1
|
Guan J, Gong X, Zeng H, Zhang W, Qin Q, Gou H, Liu X, Song B. Gastrointestinal tumor personalized immunotherapy: an integrated analysis from molecular genetics to imaging biomarkers. Therap Adv Gastroenterol 2025; 18:17562848251333527. [PMID: 40297204 PMCID: PMC12035075 DOI: 10.1177/17562848251333527] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Accepted: 03/24/2025] [Indexed: 04/30/2025] Open
Abstract
The immunotherapy landscape for gastrointestinal (GI) tumors is rapidly evolving. There is an urgent need for reliable biomarkers capable of predicting treatment outcomes to optimize therapeutic strategies and enhance patient prognosis. This review presents a comprehensive overview of biomarkers associated with the immunotherapy response of GI tumors, covering advances in molecular genetics, histopathological markers, and imaging. Key molecular biomarkers, such as microsatellite instability, tumor mutational burden, and programmed death-ligand 1 expression, remain critical for identifying patients likely to benefit from immune checkpoint inhibitors. The significance of tumor-infiltrating lymphocytes, notably the CD8+ T cell to regulatory T cell ratio, as a predictor of immunotherapy response is explored. In addition, advanced imaging techniques, including computed tomography (CT), magnetic resonance imaging, and positron emission tomography-CT, facilitate the noninvasive evaluation of tumor biology and therapeutic response. By bridging molecular and imaging data, this integrated strategy enhances precision in patient selection, treatment monitoring, and adaptive therapy design. Future studies should aim to validate these biomarkers in larger, multicenter cohorts and focus on clinical translation to advance precision medicine in GI oncology.
Collapse
Affiliation(s)
- Jian Guan
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, China
- Department of Radiology, Sichuan Provincial Corps Hospital, Chinese People’s Armed Police Forces, Leshan, China
| | - Xiaoling Gong
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, China
| | - Hanjiang Zeng
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, China
| | - Wei Zhang
- Department of Radiology, Sichuan Provincial Corps Hospital, Chinese People’s Armed Police Forces, Leshan, China
| | - Qing Qin
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Hongfeng Gou
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Xijiao Liu
- Department of Radiology, West China Hospital, Sichuan University, No. 37, Guoxue Alley, Chengdu 610041, China
- Department of Radiology, Sanya People’s Hospital, Sanya, Hainan, China
| | - Bin Song
- Department of Radiology, West China Hospital, Sichuan University, No. 37, Guoxue Alley, Chengdu 610041, China
- Department of Radiology, Sanya People’s Hospital, Sanya, Hainan, China
| |
Collapse
|
2
|
Kravchuk D, Lebedeva A, Kuznetsova O, Kavun A, Taraskina A, Belova E, Grigoreva T, Veselovsky E, Mileyko V, Nikulin V, Nekrasova L, Tryakin A, Fedyanin M, Ivanov M. Dynamics of blood microsatellite instability (bMSI) burden predicts outcome of a patient treated with immune checkpoint inhibitors: a case report of hyperprogressive disease. Front Immunol 2025; 16:1492296. [PMID: 39975556 PMCID: PMC11836019 DOI: 10.3389/fimmu.2025.1492296] [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: 09/06/2024] [Accepted: 01/16/2025] [Indexed: 02/21/2025] Open
Abstract
Microsatellite instability (MSI) is a widely studied molecular signature, which is associated with long-term benefit in patients treated with immune checkpoint inhibitor therapy. This approach has been proven to be effective in the treatment of patients with MSI-positive colorectal cancer (CRC). Analysis of serial liquid biopsy samples allows to detect changes in the tumor in response to therapy. Typically, somatic mutations are used for tracing the dynamics of the tumor, and the assessment of DNA signatures such as MSI is not currently used for these purposes. Here, we describe a case of a MSI-positive CRC, who received nivolumab monotherapy. Sequential sampling of the patient's plasma demonstrated an increase in MSI burden (bMSI), which was found to correlate with the increase of driver mutation burden one month after starting nivolumab, and hyperprogressive disease. Thus, analysis of bMSI in liquid biopsy via NGS may be a promising method for timely assessment of the treatment effectiveness received by patients with MSI-positive CRC.
Collapse
Affiliation(s)
- Daria Kravchuk
- Moscow Multidisciplinary Clinical Center “Kommunarka” of the Department of Health of the City of Moscow, State Budgetary Institution of Healthcare, Moscow, Russia
| | - Alexandra Lebedeva
- R&D Department, OncoAtlas LLC, Moscow, Russia
- Institute for Personalized Oncology, Sechenov First Moscow State Medical University, Moscow, Russia
| | - Olesya Kuznetsova
- R&D Department, OncoAtlas LLC, Moscow, Russia
- N.N. Blokhin Russian Cancer Research Center, Moscow, Russia
| | | | | | - Ekaterina Belova
- R&D Department, OncoAtlas LLC, Moscow, Russia
- Institute for Personalized Oncology, Sechenov First Moscow State Medical University, Moscow, Russia
- Faculty of Physics, Lomonosov Moscow State University, Moscow, Russia
| | - Tatiana Grigoreva
- R&D Department, OncoAtlas LLC, Moscow, Russia
- Institute for Personalized Oncology, Sechenov First Moscow State Medical University, Moscow, Russia
| | | | - Vladislav Mileyko
- R&D Department, OncoAtlas LLC, Moscow, Russia
- Institute for Personalized Oncology, Sechenov First Moscow State Medical University, Moscow, Russia
| | | | - Lidia Nekrasova
- P. Hertsen Moscow Oncology Research Institute (MORI), Moscow, Russia
| | - Alexey Tryakin
- N.N. Blokhin Russian Cancer Research Center, Moscow, Russia
| | - Mikhail Fedyanin
- Moscow Multidisciplinary Clinical Center “Kommunarka” of the Department of Health of the City of Moscow, State Budgetary Institution of Healthcare, Moscow, Russia
- N.N. Blokhin Russian Cancer Research Center, Moscow, Russia
- Federal State Budgetary Institution “National Medical and Surgical Center named after N.I. Pirogov” of the Ministry of Health of the Russian Federation, Moscow, Russia
| | - Maxim Ivanov
- R&D Department, OncoAtlas LLC, Moscow, Russia
- Institute for Personalized Oncology, Sechenov First Moscow State Medical University, Moscow, Russia
| |
Collapse
|
3
|
Singer M, Valerin J, Zhang Z, Zhang Z, Dayyani F, Yaghmai V, Choi A, Imagawa D, Abi-Jaoudeh N. Promising Cellular Immunotherapy for Colorectal Cancer Using Classical Dendritic Cells and Natural Killer T Cells. Cells 2025; 14:166. [PMID: 39936958 PMCID: PMC11817869 DOI: 10.3390/cells14030166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 01/15/2025] [Accepted: 01/20/2025] [Indexed: 02/13/2025] Open
Abstract
Colorectal cancer (CRC) remains one of the leading causes of cancer-related morbidity and mortality around the world. Despite advances in surgery, chemotherapy, and targeted therapies, the prognosis for patients with metastatic or advanced CRC remains poor. Immunotherapies comprising immune checkpoint inhibitors showed disappointing responses in metastatic CRC (mCRC). However, cellular immunotherapy, specifically using classical dendritic cells (cDCs), may hold unique promise in immune recognition for CRC antigens. cDCs are substantial players in immune recognition and are instrumental in orchestrating innate and adaptive immune responses by processing and presenting tumor antigens to effector cells. Natural killer T (NKT) cells are insufficiently studied but unique effector cells because of their ability to bridge innate and adaptive immune reactions and the crosstalk with dendritic cells in cancer. This review explores the therapeutic potential of using both cDCs and NKT cells as a synergistic therapy in CRC, focusing on their biological roles, strategies for harnessing their capabilities, clinical applications, and the challenges within the tumor microenvironment. Both cDCs and NKT cells can be used as a new effective approach for cell-based therapies in cancers to provide a new hope for CRC patients that are challenging to treat.
Collapse
Affiliation(s)
- Mahmoud Singer
- Department of Radiological Sciences, School of Medicine, University of California, Irvine, CA 92617, USA
| | - Jennifer Valerin
- Department of Medicine, Chao Family Comprehensive Cancer Center, University of California, Irvine, CA 92697, USA (A.C.)
| | - Zhuoli Zhang
- Department of Radiological Sciences, School of Medicine, University of California, Irvine, CA 92617, USA
| | - Zigeng Zhang
- Department of Radiological Sciences, School of Medicine, University of California, Irvine, CA 92617, USA
| | - Farshid Dayyani
- Department of Medicine, Chao Family Comprehensive Cancer Center, University of California, Irvine, CA 92697, USA (A.C.)
| | - Vahid Yaghmai
- Department of Radiological Sciences, School of Medicine, University of California, Irvine, CA 92617, USA
| | - April Choi
- Department of Medicine, Chao Family Comprehensive Cancer Center, University of California, Irvine, CA 92697, USA (A.C.)
| | - David Imagawa
- Department of Surgery, University of California Irvine, Orange, CA 92697, USA
| | - Nadine Abi-Jaoudeh
- Department of Radiological Sciences, School of Medicine, University of California, Irvine, CA 92617, USA
| |
Collapse
|
4
|
Ji K, Jia H, Liu Z, Yu G, Wen R, Zhang T, Peng Z, Man W, Tian Y, Wang C, Ling Q, Zhang W, Zhou L, Liu M, Zhu B. New insight in immunotherapy and combine therapy in colorectal cancer. Front Cell Dev Biol 2025; 12:1453630. [PMID: 39839672 PMCID: PMC11747282 DOI: 10.3389/fcell.2024.1453630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Accepted: 12/18/2024] [Indexed: 01/23/2025] Open
Abstract
The advent of immune checkpoint inhibitors (ICIs) in colorectal cancer (CRC) treatment marks a major breakthrough. These therapies have proven safer and more effective than traditional radiotherapy and targeted treatments. Immunotherapies like pembrolizumab, nivolumab, and ipilimumab have pioneered new treatment avenues, potentially improving patient outcomes and quality of life. Additionally, advances in immunotherapy have prompted detailed research into CRC therapies, especially those integrating ICIs with conventional treatments, providing new hope for patients and shaping future research and practice. This review delves into the mechanisms of various ICIs and evaluates their therapeutic potential when combined with radiotherapy, chemotherapy, and targeted therapies in clinical settings. It also sheds light on the current application and research involving ICIs in CRC treatment.
Collapse
Affiliation(s)
- Kai Ji
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
- Department of Colorectal Surgery, Shanghai Changhai Hospital, Naval Medical University, Shanghai, China
| | - Hang Jia
- Department of Colorectal Surgery, Shanghai Changhai Hospital, Naval Medical University, Shanghai, China
| | - Zixuan Liu
- Department of Colorectal Surgery, Shanghai Changhai Hospital, Naval Medical University, Shanghai, China
| | - Guanyu Yu
- Department of Colorectal Surgery, Shanghai Changhai Hospital, Naval Medical University, Shanghai, China
| | - Rongbo Wen
- Department of Colorectal Surgery, Shanghai Changhai Hospital, Naval Medical University, Shanghai, China
| | - Tianshuai Zhang
- Department of Colorectal Surgery, Shanghai Changhai Hospital, Naval Medical University, Shanghai, China
| | - Zhiying Peng
- Department of Colorectal Surgery, Shanghai Changhai Hospital, Naval Medical University, Shanghai, China
| | - Wenjiang Man
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
- Department of Colorectal Surgery, Shanghai Changhai Hospital, Naval Medical University, Shanghai, China
| | - Yucheng Tian
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
- Department of Colorectal Surgery, Shanghai Changhai Hospital, Naval Medical University, Shanghai, China
| | - Can Wang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
- Department of Colorectal Surgery, Shanghai Changhai Hospital, Naval Medical University, Shanghai, China
| | - Qianlong Ling
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
| | - Wei Zhang
- Department of Colorectal Surgery, Shanghai Changhai Hospital, Naval Medical University, Shanghai, China
| | - Leqi Zhou
- Department of Colorectal Surgery, Shanghai Changhai Hospital, Naval Medical University, Shanghai, China
| | - Mulin Liu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
| | - Bing Zhu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
| |
Collapse
|
5
|
Li C, Mao Y, Liu Y, Hu J, Su C, Tan H, Hou X, Ou M. Machine learning-based integration develops a multiple programmed cell death signature for predicting the clinical outcome and drug sensitivity in colorectal cancer. Anticancer Drugs 2025; 36:1-18. [PMID: 39132895 DOI: 10.1097/cad.0000000000001654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/13/2024]
Abstract
Tumorigenesis and treatment are closely associated with various programmed cell death (PCD) patterns. However, the coregulatory role of multiple PCD patterns in colorectal cancer (CRC) remains unknown. In this study, we developed a multiple PCD index (MPCDI) based on 19 PCD patterns using two machine learning algorithms for risk stratification, prognostic prediction, construction of nomograms, immune cell infiltration analysis, and chemotherapeutic drug sensitivity analysis. As a result, in the TCGA-COAD, GSE17536, and GSE29621 cohorts, the MPCDI can effectively distinguished survival outcomes in CRC patients and served as an independent factor for CRC patients. We then explored the immune infiltration landscape in two groups using the nine algorithms and found more overall immune infiltration in the high-MPCDI group. TIDE scores suggested that the increased immune evasion potential and immune checkpoint inhibition therapy may be less effective in the high-MPCDI group. Immunophenoscores indicated that anti-PD1, anti-cytotoxic T-lymphocyte associated antigen 4 (anti-CTLA4), and anti-PD1-CTLA4 combination therapies are less effective in the high-MPCDI group. In addition, the high-MPCDI group was more sensitive to AZD1332, Foretinib, and IGF1R_3801, and insensitive to AZD3759, AZD5438, AZD6482, Erlotinib, GSK591, IAP_5620, and Picolinici-acid, which suggests that the MPCDI can guide drug selection for CRC patients. As a new clinical classifier, the MPCDI can more accurately distinguish CRC patients who benefit from immunotherapy and develop personalized treatment strategies for CRC patients.
Collapse
Affiliation(s)
- Chunhong Li
- Central Laboratory, The Second Affiliated Hospital of Guilin Medical University
- Guangxi Health Commission Key Laboratory of Glucose and Lipid Metabolism Disorders, The Second Affiliated Hospital of Guilin Medical University
| | - Yuhua Mao
- Department of Obstetrics, The Second Affiliated Hospital of Guilin Medical University
| | - Yi Liu
- Department of Obstetrics, The Second Affiliated Hospital of Guilin Medical University
| | - Jiahua Hu
- Central Laboratory, The Second Affiliated Hospital of Guilin Medical University
- Guangxi Health Commission Key Laboratory of Glucose and Lipid Metabolism Disorders, The Second Affiliated Hospital of Guilin Medical University
| | - Chunchun Su
- Department of Laboratory Medicine, The Second Affiliated Hospital of Guilin Medical University
| | - Haiyin Tan
- School of Medical Laboratory Medicine, Guilin Medical University, Guilin, China
| | - Xianliang Hou
- Central Laboratory, The Second Affiliated Hospital of Guilin Medical University
- Guangxi Health Commission Key Laboratory of Glucose and Lipid Metabolism Disorders, The Second Affiliated Hospital of Guilin Medical University
| | - Minglin Ou
- Central Laboratory, The Second Affiliated Hospital of Guilin Medical University
- Guangxi Health Commission Key Laboratory of Glucose and Lipid Metabolism Disorders, The Second Affiliated Hospital of Guilin Medical University
| |
Collapse
|
6
|
Li X, Zhao Y, Wei S, Dai Y, Yi C. Construction of a cuproptosis-tricarboxylic acid cycle-associated lncRNA model to predict the prognosis of non-small cell lung cancer. Transl Cancer Res 2024; 13:6807-6824. [PMID: 39816567 PMCID: PMC11729758 DOI: 10.21037/tcr-24-660] [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: 04/21/2024] [Accepted: 11/15/2024] [Indexed: 01/18/2025]
Abstract
Background In cuproptosis, excess copper ions induce cell death via fatty acylation in the tricarboxylic acid (TCA) cycle. However, the effects of cuproptosis-TCA-related long non-coding RNAs (lncRNAs) on the clinical prognosis of non-small cell lung cancer (NSCLC) and the associated tumor microenvironment remain unclear. The purpose of this study is to use cuproptosis-TCA related lncRNAs to predict the prognosis of NSCLC. Methods Molecular signature databases and cuproptosis-related publications were made use of identifying cuproptosis-TCA-related genes. They were identified based on Pearson correlation analysis. The prognostic features associated with these lncRNAs were evaluated using the absolute contraction and selection operator and a receiver operating characteristic curve analysis. Additionally, downstream functional enrichment and immunoinfiltration were analyzed to examine the immunotherapeutic responses of patients with NSCLC. Results Eleven cuproptosis-TCA-associated lncRNAs were identified. A high-risk group was compared with a low-risk group based on risk scores, and the high-risk group had a significantly lower overall survival (OS). We established a prognostic risk profile, and based on these characteristics and clinical staging, a nomogram was constructed. An analysis of functional enrichment revealed the involvement of pathways associated with cellular and humoral immunity and fatty acylation. Risk scores differed significantly based on immune cells and pathways (antigen-presenting cell co-stimulation). Moreover, TP53, TTN, and MUC16 mutation status were strongly associated with risk scores, with patients identified as having a higher risk of NSCLC being more responsive to immunotherapy. Conclusions Eleven cuproptosis-TCA-associated lncRNAs can be used to predict the prognosis of NSCLC patients, thereby providing a new theoretical basis for immunotherapy.
Collapse
Affiliation(s)
- Xiang Li
- Faculty of Medicine, Hunan University of Chinese Medicine, Changsha, China
| | - Yunlong Zhao
- Faculty of Medicine, Hunan University of Chinese Medicine, Changsha, China
| | - Shengjie Wei
- Faculty of Medicine, Hunan University of Chinese Medicine, Changsha, China
| | - Yuqing Dai
- Faculty of Medicine, Hunan University of Chinese Medicine, Changsha, China
| | - Chun Yi
- Department of Pathology, Faculty of Medicine, Hunan University of Chinese Medicine, Changsha, China
| |
Collapse
|
7
|
HUANG YONGJIAN, WANG JINZHOU, XU JIUHUA, RUAN NING. Remodeling tumor microenvironment using pH-sensitive biomimetic co-delivery of TRAIL/R848 liposomes against colorectal cancer. Oncol Res 2024; 32:1765-1776. [PMID: 39449815 PMCID: PMC11497182 DOI: 10.32604/or.2024.045564] [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: 08/31/2023] [Accepted: 01/11/2024] [Indexed: 10/26/2024] Open
Abstract
Background Despite significant advancements in the development of anticancer therapies over the past few decades, the clinical management of colorectal cancer remains a challenging task. This study aims to investigate the inhibitory effects of cancer-targeting liposomes against colorectal cancer. Materials and Methods Liposomes consisting of 3β-[N-(N', N'-dimethylamino ethane)carbamoyl]-cholesterol (DC-CHOL), cholesterol (CHOL), and dioleoylphosphatidylethanolamine (DOPE) at a molar ratio of 1:1:0.5 were created and used as carriers to deliver an apoptosis-inducing plasmid encoding the tumor necrosis factor-related apoptosis-inducing ligand (pTRAIL) gene, along with the toll-like receptor (TLR7) agonist Rsiquimod (R848). The rationale behind this design is that pTRAIL can trigger cancer cell apoptosis by activating the DR4/5 receptor, while R848 can stimulate the immune microenvironment. Results Experimental results demonstrated the synergistic effects of R848 and pTRAIL encapsulated by liposomes (RTL) in suppressing the proliferation of colorectal cancer cells. Moreover, further in vivo investigations revealed the strong anti-tumor efficacy of RTL in xenograft and orthotropic in situ models of colorectal cancer. Conclusions These findings collectively highlight the therapeutic potential of R848/pTRAIL-loaded liposomes in the treatment of colorectal cancer.
Collapse
Affiliation(s)
- YONGJIAN HUANG
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, China
| | - JINZHOU WANG
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, China
| | - JIUHUA XU
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, China
| | - NING RUAN
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, China
| |
Collapse
|
8
|
Saini KS, Somara S, Ko HC, Thatai P, Quintana A, Wallen ZD, Green MF, Mehrotra R, McGuigan S, Pang L, Das S, Yadav K, Neric D, Cantini L, Joshi C, Iwamoto K, Dubbewar S, Vidal L, Chico I, Severson E, Lorini L, Badve S, Bossi P. Biomarkers in head and neck squamous cell carcinoma: unraveling the path to precision immunotherapy. Front Oncol 2024; 14:1473706. [PMID: 39439946 PMCID: PMC11493772 DOI: 10.3389/fonc.2024.1473706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 09/09/2024] [Indexed: 10/25/2024] Open
Abstract
Recent strides in understanding the molecular underpinnings of head and neck cancers have sparked considerable interest in identifying precise biomarkers that can enhance prognostication and enable personalized treatment strategies. Immunotherapy has particularly revolutionized the therapeutic landscape for head and neck squamous cell carcinoma, offering new avenues for treatment. This review comprehensively examines the application and limitations of the established and emerging/novel biomarkers for head and neck squamous cell carcinoma. Established biomarkers, including well-characterized genetic mutations, protein expressions, and clinical factors, have been extensively studied and validated in clinical practice. Novel biomarkers identified through molecular analyses, including novel genetic alterations, immune-related markers, and molecular signatures, are currently being investigated and validated in preclinical and clinical settings. Biomarkers hold the potential to deepen our understanding of head and neck squamous cell carcinoma biology and guide therapeutic strategies. The evolving paradigm of predictive biomarkers facilitates the study of individual responses to specific treatments, including targeted therapy and immunotherapy.
Collapse
Affiliation(s)
- Kamal S. Saini
- Fortrea Inc., Durham, NC, United States
- Addenbrooke’s Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
| | | | | | | | | | | | | | | | | | | | - Soma Das
- Fortrea Inc., Durham, NC, United States
| | - Kavita Yadav
- George Institute for Global Health, New Delhi, India
| | | | | | | | | | | | | | | | | | - Luigi Lorini
- Medical Oncology and Hematology Unit, IRCCS Humanitas Cancer Centre, Milan, Italy
| | - Sunil Badve
- Emory University, Atlanta, GA, United States
| | - Paolo Bossi
- Medical Oncology and Hematology Unit, IRCCS Humanitas Cancer Centre, Milan, Italy
- Università degli Studi di Brescia, Brescia, Italy
| |
Collapse
|
9
|
Wu C, Qin W, Lu W, Lin J, Yang H, Li C, Mao Y. Unraveling the immune landscape of lung adenocarcinoma: insights for tailoring therapeutic approaches. Discov Oncol 2024; 15:470. [PMID: 39331252 PMCID: PMC11436577 DOI: 10.1007/s12672-024-01396-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 09/24/2024] [Indexed: 09/28/2024] Open
Abstract
Lung adenocarcinoma (LUAD), a prevalent type of non-small cell lung cancer (NSCLC), was known for its diversity and intricate tumor microenvironment (TME). Comprehending the interaction among human immune-related genes (IRGs) and the TME is vital in the creation of accurate predictive models and specific treatments. We created a risk score based on IRGs and designed a nomogram to predict the prognosis of LUAD accurately. This involved a thorough examination of TME and the infiltration of immune cells in both high-risk and low-risk LUAD groups. Furthermore, the examination of the association between characteristic genes (BIRC5 and BMP5) and immune cells, along with immune checkpoints in the TME, was also conducted. The findings of our research unveiled unique immune profiles and interactions among individuals in the high- and low-risk categories, which contribute to variations in prognosis. LUAD demonstrated significant associations between BIRC5, BMP5, immune cells, and checkpoints, suggesting their involvement in disease advancement and resistance to medication. Furthermore, by correlating our findings with a multidrug database, we identified specific LUAD patient subsets that might benefit from tailored treatments. Our study establishes a groundbreaking prognostic model for LUAD, which not only underscores the importance of the immune context in LUAD but also paves the way for advancing precision medicine strategies in this complex malignancy.
Collapse
Affiliation(s)
- Changjiang Wu
- Department of Intensive Care Unit, Suzhou Kowloon Hospital, Shanghai Jiao Tong University School of Medicine, Suzhou, 215028, Jiangsu, China
| | - Wangshang Qin
- Genetic and Metabolic Central Laboratory, Maternal and Child Health Hospital of Guangxi Zhuang Autonomous Region, Nanning, 530003, Guangxi, China
| | - Wenqiang Lu
- Department of Thoracic Surgery, Suzhou Kowloon Hospital, Shanghai Jiao Tong University School of Medicine, Suzhou, 215028, Jiangsu, China
| | - Jingyu Lin
- Department of Science & Education, Suzhou Kowloon Hospital, Shanghai Jiao Tong University School of Medicine, Suzhou, 215028, Jiangsu, China
| | - Hongwei Yang
- Department of Clinical Laboratory, Suzhou BOE Hospital, Suzhou, 215028, Jiangsu, China
| | - Chunhong Li
- Central Laboratory, The Second Affiliated Hospital of Guilin Medical University, Guilin, 541199, Guangxi, China.
- Guangxi Health Commission Key Laboratory of Glucose and Lipid Metabolism Disorders, The Second Affiliated Hospital of Guilin Medical University, Guilin, 541199, Guangxi, China.
| | - Yiming Mao
- Department of Thoracic Surgery, Suzhou Kowloon Hospital, Shanghai Jiao Tong University School of Medicine, Suzhou, 215028, Jiangsu, China.
| |
Collapse
|
10
|
Li Z, Zhao Y, Huang P, Wu Z, Ouyang D, Nyarko AO, Ai L, Zhang Z, Chang S. Pan-cancer analysis of the immunological and oncogenic roles of ATAD2 with verification in papillary thyroid carcinoma. Sci Rep 2024; 14:22263. [PMID: 39333272 PMCID: PMC11436736 DOI: 10.1038/s41598-024-73274-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 09/16/2024] [Indexed: 09/29/2024] Open
Abstract
ATAD2 (ATPase Family AAA Domain-Containing 2) is highly expressed across varies tumor types, yet its common roles in tumor progression and immune interaction remain unclear. We analyzed the expression and alteration profiles of ATAD2, along with its diagnostic and prognostic role in pan-cancer, utilizing TCGA, GTEx, CPTAC, HPA, and cBioPortal databases. Furthermore, we examined the relationship between ATAD2 and immune infiltration utilizing single-cell sequencing data and TCGA database. Additionally, the expression and oncogenic functions of ATAD2 were verified in papillary thyroid carcinoma (PTC) through MTT, wound-healing, transwell, and flow cytometry assays. Our results revealed significant overexpression of ATAD2 in most cancers, strongly associated with poor prognosis. Amplification was the most frequent alteration type of ATAD2, with its mutation correlating with improved overall survival. ATAD2 was positively correlated with multiple inhibitory immune checkpoints and closely associated with the immunosuppressive microenvironment, particularly in PTC. In vitro experiments demonstrated that ATAD2 promoted the proliferation, migration, and invasion of PTC cells by activating the PI3K-AKT pathway and modulating the G1/S cell cycle checkpoint. Collectively, ATAD2 holds promise as a biomarker for pan-cancer diagnosis and prognosis, as well as a predictor of immunotherapeutic responsiveness and a therapeutic target to enhance the efficacy of existing anti-tumor immune therapies.
Collapse
Affiliation(s)
- Zhecheng Li
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Yaxin Zhao
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Peng Huang
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Zhaoyi Wu
- Department of Thyroid and Breast Surgery, Hunan Provincial People's Hospital, The First Affiliated Hospital of Human Normal University, Changsha, 410008, China
| | - Dengjie Ouyang
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Adolphus Osei Nyarko
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Lei Ai
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Zhejia Zhang
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, 410008, China.
| | - Shi Chang
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, 410008, China.
| |
Collapse
|
11
|
Hyeon D, Kim Y, Hwang Y, Bae JM, Kang GH, Kim K. Deep learning-based histological predictions of chromosomal instability in colorectal cancer. Am J Cancer Res 2024; 14:4495-4505. [PMID: 39417190 PMCID: PMC11477831 DOI: 10.62347/jynd6488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 08/23/2024] [Indexed: 10/19/2024] Open
Abstract
Colorectal cancer (CRC) is a lethal malignancy and a leading cause of cancer-related mortality worldwide. Chromosomal instability (CIN) is a key driver of genomic instability in CRC and is characterized by aneuploidy and somatic copy-number alterations. This study aimed to predict CIN in CRC using histological data from whole slide images (WSIs). CRC samples from TCGA were analyzed, with tumor regions segmented into tiles and nuclei for feature extraction using convolutional neural network (CNN) and morphologic analysis. Binary classification models were developed to distinguish high and low aneuploidy scores (AS) based on slide-level features. The analysis included 313 patients with 315 WSIs, resulting in over 350,000 tumor tiles and nearly 2.7 million tumor cell nuclei. The ResNet18-SSL model, pre-trained on histopathological images, demonstrated superior accuracy in tile-based AS prediction, while DenseNet121 excelled in nucleus-based prediction. Combining CNN-based and morphological features enhanced the classification accuracy of nucleus-based predictions. Additionally, significant correlations were observed between morphological features and copy-number signatures. Unsupervised clustering of nuclear features revealed that distinct groups are significantly correlated with CIN and TP53 mutations. This study underscores the potential of histological features from WSIs to predict CIN in CRC samples. Nuclear feature analysis, combined with deep-learning techniques, offers a robust method for CIN prediction, highlighting the importance of further research into the relationships between histological and molecular phenotypes.
Collapse
Affiliation(s)
- Dongwoo Hyeon
- Institute of Biomedical Research, Seoul National University HospitalSeoul, South Korea
| | - Younghoon Kim
- Department of Pathology, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of KoreaSeoul, South Korea
| | - Yaeeun Hwang
- Department of Veterinary Medicine, Seoul National UniversitySeoul, South Korea
| | - Jeong Mo Bae
- Department of Pathology, Seoul National University HospitalSeoul, South Korea
| | - Gyeong Hoon Kang
- Department of Pathology, College of Medicine, Seoul National UniversitySeoul, South Korea
| | - Kwangsoo Kim
- Department of Transdisciplinary Medicine, Institute of Convergence Medicine with Innovative Technology, Seoul National University HospitalSeoul, South Korea
- Department of Medicine, Seoul National UniversitySeoul, South Korea
| |
Collapse
|
12
|
Schwarz S, Su Z, Krohn M, Löffler MW, Schlosser A, Linnebacher M. Peptide-stimulated T cells bypass immune checkpoint inhibitor resistance and eliminate autologous microsatellite instable colorectal cancer cells. NPJ Precis Oncol 2024; 8:163. [PMID: 39075115 PMCID: PMC11286882 DOI: 10.1038/s41698-024-00645-3] [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: 01/23/2024] [Accepted: 07/09/2024] [Indexed: 07/31/2024] Open
Abstract
Two hypermutated colon cancer cases with patient-derived cell lines, peripheral and tumor-infiltrating T cells available were selected for detailed investigation of immunological response.T cells co-cultured with autologous tumor cells showed only low levels of pro-inflammatory cytokines and failed at tumor recognition. Similarly, treatment of co-cultures with immune checkpoint inhibitors (ICI) did not boost antitumor immune responses. Since proteinase inhibitor 9 (PI-9) was detected in tumor cells, a specific inhibitor (PI-9i) was used in addition to ICI in T cell cytotoxicity testing. However, only pre-stimulation with tumor-specific peptides (cryptic and neoantigenic) significantly increased recognition and elimination of tumor cells by T cells independently of ICI or PI-9i.We showed, that ICI resistant tumor cells can be targeted by tumor-primed T cells and also demonstrated the superiority of tumor-naïve peripheral blood T cells compared to highly exhausted tumor-infiltrating T cells. Future precision immunotherapeutic approaches should include multimodal strategies to successfully induce durable anti-tumor immune responses.
Collapse
Affiliation(s)
- Sandra Schwarz
- Department of General Surgery, Molecular Oncology and Immunotherapy, University Medicine Rostock, Rostock, Germany
| | - Zhaoran Su
- Department of General Surgery, Molecular Oncology and Immunotherapy, University Medicine Rostock, Rostock, Germany
| | - Mathias Krohn
- Department of General Surgery, Molecular Oncology and Immunotherapy, University Medicine Rostock, Rostock, Germany
| | - Markus W Löffler
- Department of General, Visceral and Transplant Surgery, University Hospital Tübingen, Tübingen, Germany
- Institute of Immunology, University of Tübingen, Tübingen, Germany
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ) Partner Site Tübingen, Tübingen, Germany
- Cluster of Excellence iFIT (EXC2180) 'Image-Guided and Functionally Instructed Tumor Therapies', University of Tübingen, Tübingen, Germany
- Institute for Clinical and Experimental Transfusion Medicine, Medical Faculty, University Hospital Tübingen, Tübingen, Germany
| | - Andreas Schlosser
- Rudolf-Virchow Center, Center for Integrative and Translational Bioimaging, University of Würzburg, Würzburg, Germany
| | - Michael Linnebacher
- Department of General Surgery, Molecular Oncology and Immunotherapy, University Medicine Rostock, Rostock, Germany.
| |
Collapse
|
13
|
Ajab SM, Zoughbor SH, Labania LA, Östlundh LM, Orsud HS, Olanda MA, Alkaabi O, Alkuwaiti SH, Alnuaimi SM, Al Rasbi Z. Microbiota composition effect on immunotherapy outcomes in colorectal cancer patients: A systematic review. PLoS One 2024; 19:e0307639. [PMID: 39047017 PMCID: PMC11268651 DOI: 10.1371/journal.pone.0307639] [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: 03/12/2024] [Accepted: 07/09/2024] [Indexed: 07/27/2024] Open
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) have emerged as an effective treatment for colorectal cancer (CRC). Studies indicate that the composition of gut microbiota could potentially serve as a biomarker for predicting the clinical effectiveness of immune checkpoint inhibitors. METHODS Following PRISMA guidelines, the review was conducted after registering the protocol with PROSPERO. A comprehensive literature search was carried out across five databases: PubMed, Scopus, Web of Science, Embase, and Cochrane Library. Assessment tools from the National Institutes of Health (NIH) were used to gauge the quality of the studies. RESULTS A total of 5,132 papers were identified, and three studies and one conference abstract published between 2017-2022 met the inclusion criteria and were summarized in a descriptive synthesis table. These four studies were in accord with the following findings, four main phyla, Firmicutes, Bacteroidata, Actinobacteria, and Verrucomicrobiota were associated with CRC patients' clinical response toward ICIs treatment. Ruminococcaceae was predominantly related to CRC patients responding to therapy, while the Micrococcaceae family was more common among the non-responders. Bacterial taxa such as Faecalibacterium and Prevotellaceae were associated with better responses to ICIs and could be predictive biomarkers. The signature of fecal microbiota with Akkermansia muciniphila and Eubacterium rectale enrichment, and Rothia mucilaginosa depletion could independently predict better response to ICIs in patients with CRC. CONCLUSION The findings have brought attention to the notable differences in terms of richness and composition of microbiota between patients who responded positively to the treatment and those who did not. Bacterial species and families, such as Faecalibacterium, Bifidobacterium, Lachnospiraceae, Akkermansia sp., Ruminococcaceae, and Prevotellaceae, have consistently surfaced as potential indicators of immunotherapeutic responses. Furthermore, this review also emphasizes the need for additional comprehensive, multi-center studies with larger sample sizes to validate reported microbiota and expand our understanding of the role of gut microbiota in CRC ICIs therapy. PROSPERO ID: CRD42021277691.
Collapse
Affiliation(s)
- Suad Mohamed Ajab
- Institute of Public Health, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, Abu Dhabi, United Arab Emirates
| | - Sumaya Hasan Zoughbor
- Microbiology and Immunology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, Abu Dhabi, United Arab Emirates
| | - Lena Abdulbaset Labania
- Microbiology and Immunology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, Abu Dhabi, United Arab Emirates
| | | | - Hiba Salaheldin Orsud
- Microbiology and Immunology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, Abu Dhabi, United Arab Emirates
| | - Marie Antonette Olanda
- Microbiology and Immunology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, Abu Dhabi, United Arab Emirates
| | - Obaid Alkaabi
- Microbiology and Immunology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, Abu Dhabi, United Arab Emirates
| | - Shamma Hamad Alkuwaiti
- Microbiology and Immunology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, Abu Dhabi, United Arab Emirates
| | - Shaikha Mohammed Alnuaimi
- Microbiology and Immunology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, Abu Dhabi, United Arab Emirates
| | - Zakeya Al Rasbi
- Microbiology and Immunology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, Abu Dhabi, United Arab Emirates
| |
Collapse
|
14
|
Yu H, Liu Q, Wu K, Tang S. Biomarkers to predict efficacy of immune checkpoint inhibitors in colorectal cancer patients: a systematic review and meta-analysis. Clin Exp Med 2024; 24:143. [PMID: 38960935 PMCID: PMC11222262 DOI: 10.1007/s10238-024-01408-x] [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: 11/01/2023] [Accepted: 06/18/2024] [Indexed: 07/05/2024]
Abstract
Immune checkpoint inhibitors (ICIs) are approved to treat colorectal cancer (CRC) with mismatch-repair gene deficiency, but the response rate remains low. Value of current biomarkers to predict CRC patients' response to ICIs is unclear due to heterogeneous study designs and small sample sizes. Here, we aim to assess and quantify the magnitude of multiple biomarkers for predicting the efficacy of ICIs in CRC patients. We systematically searched MEDLINE, Embase, the Cochrane Library, and Web of Science databases (to June 2023) for clinical studies examining biomarkers for efficacy of ICIs in CRC patients. Random-effect models were performed for meta-analysis. We pooled odds ratio (OR) and hazard ratio (HR) with 95% confidence interval (CI) for biomarkers predicting response rate and survival. 36 studies with 1867 patients were included in systematic review. We found that a lower pre-treatment blood neutrophil-to-lymphocyte ratio (n=4, HR 0.37, 95%CI 0.21-0.67) predicts good prognosis, higher tumor mutation burden (n=10, OR 4.83, 95%CI 2.16-10.78) predicts response to ICIs, and liver metastasis (n=16, OR 0.32, 95%CI 0.16-0.63) indicates resistance to ICIs, especially when combined with VEGFR inhibitors. But the predictive value of tumor PD-L1 expression (n=9, OR 1.01, 95%CI 0.48-2.14) was insignificant in CRC. Blood neutrophil-to-lymphocyte ratio, tumor mutation burden, and liver metastasis, but not tumor PD-L1 expression, function as significant biomarkers to predict efficacy of ICIs in CRC patients. These findings help stratify CRC patients suitable for ICI treatments, improving efficacy of immunotherapy through precise patient management. (PROSPERO, CRD42022346716).
Collapse
Affiliation(s)
- Hang Yu
- Cancer Institute, Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, 270 Dong-An Road, Shanghai, 200032, People's Republic of China
| | - Qingquan Liu
- School of Clinical Medicine, Shanghai Medical College, Fudan University, Shanghai, 200032, People's Republic of China
| | - Keting Wu
- School of Clinical Medicine, Shanghai Medical College, Fudan University, Shanghai, 200032, People's Republic of China
| | - Shuang Tang
- Cancer Institute, Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, 270 Dong-An Road, Shanghai, 200032, People's Republic of China.
- Shanghai Key Laboratory of Radiation Oncology, Shanghai, 200032, People's Republic of China.
| |
Collapse
|
15
|
Dawood S, Natarajan V, Danchaivijitr P. Comprehensive molecular profiling identifies actionable biomarkers for patients from Thailand and the United Arab Emirates with advanced malignancies. Front Oncol 2024; 14:1374087. [PMID: 38800398 PMCID: PMC11116666 DOI: 10.3389/fonc.2024.1374087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Accepted: 04/03/2024] [Indexed: 05/29/2024] Open
Abstract
Background Comprehensive molecular profiling of tissue samples that can help guide therapy management is not widely available across the globe. Methods Comprehensive molecular profiling through Caris Molecular Intelligence involves the analysis of DNA through next-generation sequencing, chromogenic or fluorescent in situ hybridization, pyrosequencing, and copy number alterations; RNA through whole-transcriptome sequencing and multiplex PCR of RNA; and protein through immunohistochemistry. Results Here we describe the experience of molecular profiling of tumor tissue samples from patients diagnosed with advanced solid tumors and treated in two countries, the United Arab Emirates and Thailand. Tumor cancer cases submitted to Caris Life Sciences (Phoenix, Arizona, USA) for molecular profiling from the UAE and Thailand were retrospectively analyzed (data accessed between 2019 and 2020) for their molecular alterations and clinical biomarkers, without regard to ethnicity. A total of 451 samples from 35 distinct types of advanced cancers were examined for mutations, amplifications, overexpression, exon copy number alterations, microsatellite instability, deficient mismatch repair, tumor mutational burden, and fusions. Interrogating each step of the biological pathway, from DNA to RNA to distinct protein, identified an alteration with an associated therapy for 75% of these tumor samples. The most common alterations identified included elevated PDL-1 that can be targeted with an immune checkpoint inhibitors and amplification of HER2 for which a variety of anti HER2 therapies are available. Conclusion Comprehensive molecular profiling in patients with advanced malignancies can help optimize therapeutic management allowing for improved prognostic outcome.
Collapse
Affiliation(s)
- Shaheenah Dawood
- Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, United Arab Emirates
- Oncology Department, Mediclinic City Hospital, Dubai, United Arab Emirates
| | | | - Pongwut Danchaivijitr
- Division of Medical Oncology, Department of Medicine, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, Thailand
| |
Collapse
|
16
|
Taflin N, Sandow L, Thawani R, Ye S, Kardosh A, Corless CL, Chen EY. Survival in metastatic microsatellite-stable colorectal cancer correlated with tumor mutation burden and mutations identified by next-generation sequencing. J Gastrointest Oncol 2024; 15:681-688. [PMID: 38756628 PMCID: PMC11094494 DOI: 10.21037/jgo-23-809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 03/08/2024] [Indexed: 05/18/2024] Open
Abstract
Background Next-generation sequencing (NGS) identifies mutations and molecular abnormalities within tumors, including tumor mutation burden (TMB). If a solid tumor has high TMB, immune checkpoint inhibitors (ICIs) are approved as an option for treatment. Studies have been inconclusive regarding how effective ICI are in treating patients with colorectal cancer (CRC), and it is unclear if high TMB is a good prognostic marker for CRC. We collected data from NGS of CRC and correlated survival to both TMB and mutations of interest, as well as investigated the efficacy of ICI. Methods This was a retrospective cohort analysis at a single institution, collecting NGS data from January 2018 to December 2020 in patients with CRC who were microsatellite-stable (MSS), n=161. Demographics, clinical data, and results from NGS were collected, and a survival analysis looking at TMB and selected mutations of interest was performed. Patients who were treated with ICI were assessed in a descriptive subset analysis. Results Patients with CRC who were MSS and had high TMB trended towards worse survival [hazard ratio (HR) =1.38] though the result was not significant (P=0.28). Survival was significantly worse in patients with a KRAS mutation (HR =1.71, P=0.04) and/or a CDKN2A mutation (HR =4.45, P<0.001). In this study population, 12 patients with high TMB had treatment with ICI, with nine of these patients having shorter progression-free survival (PFS) between 0.7 and 4.1 months, and three patients having longer PFS of 26.3, 24.7, and 13.2 months. Conclusions High TMB in MSS CRC did not show statistical difference in outcome. Mutations in KRAS and/or CDKN2A correlated with worse prognosis. Some patients with MSS CRC and high TMB responded to ICI, though there is a need to identify a better biomarker to predict which patients will have a good response to ICI therapy.
Collapse
Affiliation(s)
- Nicholas Taflin
- Division of Oncology, Department of Internal Medicine, The University of Utah, Salt Lake City, UT, USA
| | - Lyndsey Sandow
- Department of Medicine, Oregon Health & Science University, Portland, OR, USA
| | - Rajat Thawani
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Chicago, IL, USA
| | - Shangyuan Ye
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
| | - Adel Kardosh
- Division of Hematology & Oncology, Department of Medicine, Oregon Health & Science University, Portland, OR, USA
| | | | - Emerson Y. Chen
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
- Division of Hematology & Oncology, Department of Medicine, Oregon Health & Science University, Portland, OR, USA
| |
Collapse
|
17
|
Johnson B, Morris V, Wang X, Dasari A, Raghav K, Shen JP, Lee MS, Huey R, Parseghian C, Willis J, Wolff R, Drusbosky LM, Overman MJ, Kopetz S. Comprehensive Landscape of BRAF Variant Classes, Clonalities, and Co-Mutations in Metastatic Colorectal Cancer Using ctDNA Profiling. Cancers (Basel) 2024; 16:737. [PMID: 38398128 PMCID: PMC10886949 DOI: 10.3390/cancers16040737] [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: 11/30/2023] [Revised: 01/30/2024] [Accepted: 02/07/2024] [Indexed: 02/25/2024] Open
Abstract
Although V600E accounts for the majority of the BRAF mutations in metastatic colorectal cancer (mCRC), non-V600 BRAF variants have been shown in recent years to represent a distinct molecular subtype. This study provides a comprehensive profile of BRAF variants in mCRC using a large genomic database of circulating tumor DNA (ctDNA) and analyzing clinical outcomes in a cohort of patients with atypical (non-V600) BRAF variants (aBRAF; class II, class III, unclassified). Overall, 1733 out of 14,742 mCRC patients in the ctDNA cohort had at least one BRAF variant. Patients with atypical BRAF variants tended to be younger and male. In contrast to BRAFV600E, BRAF class II and III variants and their co-occurrence with KRAS/NRAS mutations were increased at baseline and especially with those patients predicted to have prior anti-EGFR exposure. Our clinical cohort included 38 patients with atypical BRAF mCRC treated at a large academic referral center. While there were no survival differences between atypical BRAF classes, concurrent RAS mutations or liver involvement was associated with poorer prognosis. Notably, patients younger than 50 years of age had extremely poor survival. In these patients, the high-frequency KRAS/NRAS co-mutation and its correlation with poorer prognosis underlines the urgent need for novel therapeutic strategies. This study represents one of the most comprehensive characterizations to date of atypical BRAF variants, utilizing both ctDNA and clinical cohorts.
Collapse
Affiliation(s)
- Benny Johnson
- Department of Gastrointestinal Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Van Morris
- Department of Gastrointestinal Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Xuemei Wang
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Arvind Dasari
- Department of Gastrointestinal Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Kanwal Raghav
- Department of Gastrointestinal Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - John Paul Shen
- Department of Gastrointestinal Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Michael S. Lee
- Department of Gastrointestinal Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Ryan Huey
- Department of Gastrointestinal Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Christine Parseghian
- Department of Gastrointestinal Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jason Willis
- Department of Gastrointestinal Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Robert Wolff
- Department of Gastrointestinal Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | | | - Michael J. Overman
- Department of Gastrointestinal Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Scott Kopetz
- Department of Gastrointestinal Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
18
|
Rodrigo JP, Sánchez-Canteli M, Otero-Rosales M, Martínez-Camblor P, Hermida-Prado F, García-Pedrero JM. Tumor mutational burden predictability in head and neck squamous cell carcinoma patients treated with immunotherapy: systematic review and meta-analysis. J Transl Med 2024; 22:135. [PMID: 38311741 PMCID: PMC10840180 DOI: 10.1186/s12967-024-04937-x] [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: 11/20/2023] [Accepted: 01/27/2024] [Indexed: 02/06/2024] Open
Abstract
BACKGROUND Tumor mutational burden (TMB) has been demonstrated to predict the response to immune checkpoint inhibitors (ICIs) in various cancers. However, the role of TMB in head and neck squamous cell carcinoma (HNSCC) has not yet been specifically addressed. Since HNSCC patients exhibit a rather limited response to ICIs, there is an unmet need to develop predictive biomarkers to improve patient selection criteria and the clinical benefit of ICI treatment. METHODS We conducted a systematic review and meta-analysis according to Preferred Reporting Items for Systematic Reviews and Meta-analyses (PRISMA) reporting guidelines. HNSCC cohort studies were selected when TMB prior to ICI treatment was evaluated, TMB cutoff value was available, and the prognostic value of TMB was evaluated by time-to-event survival analysis. A total of 11 out of 1960 articles were analyzed, including 1200 HNSCC patients. RESULTS The results showed that those patients harboring high TMB exhibited a significantly superior overall response rate (OR = 2.62; 95% CI 1.74-3.94; p < 0.0001) and a survival advantage (HR = 0.53; 95% CI 0.39-0.71; p < 0.0001) after ICI treatment. CONCLUSION This is the first meta-analysis to demonstrate a higher response and clinical benefit from ICI therapy in HNSCC patients with high TMB.
Collapse
Affiliation(s)
- Juan P Rodrigo
- Department of Otolaryngology, Hospital Universitario Central de Asturias and Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Instituto Universitario de Oncología del Principado de Asturias, University of Oviedo, 33011, Oviedo, Spain
- Ciber de Cáncer, CIBERONC, 28029, Madrid, Spain
| | - Mario Sánchez-Canteli
- Ciber de Cáncer, CIBERONC, 28029, Madrid, Spain.
- Department of Otolaryngology, Hospital Universitario de Cabueñes and Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011, Oviedo, Spain.
| | - María Otero-Rosales
- Department of Otolaryngology, Hospital Universitario Central de Asturias and Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Instituto Universitario de Oncología del Principado de Asturias, University of Oviedo, 33011, Oviedo, Spain
- Ciber de Cáncer, CIBERONC, 28029, Madrid, Spain
| | - Pablo Martínez-Camblor
- Department of Anesthesiology, Dartmouth Hitchcock Medical Center, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
- Faculty of Health Sciences, Universidad Autonoma de Chile, 7500912, Providencia, Chile
| | - Francisco Hermida-Prado
- Department of Otolaryngology, Hospital Universitario Central de Asturias and Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Instituto Universitario de Oncología del Principado de Asturias, University of Oviedo, 33011, Oviedo, Spain
- Ciber de Cáncer, CIBERONC, 28029, Madrid, Spain
| | - Juana M García-Pedrero
- Department of Otolaryngology, Hospital Universitario Central de Asturias and Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Instituto Universitario de Oncología del Principado de Asturias, University of Oviedo, 33011, Oviedo, Spain
- Ciber de Cáncer, CIBERONC, 28029, Madrid, Spain
| |
Collapse
|
19
|
Ma C, Teng Q, Shang L, Du F, Li L. Tumor mutation load better predicts the prognosis of patients treated with immune checkpoint inhibitors in upper gastrointestinal cancers: A systematic review and meta-analysis. Cancer Rep (Hoboken) 2024; 7:e1959. [PMID: 38204354 PMCID: PMC10849990 DOI: 10.1002/cnr2.1959] [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: 08/10/2023] [Revised: 11/21/2023] [Accepted: 12/04/2023] [Indexed: 01/12/2024] Open
Abstract
BACKGROUND Tumor mutational load (TML) has emerged as a potential biomarker for multiple solid tumors. However, data on its prognostic impact on upper gastrointestinal (UGI) cancer are limited. Therefore, the aim of this systematic review and meta-analysis was to assess the prognostic value of TML for the survival of patients with UGI cancer. METHOD A comprehensive search of the PubMed, Embase, Cochrane Library, and Web of Science databases was conducted up to February 13, 2023. Eleven studies met our inclusion criteria. Hazard ratios (HRs) for progression-free survival and overall survival and their 95% confidence intervals (CIs) were calculated. Subsequently, the combined HR and its 95% CI were calculated for UGI tract cancers in the high and low TML groups. I2 statistics and p-values were used to evaluate heterogeneity. Publication bias, sensitivity, and subgroup analyses were performed to determine sources of heterogeneity. RESULTS In total, 932 patients with UGI tract cancer from 11 publications were included. The high TML group treated with immunotherapy showed significantly improved overall survival (HR = 0.68; 95% CI: 0.53, 0.86; p = .001) and progression-free survival (HR = 0.74; 95% CI: 0.58, 0.95; p = .020) compared with the low TML group. CONCLUSION Our study demonstrated that patients with UGI tumors and higher TML have a better prognosis with immunotherapy, suggesting that TML is a promising predictive biomarker for immunotherapy. REGISTRATION The study protocol was registered with the International Prospective Register of Systematic Reviews (PROSPERO Registration No: CRD42023405596).
Collapse
Affiliation(s)
- Chenghao Ma
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital, Cheeloo College of MedicineShandong UniversityJinanChina
| | - Qiong Teng
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital, Cheeloo College of MedicineShandong UniversityJinanChina
| | - Liang Shang
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital, Cheeloo College of MedicineShandong UniversityJinanChina
- Department of Gastrointestinal SurgeryShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanChina
| | - Fengying Du
- Department of Gastrointestinal SurgeryShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanChina
| | - Leping Li
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital, Cheeloo College of MedicineShandong UniversityJinanChina
- Department of Gastrointestinal SurgeryShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanChina
| |
Collapse
|
20
|
Xie T, Fu DJ, Li KJ, Guo JD, Xiao ZM, Li Z, Zhao SC. Identification of a basement membrane gene signature for predicting prognosis and estimating the tumor immune microenvironment in prostate cancer. Aging (Albany NY) 2024; 16:1581-1604. [PMID: 38240702 PMCID: PMC10866409 DOI: 10.18632/aging.205445] [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: 08/23/2023] [Accepted: 12/01/2023] [Indexed: 02/06/2024]
Abstract
Basement membrane plays an important role in tumor invasion and metastasis, which is closely related to prognosis. However, the prognostic value and biology of basement membrane genes (BMGs) in prostate cancer (PCa) remain unknown. In the TCGA training set, we used differentially expressed gene analysis, protein-protein interaction networks, univariate and multivariate Cox regression, and least absolute shrinkage and selection operator regression to construct a basement membrane-related risk model (BMRM) and validated its effectiveness in the MSKCC validation set. Furthermore, the accurate nomogram was constructed to improve clinical applicability. Patients with PCa were divided into high-risk and low-risk groups according to the optimal cut-off value of the basement membrane-related risk score (BMRS). It was found that BMRS was significantly associated with RFS, T-stage, Gleason score, and tumor microenvironmental characteristics in PCa patients. Further analysis showed that the model grouping was closely related to tumor immune microenvironment characteristics, immune checkpoint inhibitors, and chemotherapeutic drug sensitivity. In this study, we developed a new BMGs-based prognostic model to determine the prognostic value of BMGs in PCa. Finally, we confirmed that THBS2, a key gene in BMRM, may be an important link in the occurrence and progression of PCa. This study provides a novel perspective to assess the prognosis of PCa patients and provides clues for the selection of future personalized treatment regimens.
Collapse
Affiliation(s)
- Tao Xie
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
- Department of Urology, The Third Affiliated Hospital of Southern Medical University, Guangzhou 510500, China
| | - Du-Jiang Fu
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Kang-Jing Li
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Jia-Ding Guo
- Department of Urology, The Third Affiliated Hospital of Southern Medical University, Guangzhou 510500, China
| | - Zhao-Ming Xiao
- Department of Urology, The Third Affiliated Hospital of Southern Medical University, Guangzhou 510500, China
| | - Zhijie Li
- Department of Geriatric Medicine, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen 518020, China
| | - Shan-Chao Zhao
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
- Department of Urology, The Third Affiliated Hospital of Southern Medical University, Guangzhou 510500, China
| |
Collapse
|
21
|
Noori M, Jafari-Raddani F, Davoodi-Moghaddam Z, Delshad M, Safiri S, Bashash D. Immune checkpoint inhibitors in gastrointestinal malignancies: an Umbrella review. Cancer Cell Int 2024; 24:10. [PMID: 38183112 PMCID: PMC10771001 DOI: 10.1186/s12935-023-03183-3] [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: 08/19/2023] [Accepted: 12/17/2023] [Indexed: 01/07/2024] Open
Abstract
In the Modern era, immune checkpoint inhibitors (ICIs) have been the cornerstone of success in the treatment of several malignancies. Despite remarkable therapeutic advances, complex matrix together with significant molecular and immunological differences have led to conflicting outcomes of ICI therapy in gastrointestinal (GI) cancers. As far we are aware, to date, there has been no study to confirm the robustness of existing data, and this study is the first umbrella review to provide a more comprehensive picture about ICIs' efficacy and safety in GI malignancies. Systematic search on PubMed, Scopus, Web of Science, EMBASE, and Cochrane library identified 14 meta-analyses. The pooled analysis revealed that ICIs application, especially programmed death-1 (PD-1) inhibitors such as Camrelizumab and Sintilimab, could partially improve response rates in patients with GI cancers compared to conventional therapies. However, different GI cancer types did not experience the same efficacy; it seems that hepatocellular carcinoma (HCC) and esophageal cancer (EC) patients are likely better candidates for ICI therapy than GC and CRC patients. Furthermore, application of ICIs in a combined-modal strategy are perceived opportunity in GI cancers. We also assessed the correlation of PD-L1 expression as well as microsatellite status with the extent of the response to ICIs; overall, high expression of PD-L1 in GI cancers is associated with better response to ICIs, however, additional studies are required to precisely elaborate ICI responses with respect to microsatellite status in different GI tumors. Despite encouraging ICI efficacy in some GI cancers, a greater number of serious and fatal adverse events have been observed; further highlighting the fact that ICI therapy in GI cancers is not without cost, and further studies are required to utmost optimization of this approach in GI cancers.
Collapse
Affiliation(s)
- Maryam Noori
- Student Research Committee, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Farideh Jafari-Raddani
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zeinab Davoodi-Moghaddam
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahda Delshad
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Laboratory Sciences, School of Allied Medical Sciences, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Saeid Safiri
- Department of Community Medicine, Faculty of Medicine, Social Determinants of Health Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Davood Bashash
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
22
|
Zhu M, Benson AB. An update on pharmacotherapies for colorectal cancer: 2023 and beyond. Expert Opin Pharmacother 2024; 25:91-99. [PMID: 38224000 DOI: 10.1080/14656566.2024.2304654] [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: 10/03/2023] [Accepted: 01/09/2024] [Indexed: 01/16/2024]
Abstract
INTRODUCTION Colorectal cancer (CRC) is one of the most prevalent and lethal cancers worldwide. The treatment of metastatic colorectal cancer (mCRC) is difficult, and mCRC has a survival rate of only 13-17% compared with 70-90% in locoregional CRC. There is ongoing research effort on pharmacotherapy for CRC to improve the treatment outcome. AREAS COVERED We reviewed the current literature and ongoing clinical trials on CRC pharmacotherapy, with a focus on targeted therapy based on the results of genetic testing. The pharmacotherapies covered in this article include novel agents targeting EGFR and EGFR-related pathways, agents targeting the VEGF pathway, immunotherapy options depending on the MMR/MSI status, and new therapies targeting genetic fusions such as NTRK. We also briefly discuss the value of next-generation sequencing (NGS) in treatment selection and response monitoring. EXPERT OPINION We advocate for the early and routine use of NGS to genetically characterize CRC to assist with pharmacotherapy selection. Targeted therapy is a promising field of ongoing research and improves CRC treatment outcome.
Collapse
Affiliation(s)
- Mengou Zhu
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Al B Benson
- Department of Medicine, Division of Hematology and Oncology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Robert H. Lurie Comprehensive Cancer Center, Chicago, IL, USA
| |
Collapse
|
23
|
Li S, Zheng H, Ge Q, Xia S, Zhang K, Wang C, Wang F. Effectiveness and Safety of Apatinib Plus Programmed Cell Death Protein 1 Blockades for Patients with Treatment-refractory Metastatic Colorectal Cancer: A Retrospective Exploratory Study. J Cancer Prev 2023; 28:106-114. [PMID: 37830117 PMCID: PMC10564635 DOI: 10.15430/jcp.2023.28.3.106] [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: 02/27/2023] [Revised: 06/21/2023] [Accepted: 08/21/2023] [Indexed: 10/14/2023] Open
Abstract
This study aimed to investigate the efficacy and safety of apatinib plus programmed cell death protein 1 (PD-1) blockades for patients with metastatic colorectal cancer (CRC) who were refractory to the standard regimens. In this retrospective study, patients with metastatic CRC who received apatinib plus PD-1 blockades in clinical practice were included. The initial dosage of apatinib was 250 mg or 500 mg, and PD-1 blockades were comprised of camrelizumab, sintilimab and pembrolizumab. Efficacy and safety data were collected through the hospital's electronic medical record system. From October 2018 to March 2022, a total of 43 patients with metastatic CRC were evaluated for efficacy and safety. The results showed an objective response rate of 25.6% (95% CI, 13.5%-41.2%) and a disease control rate of 72.1% (95% CI, 56.3%-84.7%). The median progression-free survival (PFS) of the cohort was 5.8 months (95% CI, 3.81-7.79), and the median overall survival (OS) was 10.3 months (95% CI, 5.75-14.85). The most common adverse reactions were fatigue (76.7%), hypertension (72.1%), diarrhea (62.8%), and hand-foot syndrome (51.2%). Multivariate Cox regression analysis revealed that Eastern Cooperative Oncology Group (ECOG) performance status and location of CRC (left or right-side) were independent factors to predict PFS of patients with metastatic CRC treated with the combination regimen. Consequently, the combination of apatinib and PD-1 blockades demonstrated potential efficacy and acceptable safety for patients with treatment-refractory metastatic CRC. This conclusion should be confirmed in prospective clinical trials subsequently.
Collapse
Affiliation(s)
- Shenglong Li
- Department of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Hao Zheng
- Department of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Qinghong Ge
- Department of Internal Medicine, Harbin Orthopedics Hospital, Harbin, China
| | - Shuli Xia
- Department of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Ke Zhang
- Department of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Chunjing Wang
- Department of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Fujing Wang
- Department of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
24
|
Johnson RL, Ganesan S, Thangavelu A, Theophilou G, de Jong D, Hutson R, Nugent D, Broadhead T, Laios A, Cummings M, Orsi NM. Immune Checkpoint Inhibitors Targeting the PD-1/PD-L1 Pathway in Advanced, Recurrent Endometrial Cancer: A Scoping Review with SWOT Analysis. Cancers (Basel) 2023; 15:4632. [PMID: 37760602 PMCID: PMC10527181 DOI: 10.3390/cancers15184632] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 09/06/2023] [Accepted: 09/15/2023] [Indexed: 09/29/2023] Open
Abstract
Results of recent clinical trials using the immune check point inhibitors (ICI) pembrolizumab or dostarlimab with/without lenvatinib has led to their approval for specific molecular subgroups of advanced recurrent endometrial cancer (EC). Herein, we summarise the clinical data leading to this first tissue-agnostic approval. As this novel therapy is not yet available in the United Kingdom standard care setting, we explore the strengths, weaknesses, opportunities, and threats (SWOT) of ICI treatment in EC. Major databases were searched focusing on clinical trials using programmed cell death protein 1 (PD-1) and its ligand (PD-L1) ICI which ultimately contributed to anti-PD-1 approval in EC. We performed a data quality assessment, reviewing survival and safety analysis. We included 15 studies involving 1609 EC patients: 458 with mismatch repair deficiency (MMRd)/microsatellite instability-high (MSI-H) status and 1084 with mismatch repair proficiency/microsatellite stable (MMRp/MSS) status. Pembrolizumab/dostarlimab have been approved for MMRd ECs, with the addition of lenvatinib for MMRp cases in the recurrent setting. Future efforts will focus on the pathological assessment of biomarkers to determine molecular phenotypes that correlate with response or resistance to ICI in order to identify patients most likely to benefit from this treatment.
Collapse
Affiliation(s)
- Racheal Louise Johnson
- Department of Gynaecological Oncology, St James’s University Hospital, Leeds LS9 7TF, UK
| | - Subhasheenee Ganesan
- Department of Gynaecological Oncology, St James’s University Hospital, Leeds LS9 7TF, UK
| | - Amudha Thangavelu
- Department of Gynaecological Oncology, St James’s University Hospital, Leeds LS9 7TF, UK
| | - Georgios Theophilou
- Department of Gynaecological Oncology, St James’s University Hospital, Leeds LS9 7TF, UK
| | - Diederick de Jong
- Department of Gynaecological Oncology, St James’s University Hospital, Leeds LS9 7TF, UK
| | - Richard Hutson
- Department of Gynaecological Oncology, St James’s University Hospital, Leeds LS9 7TF, UK
| | - David Nugent
- Department of Gynaecological Oncology, St James’s University Hospital, Leeds LS9 7TF, UK
| | - Timothy Broadhead
- Department of Gynaecological Oncology, St James’s University Hospital, Leeds LS9 7TF, UK
| | - Alexandros Laios
- Department of Gynaecological Oncology, St James’s University Hospital, Leeds LS9 7TF, UK
| | - Michele Cummings
- Leeds Institute of Medical Research, St James’s University Hospital, The University of Leeds, Leeds LS9 7TF, UK
| | - Nicolas Michel Orsi
- Leeds Institute of Medical Research, St James’s University Hospital, The University of Leeds, Leeds LS9 7TF, UK
| |
Collapse
|
25
|
Wankhede D, Grover S, Hofman P. The prognostic value of TMB in early-stage non-small cell lung cancer: a systematic review and meta-analysis. Ther Adv Med Oncol 2023; 15:17588359231195199. [PMID: 37667779 PMCID: PMC10475237 DOI: 10.1177/17588359231195199] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 07/31/2023] [Indexed: 09/06/2023] Open
Abstract
Background Tumor mutation burden (TMB) has been validated as a predictive biomarker for immunotherapy response and survival in numerous cancer types. Limited data is available on the inherent prognostic role of TMB in early-stage tumors. Objective To evaluate the prognostic role of TMB in early-stage, resected non-small cell lung cancer (NSCLC). Design Systematic review and meta-analysis of pertinent prospective and retrospective studies. Data sources and methods Publication search was performed in PubMed, Embase, Cochrane Library, and Web of Science databases. Based on the level of heterogeneity, a random- or fixed-effects model was used to calculate pooled effects of hazard ratio (HR) for overall survival (OS) and disease-free survival (DFS). The source of heterogeneity was investigated using sensitivity analysis, subgroup analysis, and publication bias assessment. Results Ten studies comprising 2520 patients were included in this analysis. There was no statistically significant difference in OS (HR, 1.18, 95% CI, 0.70, 1.33; p 0.53, I2 = 80%; phet < 0.0001) and DFS (HR, 1.18, 95% CI, 0.91, 1.52; p = 0.53, I2 = 75%; phet = 0.0001) between the high-TMB and low-TMB group. Subgroup analyses indicated that East Asian ethnicity, and TMB detected using whole exome sequencing, and studies with <100 patients had poor DFS in the high-TMB group. Conclusion The inherent prognostic role of TMB is limited in early-stage NSCLC. Ethnic differences in mutation burden must be considered while designing future trials on neoadjuvant immunotherapy. Further research in the harmonization and standardization of panel-based TMB is essential for its widespread clinical utility.Registration: CRD42023392846.
Collapse
Affiliation(s)
- Durgesh Wankhede
- German Cancer Research Center, Im Neuenheimer Feld 580, 69120, Heidelberg, Germany
| | - Sandeep Grover
- Center for Human Genetics, Universitatsklinikum Giessen und Marburg – Standort Marburg, Marburg, Germany
| | - Paul Hofman
- Laboratory of Clinical and Experimental Pathology, Pasteur Hospital, University Côte d’Azur, Nice, France
- Institute for Research on Cancer and Ageing, Nice (IRCAN), INSERM U1081 and UMR CNRS 7284, Team 4, Nice, France
- Hospital-Integrated Biobank BB-0033-00025, Pasteur Hospital, Nice, France
- University Hospital Institute RespirERA, Nice, France
- University Hospital Federation OncoAge, CHU de Nice, University Côte d’Azur, Nice, France
| |
Collapse
|
26
|
Hou C, Wu X, Li C, Wang C, Liu J, Luo Q. A cuproptosis-associated long non-coding RNA signature for the prognosis and immunotherapy of lung squamous cell carcinoma. BIOMOLECULES & BIOMEDICINE 2023; 23:624-633. [PMID: 36724022 PMCID: PMC10351099 DOI: 10.17305/bb.2022.8481] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Revised: 01/04/2023] [Accepted: 01/04/2023] [Indexed: 06/18/2023]
Abstract
Cuproptosis, a copper-induced mechanism of mitochondrial-related cell death, has been implicated as a breakthrough in the treatment of cancer and has become a new treatment strategy. Furthermore, long non-coding RNA (lncRNA) can change the biological activities of tumor cells. Worldwide, lung squamous cell carcinoma (LUSC) is among the most common annoying tumors. LncRNAs related to cuproptosis are not researched at LUSC. Our research intends to develop a signature on the basis of cuproptosis-associated lncRNAs, which can predict LUSC prognosis and investigate LUSC immunological features. The TCGA database was used to retrieve LUSC transcriptome, clinical, and gene mutation data. For statistical analysis, we utilized the R program. We created a signature consisting of three cuproptosis-related lncRNAs in this investigation (including AC002467.1, LINC01740, and LINC02345). Survival analyses and Receiver Operating Characteristic curves demonstrated that this signature possessed powerful predictive capability. The signature's ability to predict was confirmed by a Receiver Operating Characteristic curve and principal component analysis. Notably, the patient with a high-risk score and a high tumor mutation burden level had a lower survival time. Furthermore, the tumor immune dysfunction and exclusion analysis showed that these individuals with low-risk scores may benefit from immunotherapy. The signature constructed by three cuproptosis-associated lncRNAs may be prognostic markers of LUSC. It contributes to immunotherapy and offers LUSC's therapy a new treatment direction.
Collapse
Affiliation(s)
- Chunlan Hou
- Department of Clinical Medicine, Southwest Medical University, Luzhou, Sichuan, China
| | - Xiuping Wu
- Department of Clinical Medicine, Southwest Medical University, Luzhou, Sichuan, China
| | - Caoyang Li
- Department of Clinical Medicine, Southwest Medical University, Luzhou, Sichuan, China
| | - Chao Wang
- Department of Cardiothoracic Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Jinbo Liu
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Qing Luo
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| |
Collapse
|
27
|
Li LL, Yu CF, Xie HT, Chen Z, Jia BH, Xie FY, Cai YF, Xue P, Zhu SJ. Biomarkers and factors in small cell lung cancer patients treated with immune checkpoint inhibitors: A meta-analysis. Cancer Med 2023. [PMID: 37161541 DOI: 10.1002/cam4.5800] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2023] [Revised: 02/18/2023] [Accepted: 02/25/2023] [Indexed: 05/11/2023] Open
Abstract
OBJECTIVE The aim of this meta-analysis was to summarize the available results of immunotherapy predictors for small cell lung cancer (SCLC) and to provide evidence-based information for their potential predictive value of efficacy. METHODS We searched PubMed, EMBASE, Web of Science, The Cochrane Library, and ClinicalTrials (from January 1, 1975 to November 1, 2021). The hazard ratios (HR) and its 95% confidence intervals (CIs) and tumor response rate of the included studies were extracted. RESULTS Eleven studies were eventually included and the pooled results showed that programmed cell death ligand 1 (PD-L1) positive: objective response rate (ORR) (relative risk [RR] = 1.39, 95% CI [0.48, 4.03], p = 0.54), with high heterogeneity (p = 0.05, I2 = 56%); disease control rate [DCR] (RR = 1.31, 95% CI [0.04, 38.57], p = 0.88), with high heterogeneity (p = 0.04, I2 = 75%); overall survival (OS) (HR = 0.89, 95% CI [0.74, 1.07], p = 0.22); and progression-free survival (PFS) (HR = 0.83, 95% CI [0.59, 1.16], p = 0.27), with high heterogeneity (p = 0.005, I2 = 73.1%). TMB-High (TMB-H): OS (HR = 0.86, 95% CI [0.74, 1.00], p = 0.05); PFS (HR = 0.71, 95% CI [0.6, 0.85], p < 0.001). Lactate dehydrogenase (LDH) >upper limit of normal (ULN): OS (HR = 0.95, 95% CI [0.81, 1.11], p = 0.511). Asian patients: OS (HR = 0.87, 95% CI [0.72, 1.04], p = 0.135); White/Non-Asian patients: OS (HR = 0.83, 95% CI [0.76, 0.90], p < 0.001). Liver metastasis patients: OS (HR = 0.93, 95% CI [0.83, 1.05], p = 0.229); PFS (HR = 0.84, 95% CI [0.67, 1.06], p = 0.141). Central nervous system (CNS) metastasis patients: OS (HR = 0.91, 95% CI [0.71, 1.17], p = 0.474); PFS (HR = 1.03, 95% CI [0.66, 1.60], p = 0.903). CONCLUSION The available research results do not support the recommendation of PD-L1 positive and TMB-H as predictors for the application of immune checkpoint inhibitors (ICIs) in SCLC patients. LDH, baseline liver metastasis and CNS metastasis may be used as markers/influencing factors for predicting the efficacy of ICIs in SCLC patients. Non-Asian SCLC patients had better efficacy with ICIs in our results.
Collapse
Affiliation(s)
- Lin-Lu Li
- Department of Oncology, Wangjing Hospital, China Academy of Chinese Medical Sciences, 100102, Beijing, China
- Graduate School, Beijing University of Chinese Medicine, 100029, Beijing, China
| | - Cheng-Feng Yu
- Department of Oncology, Wangjing Hospital, China Academy of Chinese Medical Sciences, 100102, Beijing, China
| | - Hong-Ting Xie
- Department of Oncology, Wangjing Hospital, China Academy of Chinese Medical Sciences, 100102, Beijing, China
- Graduate School, Beijing University of Chinese Medicine, 100029, Beijing, China
| | - Zheng Chen
- Department of Oncology, Wangjing Hospital, China Academy of Chinese Medical Sciences, 100102, Beijing, China
- Graduate School, Beijing University of Chinese Medicine, 100029, Beijing, China
| | - Bo-Hui Jia
- Beijing Sihui West District Hospital, 100082, Beijing, China
| | - Fei-Yu Xie
- Department of Oncology, Wangjing Hospital, China Academy of Chinese Medical Sciences, 100102, Beijing, China
- Graduate School, Beijing University of Chinese Medicine, 100029, Beijing, China
| | - Ya-Fang Cai
- Department of Oncology, Wangjing Hospital, China Academy of Chinese Medical Sciences, 100102, Beijing, China
- Graduate School, Beijing University of Chinese Medicine, 100029, Beijing, China
| | - Peng Xue
- Department of Oncology, Wangjing Hospital, China Academy of Chinese Medical Sciences, 100102, Beijing, China
| | - Shi-Jie Zhu
- Department of Oncology, Wangjing Hospital, China Academy of Chinese Medical Sciences, 100102, Beijing, China
| |
Collapse
|
28
|
Xu XT, Qian Y, Tian MX, Ding CC, Guo H, Tang J, Pi GL, Wu Y, Dai Z, Jin X. Predictive impact of prognostic nutritional index in patients with cancer treated with immune checkpoint inhibitors: a systematic review and meta-analysis. Nutr Cancer 2023:1-14. [PMID: 37140894 DOI: 10.1080/01635581.2023.2203355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
A few previous studies have investigated the prognostic value of the prognostic nutritional index (PNI) in patients treated with immune checkpoint inhibitors (ICIs); however, the results are inconsistent. Therefore, this study aimed to clarify the prognostic significance of PNI. The PubMed, Embase, and Cochrane Library databases were searched. A meta-analysis of the impact of PNI on overall survival (OS), progression-free survival (PFS), objective response rate (ORR), disease control rate (DCR), and rate of adverse events (AEs) in patients treated with ICIs was performed. Twenty-three studies involving 2,386 patients were included. Low PNI was associated with significantly poor OS (hazard ratio [HR] = 2.26, 95% confidence interval [CI]: 1.81-2.82, P < .001) and short PFS (HR = 1.75, 95% CI: 1.54-1.99, P < .001). Patients with low PNI tended to have a low ORR (odds ratio [OR] = 0.47, 95% CI: 0.34-0.65, P < .001) and DCR (OR = 0.43, 95% CI: 0.34-0.56, P < .001). However, the subgroup analysis demonstrated no significant association between PNI and survival time in patients receiving a programmed death ligand-1 inhibitor. PNI was significantly associated with survival time and treatment efficacy in patients treated with ICIs.
Collapse
Affiliation(s)
- Xin-Tian Xu
- Department of Pharmacy, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Yu Qian
- Department of Thoracic Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Meng-Xing Tian
- Department of Clinical Nutrition, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Chen-Chen Ding
- Department of Clinical Nutrition, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Huan Guo
- Department of Clinical Nutrition, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Jing Tang
- Department of Thoracic Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Guo-Liang Pi
- Department of Radiation Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Yuan Wu
- Department of Radiation Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Zhu Dai
- Department of Clinical Nutrition, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Xin Jin
- Department of Clinical Nutrition, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| |
Collapse
|
29
|
Li J, Xu X. Immune Checkpoint Inhibitor-Based Combination Therapy for Colorectal Cancer: An Overview. Int J Gen Med 2023; 16:1527-1540. [PMID: 37131870 PMCID: PMC10149070 DOI: 10.2147/ijgm.s408349] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Accepted: 04/19/2023] [Indexed: 05/04/2023] Open
Abstract
Colorectal cancer (CRC) is one of the most common diseases in the world. Tumor immunotherapy is an innovative cancer treatment that acts by activating the human body's autoimmune system. Immune checkpoint block has been shown to be effective in DNA deficient mismatch repair/microsatellite instability-high CRC. However, the therapeutic effect for proficient mismatch repair/microsatellite stability patients still requires further study and optimization. At present, the main CRC strategy is to combine other therapeutic methods, such as chemotherapy, targeted therapy, and radiotherapy. Here, we review the current status and the latest progress of immune checkpoint inhibitors in the treatment of CRC. At the same time, we consider therapeutic opportunities for transforming cold to hot, as well as perspectives on possible future therapies, which may be in great demand for drug-resistant patients.
Collapse
Affiliation(s)
- Jingjing Li
- Department of Gastroenterology, Shidong Hospital of Shanghai, Shanghai, People’s Republic of China
| | - Xuanfu Xu
- Department of Gastroenterology, Shidong Hospital of Shanghai, Shanghai, People’s Republic of China
| |
Collapse
|
30
|
Jiang X, Hu Y, Peng J, Luo X, Su L, Tang Y. Fangchinoline Exerts Anticancer Effects on Colorectal Cancer Cells by Evoking Cell Apoptosis via Endoplasmic Reticulum Stress. Bull Exp Biol Med 2023; 174:639-646. [PMID: 37052856 PMCID: PMC10098992 DOI: 10.1007/s10517-023-05761-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Indexed: 04/14/2023]
Abstract
We studied the anti-tumor effect of fangchinoline (FAN) against human colorectal cancer cell lines CCL-244 and SW480 and analyzed the mechanism of FAN action. The cell viability and apoptosis were assessed by MTT test and Annexin V-PI staining; caspase-3 activity was measured by Western blotting. The expression of endoplasmic reticulum stress-related proteins was assessed by real-time PCR, Western blotting, and gene transfection. It was found that FAN inhibited cell growth and induced apoptosis in human colorectal cancer cell lines CCL-244 and SW480 in a dose-dependent manner. The caspase-3 inhibitor Ac-DEVD-CHO could reverse the inhibitory effect of FAN. Moreover, FAN significantly increased the expression of endoplasmic reticulum stress-related proteins p-PERK, p-eIF2α, ATF4, and CHOP in CCL-244 and SW480 cells. In addition, endoplasmic reticulum stress inhibitor 4-phenylbutyric acid or CHOP knockdown could prevent FAN-induced apoptosis. Thus, FAN induced apoptosis of human colorectal cancer through activation of endoplasmic reticulum stress.
Collapse
Affiliation(s)
- Xiuling Jiang
- Medical School of Yangzhou Polytechnic College, Yangzhou, China
| | - Yougen Hu
- Department of Surgery of Jiangsu, Provincial Corps Hospital, Yangzhou, China
| | - Jianming Peng
- Medical School of Yangzhou Polytechnic College, Yangzhou, China
| | - Xue Luo
- Medical School of Yangzhou Polytechnic College, Yangzhou, China
| | - Landi Su
- Medical School of Yangzhou Polytechnic College, Yangzhou, China
| | - Yuanjie Tang
- Department of Surgery of Jiangsu, Provincial Corps Hospital, Yangzhou, China.
| |
Collapse
|
31
|
Di Sario G, Rossella V, Famulari ES, Maurizio A, Lazarevic D, Giannese F, Felici C. Enhancing clinical potential of liquid biopsy through a multi-omic approach: A systematic review. Front Genet 2023; 14:1152470. [PMID: 37077538 PMCID: PMC10109350 DOI: 10.3389/fgene.2023.1152470] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Accepted: 03/20/2023] [Indexed: 04/05/2023] Open
Abstract
In the last years, liquid biopsy gained increasing clinical relevance for detecting and monitoring several cancer types, being minimally invasive, highly informative and replicable over time. This revolutionary approach can be complementary and may, in the future, replace tissue biopsy, which is still considered the gold standard for cancer diagnosis. "Classical" tissue biopsy is invasive, often cannot provide sufficient bioptic material for advanced screening, and can provide isolated information about disease evolution and heterogeneity. Recent literature highlighted how liquid biopsy is informative of proteomic, genomic, epigenetic, and metabolic alterations. These biomarkers can be detected and investigated using single-omic and, recently, in combination through multi-omic approaches. This review will provide an overview of the most suitable techniques to thoroughly characterize tumor biomarkers and their potential clinical applications, highlighting the importance of an integrated multi-omic, multi-analyte approach. Personalized medical investigations will soon allow patients to receive predictable prognostic evaluations, early disease diagnosis, and subsequent ad hoc treatments.
Collapse
|
32
|
Li J, Hou H, Sun J, Ding Z, Xu Y, Li G. Systematic pan-cancer analysis identifies transmembrane protein 158 as a potential therapeutic, prognostic and immunological biomarker. Funct Integr Genomics 2023; 23:105. [PMID: 36977915 DOI: 10.1007/s10142-023-01032-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 03/12/2023] [Accepted: 03/15/2023] [Indexed: 03/30/2023]
Abstract
The purpose of this study was to investigate the expression significance, predictive value, immunologic function, and biological role of transmembrane protein 158 (TMEM158) in the development of pan-cancer. To achieve this, we utilized data from multiple databases, including TCGA, GTEx, GEPIA, and TIMER, to collect gene transcriptome, patient prognosis, and tumor immune data. We evaluated the association of TMEM158 with patient prognosis, tumor mutational burden (TMB), and microsatellite instability (MSI) in pan-cancer samples. We performed immune checkpoint gene co-expression analysis and gene set enrichment analysis (GSEA) to better understand the immunologic function of TMEM158. Our findings revealed that TMEM158 was significantly differentially expressed between most types of cancer tissues and their adjacent normal tissues and was associated with prognosis. Moreover, TMEM158 was significantly correlated with TMB, MSI, and tumor immune cell infiltration in multiple cancers. Co-expression analysis of immune checkpoint genes showed that TMEM158 was related to the expression of several common immune checkpoint genes, especially CTLA4 and LAG3. Gene enrichment analysis further revealed that TMEM158 was involved in multiple immune-related biological pathways in pan-cancer. Overall, this systematic pan-cancer analysis suggests that TMEM158 is generally highly expressed in various cancer tissues and is closely related to patient prognosis and survival across multiple cancer types. TMEM158 may serve as a significant predictor of cancer prognosis and modulate immune responses to various types of cancer.
Collapse
Affiliation(s)
- Jiayi Li
- School of Management, Shandong University, Jinan, 250100, Shandong, China
- School of Graduate, Hanyang University, Seoul, 04763, South Korea
| | - Haiguang Hou
- Department of Anatomy, School of Basic Medical Sciences, Shandong University, Jinan, 250012, Shandong, China
| | - Jinhao Sun
- Department of Anatomy, School of Basic Medical Sciences, Shandong University, Jinan, 250012, Shandong, China
| | - Zhaoxi Ding
- Department of Anatomy, School of Basic Medical Sciences, Shandong University, Jinan, 250012, Shandong, China
| | - Yingkun Xu
- Department of Breast and Thyroid Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Guibao Li
- Department of Anatomy, School of Basic Medical Sciences, Shandong University, Jinan, 250012, Shandong, China.
| |
Collapse
|
33
|
Su A, Pedraza R, Kennecke H. Developments in Checkpoint Inhibitor Therapy for the Management of Deficient Mismatch Repair (dMMR) Rectal Cancer. Curr Oncol 2023; 30:3672-3683. [PMID: 37185392 PMCID: PMC10136520 DOI: 10.3390/curroncol30040279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/22/2023] [Accepted: 03/24/2023] [Indexed: 03/29/2023] Open
Abstract
Deficient mismatch repair (dMMR)/microsatellite instability-high (MSIH) colorectal cancer is resistant to conventional chemotherapy but responds to immune checkpoint inhibition (ICI). We review the standard of care in locally advanced dMMR rectal cancer with a focus on ICI. We also present a case report to highlight the treatment complexities and unique challenges of this novel treatment approach. ICI can lead to immune related adverse events (irAEs), resulting in early treatment discontinuation as well as new challenges to surveillance and surgical management. Overall, neoadjuvant ICI can lead to robust treatment responses, but its impact on durable response and organ preservation requires further study.
Collapse
|
34
|
San-Román-Gil M, Torres-Jiménez J, Pozas J, Esteban-Villarrubia J, Albarrán-Fernández V, Álvarez-Ballesteros P, Chamorro-Pérez J, Rosero-Rodríguez D, Orejana-Martín I, Martínez-Delfrade Í, Reguera-Puertas P, Fuentes-Mateos R, Ferreiro-Monteagudo R. Current Landscape and Potential Challenges of Immune Checkpoint Inhibitors in Microsatellite Stable Metastatic Colorectal Carcinoma. Cancers (Basel) 2023; 15:863. [PMID: 36765821 PMCID: PMC9913409 DOI: 10.3390/cancers15030863] [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: 12/22/2022] [Revised: 01/25/2023] [Accepted: 01/27/2023] [Indexed: 01/31/2023] Open
Abstract
Colorectal cancer (CRC) is the third most frequent cancer and the second most common cause of cancer-related death in Europe. High microsatellite instability (MSI-H) due to a deficient DNA mismatch repair (dMMR) system can be found in 5% of metastatic CRC (mCRC) and has been established as a biomarker of response to immunotherapy in these tumors. Therefore, immune checkpoint inhibitors (ICIs) in mCRC with these characteristics were evaluated with results showing remarkable response rates and durations of response. The majority of mCRC cases have high levels of DNA mismatch repair proteins (pMMR) with consequent microsatellite stability or low instability (MSS or MSI-low), associated with an inherent resistance to ICIs. This review aims to provide a comprehensive analysis of the possible approaches to overcome the mechanisms of resistance and evaluates potential biomarkers to establish the role of ICIs in pMMR/MSS/MSI-L (MSS) mCRC.
Collapse
Affiliation(s)
- María San-Román-Gil
- Medical Oncology Department, Ramón y Cajal University Hospital, 28034 Madrid, Spain
| | - Javier Torres-Jiménez
- Medical Oncology Department, Clínico San Carlos University Hospital, 28040 Madrid, Spain
| | - Javier Pozas
- Medical Oncology Department, Ramón y Cajal University Hospital, 28034 Madrid, Spain
| | | | | | | | - Jesús Chamorro-Pérez
- Medical Oncology Department, Ramón y Cajal University Hospital, 28034 Madrid, Spain
| | | | | | | | | | | | | |
Collapse
|
35
|
Tan L, Li QS, Peng D, Cheng Y. Does PD-1 blockade play a decisive role in the pathological complete remission of unresectable MSS, BRAF V600E-mutated metastatic colorectal cancer: A case report. Front Oncol 2023; 12:976622. [PMID: 36713544 PMCID: PMC9880525 DOI: 10.3389/fonc.2022.976622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 12/19/2022] [Indexed: 01/15/2023] Open
Abstract
Background Colorectal cancer (CRC) ranks third in highest incidence among human cancers. With the continuous development of anti-cancer drugs, CRC patients are treated more and more effectively. However, the treatment of patients with unresectable metastatic CRC (mCRC) remains a core point for surgeons worldwide, especially for those with microsatellite stability (MSS) and BRAF V600E mutation, who have been reported to have the worst prognosis. Case description We report a case of pathological complete remission in a patient with unresectable MSS, BRAF V600E-mutated metastatic rectal cancer after using Vemurafenib and Cetuximab in combination with Camrelizumab. Conclusions This case suggested that Vemurafenib and Cetuximab combined with Camrelizumab is effective in the treatment of MSS, BRAF V600E-mutated mCRC. To benefit more patients, further studies need to be completed.
Collapse
Affiliation(s)
- Li Tan
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Qing-Shu Li
- Department of Pathology, School of Basic Medicine, Chongqing Medical University, Chongqing, China
| | - Dong Peng
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China,*Correspondence: Yong Cheng, ; Dong Peng,
| | - Yong Cheng
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China,*Correspondence: Yong Cheng, ; Dong Peng,
| |
Collapse
|
36
|
Hu M, Chong R, Liu W, Liu S, Liu X. Characteristic of molecular subtype based on lysosome-associated genes reveals clinical prognosis and immune infiltration of gastric cancer. Front Oncol 2023; 13:1155418. [PMID: 37197421 PMCID: PMC10183605 DOI: 10.3389/fonc.2023.1155418] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 04/11/2023] [Indexed: 05/19/2023] Open
Abstract
Background Lysosome are involved in nutrient sensing, cell signaling, cell death, immune responses and cell metabolism, which play an important role in the initiation and development of multiple tumors. However, the biological function of lysosome in gastric cancer (GC) has not been revealed. Here, we aim to screen lysosome-associated genes and established a corresponding prognostic risk signature for GC, then explore the role and underlying mechanisms. Methods The lysosome-associated genes (LYAGs) were obtained from MSigDB database. Differentially expressed lysosome-associated genes (DE-LYAGs) of GC were acquired based on the TCGA database and GEO database. According to expression profiles of DE-LYAGs, we divided the GC patients into different subgroups and then explored tumor microenvironment (TME) landscape and immunotherapy response in LYAG subtypes using GSVA, ESTIMATE and ssGSEA algorithms. Univariate Cox regression analysis, LASSO algorithm and multivariate Cox regression analysis were adopted to identify the prognostic LYAGs and then establish a risk model for patients with GC. The Kaplan-Meier analysis, Cox regression analysis and ROC analysis were utilized to evaluate the performance of the prognostic risk model. Clinical GC specimens were also used to verify the bioinformatics results by qRT-PCR assay. Results Thirteen DE-LYAGs were obtained and utilized to distinguish three subtypes in GC samples. Expression profiles of the 13 DE-LYAGs predicted prognosis, tumor-related immunological abnormalities and pathway dysregulation in these three subtypes. Furthermore, we constructed a prognostic risk model for GC based on DEG in the three subtypes. The Kaplan-Meier analysis suggested that higher risk score related to short OS rate. The Cox regression analysis and ROC analysis indicated that risk model had independent and excellent ability in predicting prognosis of GC patients. Mechanistically, a remarkable difference was observed in immune cell infiltration, immunotherapy response, somatic mutation landscape and drug sensitivity. qRT-PCR results showed that compared with corresponding adjacent normal tissues, most screened genes showed significant abnormal expressions and the expression change trends were consistent with the bioinformatics results. Conclusions We established a novel signature based on LYAGs which could be served as a prognostic biomarker for GC. Our study might provide new insights into individualized prognostication and precision treatment for GC.
Collapse
Affiliation(s)
- Maodong Hu
- Department of Gastroenterology, Huangdao District People’s Hospital, Qingdao, China
| | - Ruifeng Chong
- Department of General Surgery, Chengyang District People’s Hospital, Qingdao, China
| | - Weilin Liu
- General Surgery Department, Qingdao Hongdao People's Hospital, Chengyang District Center for Disease Control and Prevention, Qingdao, China
| | - Shuangyong Liu
- Department of Thyroid and Breast Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xiaolei Liu
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
- *Correspondence: Xiaolei Liu,
| |
Collapse
|
37
|
Deng X, Liu X, Hu B, Liu J, Fu B, Zhang W. Upregulation of MTHFD2 is associated with PD‑L1 activation in bladder cancer via the PI3K/AKT pathway. Int J Mol Med 2022; 51:14. [PMID: 36601741 PMCID: PMC9869724 DOI: 10.3892/ijmm.2022.5217] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 12/05/2022] [Indexed: 01/01/2023] Open
Abstract
Methylenetetrahydrofolate dehydrogenase 2 (MTHFD2) has been implicated in the etiology of various human malignant tumors; however, its exact role in bladder cancer (BC) remains to be explored. Through reverse transcription‑quantitative PCR, western blotting and immunohistochemistry detection of BC tissue, combined with The Cancer Genome Atlas (TCGA) database analysis, the present study demonstrated that MTHFD2 was upregulated in BC tissues. MTHFD2 expression in patients with BC was frequently associated with worse prognosis, tumor immune cell infiltration and programmed death‑ligand 1 (PD‑L1) expression. Subsequently, using short hairpin RNA, the expression levels of MTHFD2 were knocked down in BC cell lines, and the results revealed that the tumor cell proliferation and colony formation abilities of cells were greatly reduced, as determined by Cell Counting Kit 8 and colony formation assays, as was the expression of PD‑L1, as determined by western blotting. These findings were also confirmed in a xenograft nude mouse model. Simultaneously, it was revealed that abnormal expression of MTHFD2 was closely associated with the PI3K/AKT signaling pathway in both RNA‑sequencing and TCGA datasets. This observation was verified in vitro by detecting the protein expression levels of PI3K and AKT by western blotting. The activation of PI3K and AKT was enhanced in BC cells (T24) following stimulation with 740Y‑P, a PI3K activator, and cellular activities and PD‑L1 expression levels were restored. Finally, it was demonstrated that the MTHFD2 levels were correlated with chemosensitivity to traditional BC chemotherapeutic agents and various PI3K/AKT‑targeted drugs, as determined by analyzing the Genomics of Drug Sensitivity in Cancer database. Overall, the present findings revealed that upregulation of MTHFD2 was associated with PD‑L1 activation in BC via the PI3K/AKT signaling pathway, suggesting that it could be a promising marker of chemotherapy and immunotherapy for BC.
Collapse
Affiliation(s)
- Xinxi Deng
- Department of Urology, The First Affiliated Hospital of Nanchang University, Jiangxi Institute of Urology, Nanchang, Jiangxi 330006, P.R. China,Department of Urology, Jiujiang No. 1 People's Hospital (Affiliated Jiujiang Hospital of Nanchang University), Jiujiang, Jiangxi 332000, P.R. China
| | - Xiaoqiang Liu
- Department of Urology, The First Affiliated Hospital of Nanchang University, Jiangxi Institute of Urology, Nanchang, Jiangxi 330006, P.R. China
| | - Bing Hu
- Department of Urology, The First Affiliated Hospital of Nanchang University, Jiangxi Institute of Urology, Nanchang, Jiangxi 330006, P.R. China
| | - Jianyun Liu
- Jiangxi Provincial Key Lab of System Biomedicine, Jiujiang University, Jiujiang, Jiangxi 332000, P.R. China
| | - Bin Fu
- Department of Urology, The First Affiliated Hospital of Nanchang University, Jiangxi Institute of Urology, Nanchang, Jiangxi 330006, P.R. China,Correspondence to: Dr Bin Fu, Department of Urology, The First Affiliated Hospital of Nanchang University, Jiangxi Institute of Urology, 17 Yong Wai Zheng Street, Donghu, Nanchang, Jiangxi 330006, P.R. China, E-mail:
| | - Wensheng Zhang
- Department of Urology, Jiujiang No. 1 People's Hospital (Affiliated Jiujiang Hospital of Nanchang University), Jiujiang, Jiangxi 332000, P.R. China,Dr Wensheng Zhang, Department of Urology, Jiujiang No. 1 People's Hospital (Affiliated Jiujiang Hospital of Nanchang University), 48 Taling South Road, Xunyang, Jiujiang, Jiangxi 332000, P.R. China, E-mail:
| |
Collapse
|
38
|
Lyu C, Wang L, Stadlbauer B, Buchner A, Pohla H. A Pan-Cancer Landscape of ABCG2 across Human Cancers: Friend or Foe? Int J Mol Sci 2022; 23:ijms232415955. [PMID: 36555598 PMCID: PMC9784838 DOI: 10.3390/ijms232415955] [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: 10/24/2022] [Revised: 12/02/2022] [Accepted: 12/06/2022] [Indexed: 12/23/2022] Open
Abstract
Emerging evidence from research or clinical studies reported that ABCG2 (ATP-binding cassette sub-family G member 2) interrelates with multidrug resistance (MDR) development in cancers. However, no comprehensive pan-cancer analysis is available at present. Therefore, we explore multiple databases, such as TCGA to investigate the potential therapeutic roles of ABCG2 across 33 different tumors. ABCG2 is expressed on a lower level in most cancers and shows a protective effect. For example, a lower expression level of ABCG2 was detrimental to the survival of adrenocortical carcinoma (TCGA-ACC), glioblastoma multiforme (GBM), and kidney renal clear cell carcinoma (KIRC) patients. Distinct associations exist between ABCG2 expression and stemness scores, microenvironmental scores, microsatellite instability (MSI), and tumor mutational burden (TMB) of tumor patients. We observed a significant positive correlation between the ABCG2 mutation site and prognosis in uterine corpus endometrial carcinoma (UCEC) patients. Moreover, transmembrane transporter activity and hormone biosynthetic-associated functions were found to be involved in the functionality of ABCG2 and its related genes. The cDNAs of cancer cell lines were collected to detect exon mutation sequences and to analyze ABCG2 mRNA expression. The mRNA expression level of ABCG2 showed a significant difference among spheres and drug-resistant cancer cell lines compared with their corresponding adherent cancer cell lines in six types of cancer. This pan-cancer study provides, for the first time, a comprehensive understanding of the multifunctionality of ABCG2 and unveils further details of the potential therapeutic role of ABCG2 in pan-cancer.
Collapse
Affiliation(s)
- Chen Lyu
- Tumor Immunology Laboratory, LIFE Center, LMU Klinikum, University Munich, 82152 Planegg, Germany
| | - Lili Wang
- Tumor Immunology Laboratory, LIFE Center, LMU Klinikum, University Munich, 82152 Planegg, Germany
| | - Birgit Stadlbauer
- Tumor Immunology Laboratory, LIFE Center, LMU Klinikum, University Munich, 82152 Planegg, Germany
- Department of Urology, LMU Klinikum, University Munich, 81377 Munich, Germany
| | - Alexander Buchner
- Tumor Immunology Laboratory, LIFE Center, LMU Klinikum, University Munich, 82152 Planegg, Germany
- Department of Urology, LMU Klinikum, University Munich, 81377 Munich, Germany
| | - Heike Pohla
- Tumor Immunology Laboratory, LIFE Center, LMU Klinikum, University Munich, 82152 Planegg, Germany
- Department of Urology, LMU Klinikum, University Munich, 81377 Munich, Germany
- Correspondence:
| |
Collapse
|
39
|
Ke L, Li S, Cui H. The prognostic role of tumor mutation burden on survival of breast cancer: a systematic review and meta-analysis. BMC Cancer 2022; 22:1185. [PMID: 36397030 PMCID: PMC9673350 DOI: 10.1186/s12885-022-10284-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 11/04/2022] [Indexed: 11/18/2022] Open
Abstract
Background As a potential genetic biomarker, tumor mutation burden (TMB) has made progress in numerous tumors. There are limited data regarding TMB and its prognostic role is controversial in breast cancer. This systematic review and meta-analysis were conducted to assess the prognostic value of TMB on survival of breast cancer. Methods The databases PubMed, Embase, Web of Science, and Cochrane Library were searched for articles published through May 31, 2022. Moreover, effective data were extracted from included studies and calculated pooled effects of hazard ratio (HR) for overall survival (OS) and progression-free survival (PFS) by STATA 16.0. Heterogeneity was conducted by the I2 statistic and p-value. Using publication bias evaluation, sensitivity analysis, and subgroup analysis, the origin of heterogeneity was further investigated. Results They were up to 1,722 patients collected from sixteen cohorts for this analysis. The pooled effects of HR for both OS (HR: 1.14, 95% CI: 0.83,1.58, p > 0.01) and PFS (HR: 0.96, 95% CI: 0.53,1.71, p > 0.01) indicated no statistically significant difference in the high TMB and low TMB group. In immune checkpoint inhibitors (ICIs) subgroup, high TMB patients demonstrated benefit of OS (HR: 0.72, 95% CI: 0.59,0.87, p = 0.001) and PFS (HR: 0.52, 95% CI: 0.35,0.77, p < 0.001), whereas difference was not statistically significant in the non-ICIs subgroup (OS, HR:1.76, 95% CI: 0.97,3.20, p = 0.062; PFS, HR:2.31, 95% CI: 0.89,5.97, p = 0.086). In addition, sensitivity analysis revealed that the pooled effects were stable. The funnel plot and Begg's test suggested the absence of publication bias. Conclusion Meta-analysis revealed that the prognostic relevance of TMB in breast cancer is limited in scope. High TMB may be associated with longer survival only in ICIs-based treatment, but the association is not evident in non-ICIs-based treatment. Trial registration [https://www.crd.york.ac.uk/PROSPERO], Prospective Register of Systematic Reviews (PROSPERO), identifier: CRD42022342488. Supplementary Information The online version contains supplementary material available at 10.1186/s12885-022-10284-1.
Collapse
|
40
|
Yang S, Zhao S, Ye Y, Jia L, Lou Y. Global research trends on the links between gut microbiota and cancer immunotherapy: A bibliometric analysis (2012-2021). Front Immunol 2022; 13:952546. [PMID: 36090978 PMCID: PMC9449151 DOI: 10.3389/fimmu.2022.952546] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 08/05/2022] [Indexed: 11/17/2022] Open
Abstract
Background There is a crosstalk between gut microbiota (GM) and cancer immunotherapy (CI). The purpose of this study is to use bibliometric analysis to identify the highly cited papers relating to GM/CI and explore the research status and development trends of the GM/CI research. Methods A literature search regarding GM/CI publications from 2012 to 2021 was undertaken on July 4, 2022. The article titles, journals, authors, institutions, countries, total citations, keywords, and other information were extracted from the Science Citation Index Expanded (SCIE) of Web of Science Core Collection (WoSCC). The Bibliometrix of R package and VOSviewer were used for bibliometric analysis. Results A total of 665 papers were extracted. The number of papers has increased rapidly over the past decade, especially after 2018. The United States and China had the most publications and made great contributions to this field. Th5e Univ Texas MD Anderson Canc Ctr and Univ Paris Saclay were absolutely in the leading position in GM/CI. The most influential authors were Zitvogel L and Routy B. Frontiers in Immunology had the most publications and Science had the most total citations. Historical direct citation analysis explained the historical evolution in GM/CI. Highly cited papers and high-frequency keywords illustrated the current status and trends of GM/CI. Four clusters were identified and the important topics included the role of GM and antibiotics in CI, the methods of targeting GM to improve CI outcomes, the mechanism by which GM affects CI and the application of ICIs in melanoma. “Tumor microbiome”, “proton pump inhibitors” and “prognosis” may be the new focus of attention in the next few years. Conclusion This study filtered global publications on GM/CI correlation and analyzed their bibliometric characteristics, identified the most cited papers in GM/CI, and gained insight into the status, hotspots and trends of global GM/CI research, which may inform researchers and practitioners of future directions.
Collapse
Affiliation(s)
- Shanshan Yang
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
- Oncology Department of Integrated Traditional Chinese and Western Medicine, China-Japan Friendship Hospital, Beijing, China
| | - Suya Zhao
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Yixiang Ye
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Liqun Jia
- Oncology Department of Integrated Traditional Chinese and Western Medicine, China-Japan Friendship Hospital, Beijing, China
- *Correspondence: Liqun Jia, ; Yanni Lou,
| | - Yanni Lou
- Oncology Department of Integrated Traditional Chinese and Western Medicine, China-Japan Friendship Hospital, Beijing, China
- *Correspondence: Liqun Jia, ; Yanni Lou,
| |
Collapse
|
41
|
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
|
42
|
GRAMD1A Is a Biomarker of Kidney Renal Clear Cell Carcinoma and Is Associated with Immune Infiltration in the Tumour Microenvironment. DISEASE MARKERS 2022; 2022:5939021. [PMID: 35860689 PMCID: PMC9293538 DOI: 10.1155/2022/5939021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 05/18/2022] [Accepted: 06/25/2022] [Indexed: 11/17/2022]
Abstract
Background GRAM structural domain-containing protein 1A (GRAMD1A) is upregulated in a variety of human cancer tissues and is closely associated with tumourigenesis and progression. Methods Patient RNA-sequencing data and clinicopathological information were obtained from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) datasets. The expression of GRAMD1A in kidney cancer cell lines and KIRC patients was analysed by quantitative polymerase chain reaction (qPCR). Receiver Operator Characteristic (ROC) curves, nomograms, Kaplan-Meier analysis, forest plots, and COX analysis were used to assess the diagnostic and prognostic value of GRAMD1A in KIRC, and gene set enrichment analysis (GSEA) was used to explore its potential signalling pathways. In addition, the Sangerbox website, Kaplan-Meier plotter database, and TISIDB and TIMER databases were used to further analyse the correlation of GRAMD1A with microsatellite instability (MSI), tumour mutational burden (TMB), immune checkpoint genes, and tumour-infiltrating lymphocytes (TILs). Results GRAMD1A was significantly highly expressed in KIRC and associated with shorter overall survival and relapse-free survival (P < 0.05). The AUC value of the ROC curve to identify KIRC and normal renal tissues was 0.942. Forest plot and COX analysis visualized that GRAMD1A could be an independent prognostic factor in KIRC patients (P < 0.01), and nomograms to determine the overall survival (OS) of KIRC patients also showed good efficacy (C-index: 0.776). Moreover, Spearman correlation analysis showed a positive correlation between GRAMD1A and MSI, TMB (P < 0.01). On the other hand, GRAMD1A was also found to be closely associated with immune checkpoint genes. Meanwhile, patients with KIRC with high GRAMD1A expression had a relatively low hazard ratio (HR) of death when B lymphocytes, natural killer T cells, CD4+ T lymphocytes, CD8+ T lymphocytes, and macrophages were enriched in the tumour microenvironment (TME), and a greater HR of death when regulatory T lymphocytes with tumour-specific immunosuppressive effects were significantly enriched. Last, GSEA shows that GRAMD1A is closely associated with the regulation of energy metabolism in KIRC. Conclusions GRAMD1A is a promising diagnostic and prognostic biomarker for patients with KIRC, and its biological function correlates to some extent with immune infiltration in TME.
Collapse
|
43
|
A Newly Established Cuproptosis-Associated Long Non-Coding RNA Signature for Predicting Prognosis and Indicating Immune Microenvironment Features in Soft Tissue Sarcoma. JOURNAL OF ONCOLOGY 2022; 2022:8489387. [PMID: 35847354 PMCID: PMC9279026 DOI: 10.1155/2022/8489387] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Revised: 06/01/2022] [Accepted: 06/07/2022] [Indexed: 12/16/2022]
Abstract
Cuproptosis, a new type of programmed cell death, is involved in the development and progression of malignancies. The study of cuproptosis-associated long non-coding RNAs (lncRNAs) in soft tissue sarcomas (STSs) is however limited. There is also uncertainty regarding the prognostic accuracy of cuproptosis-associated lncRNAs in STSs and their relationship to the tumor immune microenvironment. The aim of this study was to determine the prognostic significance of cuprotosis-associated lncRNAs in STSs and their relationship to the tumor immune microenvironment. Transcriptomic and clinical data from patients with STSs were obtained through The Cancer Genome Atlas (TCGA). Overall, 259 patients were randomly allocated to a training group or a testing group. In the training group, a cuproptosis-associated lncRNA signature was constructed, and the signature was verified in the testing group. On the basis of risk scores and clinical features, we later developed a hybrid nomogram. We also performed functional and tumor immune microenvironment analysis based on the cuproptosis-associated lncRNA signature. A signature of 5 cuproptosis-associated lncRNAs was created. Based on this signature, we categorized STS patients into high-risk and low-risk groups. The study showed that patients at high risk had a worse prognosis than those at low risk. A nomogram was then constructed combining clinical characteristics with the risk scores, and it was shown to have credible predictive power. Functional enrichment and tumor immune microenvironmental analyses showed that high-risk STSs tend to be immunologically sensitive tumors. In our study, we found a cuproptosis-associated lncRNAs signature, which serves as an independent prognostic indicator. Cuproptosis-associated lncRNAs may play a role in the tumor immune microenvironment, which might be a therapeutic target for patients with STSs.
Collapse
|
44
|
Significance of a Tumor Mutation Burden Gene Signature with Prognosis and Immune Feature of Gastric Cancer Patients. Int J Genomics 2022; 2022:7684606. [PMID: 35719415 PMCID: PMC9201710 DOI: 10.1155/2022/7684606] [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: 04/11/2022] [Accepted: 05/25/2022] [Indexed: 12/14/2022] Open
Abstract
Gastric cancer (GC) is a common digestive tumor which ranks the fourth most common malignancy worldwide. Immunotherapy is a promising treatment for GC, especially for advanced gastric cancer (AGC). However, in clinical practice, not all patients are sensitive to immunotherapy. Recent studies showed that tumor mutation burden (TMB) is closely correlated with the response of immunotherapy. The current study identified a TMB-related genes' signature to predict the prognosis and immune feature of GC patients. Firstly, we acquired the TMB data and expression data from The Cancer Genome Atlas (TCGA) and the National Center for Biotechnology Information (NCBI) GEO databases. Then, we extracted TMB-related genes from the expression data of TCGA and two GEO cohorts. By using univariate Cox analysis, we identified that the 429 genes were correlated to GC patients' overall survival. Subsequently, an immune prognostic signature was constructed by using the least absolute shrinkage and selection operator analysis (LASSO) and multivariate Cox regression analysis. The signature could be utilized to predict the prognosis of GC patients. In addition, the signature showed a closed correlation with immune feature of GC patients. In conclusion, our risk signature could offer hints for the prognosis of GC patients and might provide insights to formulate new immunotherapy strategies for GC patients.
Collapse
|
45
|
Long AH, Morgenstern DA, Leruste A, Bourdeaut F, Davis KL. Checkpoint Immunotherapy in Pediatrics: Here, Gone, and Back Again. Am Soc Clin Oncol Educ Book 2022; 42:1-14. [PMID: 35580293 DOI: 10.1200/edbk_349799] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The role of immune checkpoint inhibitors (ICIs) in the treatment of pediatric cancers continues to evolve. Such therapies function by augmenting existing antitumor T-cell responses that have been rendered ineffective by inhibitory pathways. Although ICIs have proven highly effective for adult cancers, initial phase I/II clinical trials using single-agent ICIs against unselected pediatric cancers have been overall disappointing. With the exception of pediatric classic Hodgkin lymphoma, responses to ICIs have been infrequent, likely stemming from an inherent difference in the immunogenicity of childhood cancers, which, on average, have far fewer neoantigens than adult cancers. Recently, however, hope has reemerged that certain subsets of children with cancer may benefit from ICI therapies. In preliminary studies, patients with both pediatric hypermutated and SMARCB1-deficient cancers have had impressive responses to ICI therapies, likely as a result of underlying biologies that enhance neoantigen expression and tumoral inflammation. Dedicated trials are ongoing to fully evaluate the efficacy of ICIs for patients with these subsets of pediatric cancer.
Collapse
Affiliation(s)
- Adrienne H Long
- Division of Hematology, Oncology, and Stem Cell Transplant, Department of Pediatrics, Stanford University, Stanford, CA
| | - Daniel A Morgenstern
- Division of Haematology/Oncology, Department of Paediatrics, Hospital for Sick Children and University of Toronto, Toronto, Ontario, Canada
| | - Amaury Leruste
- SIREDO Oncology Center, Institut Curie, Paris Sciences et Lettres University, Paris, France
| | - Franck Bourdeaut
- SIREDO Oncology Center, Institut Curie, Laboratory of Translational Research in Pediatric Oncology, Paris, France
| | - Kara L Davis
- Division of Hematology, Oncology, and Stem Cell Transplant, Department of Pediatrics, Stanford University, Stanford, CA.,Center for Cancer Cellular Therapy, Stanford University, Stanford, CA
| |
Collapse
|