/07/2022] [Accepted: 10/14/2022] [Indexed: 12/02/2022] Open
Abstract
Oxidative stress-mediated excessive accumulation of ROS in the body destroys cell homeostasis and participates in various diseases. However, the relationship between oxidative stress-related genes (ORGs) and tumor microenvironment (TME) in gastric cancer remains poorly understood. For improving the treatment strategy of GC, it is necessary to explore the relationship among them. We describe the changes of ORGs in 732 gastric cancer samples from two data sets. The two different molecular subtypes revealed that the changes of ORGs were associated with clinical features, prognosis, and TME. Subsequently, the OE_score was related to RFS, as confirmed by the correlation between OE_score and TME, TMB, MSI, immunotherapy, stem cell analysis, chemotherapeutic drugs, etc. OE_score can be used as an independent predictive marker for the treatment and prognosis of gastric cancer. Further, a Norman diagram was established to improve clinical practicability. Our research showed a potential role of ORGs in clinical features, prognosis, and tumor microenvironment of gastric cancer. Our research findings broaden the understanding of gastric cancer ORGs as a potential target for individualized treatment of gastric cancer and a new direction to evaluate the prognosis.
Collapse
Affiliation(s)
- Zihao Bai
- Graduate Department, Shanxi Medical University, Taiyuan, China
| | - Yihua Bai
- Graduate Department, Shanxi Medical University, Taiyuan, China
| | - Changzhong Fang
- Graduate Department, Shanxi Medical University, Taiyuan, China
| | - Wenliang Chen
- Department of General Surgery, The 2nd Affiliated Hospital of Shanxi Medical University, Taiyuan, China,Correspondence: Wenliang Chen
| |
Collapse
|
14
|
Pancancer Analysis of Revealed TDO2 as a Biomarker of Prognosis and Immunotherapy. DISEASE MARKERS 2022; 2022:5447017. [PMID: 36118672 PMCID: PMC9481368 DOI: 10.1155/2022/5447017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 08/27/2022] [Indexed: 12/17/2022]
Abstract
Background Tryptophan 2,3-dioxygenase (TDO) encoded by TDO2, a rate-limiting enzyme in the kynurenine pathway, catabolizes tryptophan to kynurenine, evades immune surveillance, and promotes tumor growth. Although accumulating evidence suggests a crucial role of TDO2 during tumor formation and development, systematic evaluation of TDO2 across human cancers has rarely been reported. Methods To shed more light on the role of TDO2 in human cancer, we explored the expression profiles of TDO2 and identified its prognostic value in pancancer analysis through TCGA, CCLE, and GTEx databases. We further utilized TCGA data to evaluate the association between TDO2 and tumor immunological features, such as mismatch repair (MMR), tumor immune infiltration, immune checkpoint-related genes, tumor mutational burden (TMB), microsatellite instability (MSI), and DNA methyltransferase (DNMT). Results TDO2 exhibited different expression levels in various cancer cell lines. Frequently, TDO2 was detected to be highly expressed in the majority of cancers. In addition, high TDO2 expression was correlated with an unfavorable prognosis for patients in KIRP, LGG, TGCT, and UVM. Moreover, high TDO2 expression level positively correlated with higher immune infiltration, especially dendritic cells. Additionally, there is a close relationship between TDO2 and immune checkpoint-related gene markers, such as LAIR1, CD276, NRP1, CD80, and CD86. Finally, correlation analysis has demonstrated a high-correlation between TDO2 and TMB, MSI, MMR, and DNMT of multiple cancer types. Conclusion Therefore, our results suggest that TDO2 can function as a potential prognostic biomarker due to its role in tumor immunity regulation.
Collapse
|
15
|
Sarhadi VK, Armengol G. Molecular Biomarkers in Cancer. Biomolecules 2022; 12:1021. [PMID: 35892331 PMCID: PMC9331210 DOI: 10.3390/biom12081021] [Citation(s) in RCA: 154] [Impact Index Per Article: 51.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 07/18/2022] [Accepted: 07/20/2022] [Indexed: 11/17/2022] Open
Abstract
Molecular cancer biomarkers are any measurable molecular indicator of risk of cancer, occurrence of cancer, or patient outcome. They may include germline or somatic genetic variants, epigenetic signatures, transcriptional changes, and proteomic signatures. These indicators are based on biomolecules, such as nucleic acids and proteins, that can be detected in samples obtained from tissues through tumor biopsy or, more easily and non-invasively, from blood (or serum or plasma), saliva, buccal swabs, stool, urine, etc. Detection technologies have advanced tremendously over the last decades, including techniques such as next-generation sequencing, nanotechnology, or methods to study circulating tumor DNA/RNA or exosomes. Clinical applications of biomarkers are extensive. They can be used as tools for cancer risk assessment, screening and early detection of cancer, accurate diagnosis, patient prognosis, prediction of response to therapy, and cancer surveillance and monitoring response. Therefore, they can help to optimize making decisions in clinical practice. Moreover, precision oncology is needed for newly developed targeted therapies, as they are functional only in patients with specific cancer genetic mutations, and biomarkers are the tools used for the identification of these subsets of patients. Improvement in the field of cancer biomarkers is, however, needed to overcome the scientific challenge of developing new biomarkers with greater sensitivity, specificity, and positive predictive value.
Collapse
Affiliation(s)
- Virinder Kaur Sarhadi
- Department of Oral and Maxillofacial Diseases, Helsinki University Hospital and University of Helsinki, 00290 Helsinki, Finland;
| | - Gemma Armengol
- Department of Animal Biology, Plant Biology, and Ecology, Faculty of Biosciences, Universitat Autònoma de Barcelona, 08193 Barcelona, Catalonia, Spain
| |
Collapse
|
16
|
Vošmik M, Kopecký J, John S, Kubeček O, Lochman P, Banni AM, Hruška L, Sirák I. Combined Therapy of Locally Advanced Oesophageal and Gastro-Oesophageal Junction Adenocarcinomas: State of the Art and Aspects of Predictive Factors. Cancers (Basel) 2021; 13:4591. [PMID: 34572818 PMCID: PMC8469285 DOI: 10.3390/cancers13184591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Revised: 09/07/2021] [Accepted: 09/08/2021] [Indexed: 11/17/2022] Open
Abstract
The following main treatment approaches are currently used in locally advanced adenocarcinomas of the oesophagus and gastrooesophageal junction (GOJ): preoperative chemoradiotherapy and surgery, and perioperative chemotherapy and surgery. While preoperative chemoradiotherapy is used primarily in oesophageal tumours, perioperative chemotherapy is the treatment of choice in Western countries for gastric cancer. The optimal treatment strategy for GOJ adenocarcinoma is still not clear. In comparison to other malignancies, biomarkers are used as predictive factors in distal oesophageal and GOJ adenocarcinomas in a very limited way, and moreover, only in metastatic stages (e.g., HER2 status, or microsatellite instability status). The aim of the article is to provide an overview of current treatment options in locally advanced adenocarcinomas of oesophagus and GOJ based on the latest evidence, including the possible potential of predictive biomarkers in optimizing treatment.
Collapse
Affiliation(s)
- Milan Vošmik
- Department of Oncology and Radiotherapy, Charles University, Faculty of Medicine in Hradec Králové and University Hospital Hradec Králové, 500 05 Hradec Králové, Czech Republic; (J.K.); (S.J.); (O.K.); (A.M.B.); (L.H.); (I.S.)
| | - Jindřich Kopecký
- Department of Oncology and Radiotherapy, Charles University, Faculty of Medicine in Hradec Králové and University Hospital Hradec Králové, 500 05 Hradec Králové, Czech Republic; (J.K.); (S.J.); (O.K.); (A.M.B.); (L.H.); (I.S.)
| | - Stanislav John
- Department of Oncology and Radiotherapy, Charles University, Faculty of Medicine in Hradec Králové and University Hospital Hradec Králové, 500 05 Hradec Králové, Czech Republic; (J.K.); (S.J.); (O.K.); (A.M.B.); (L.H.); (I.S.)
| | - Ondřej Kubeček
- Department of Oncology and Radiotherapy, Charles University, Faculty of Medicine in Hradec Králové and University Hospital Hradec Králové, 500 05 Hradec Králové, Czech Republic; (J.K.); (S.J.); (O.K.); (A.M.B.); (L.H.); (I.S.)
| | - Petr Lochman
- Department of Surgery, Charles University, Faculty of Medicine in Hradec Králové and University Hospital Hradec Králové, 500 05 Hradec Králové, Czech Republic;
- Department of Field Surgery, Faculty of Military Health Sciences, University of Defence, 500 05 Hradec Králové, Czech Republic
| | - Aml Mustafa Banni
- Department of Oncology and Radiotherapy, Charles University, Faculty of Medicine in Hradec Králové and University Hospital Hradec Králové, 500 05 Hradec Králové, Czech Republic; (J.K.); (S.J.); (O.K.); (A.M.B.); (L.H.); (I.S.)
| | - Libor Hruška
- Department of Oncology and Radiotherapy, Charles University, Faculty of Medicine in Hradec Králové and University Hospital Hradec Králové, 500 05 Hradec Králové, Czech Republic; (J.K.); (S.J.); (O.K.); (A.M.B.); (L.H.); (I.S.)
| | - Igor Sirák
- Department of Oncology and Radiotherapy, Charles University, Faculty of Medicine in Hradec Králové and University Hospital Hradec Králové, 500 05 Hradec Králové, Czech Republic; (J.K.); (S.J.); (O.K.); (A.M.B.); (L.H.); (I.S.)
| |
Collapse
|