1
|
Wang Q, Yu M, Zhang S. The characteristics of the tumor immune microenvironment in colorectal cancer with different MSI status and current therapeutic strategies. Front Immunol 2025; 15:1440830. [PMID: 39877377 PMCID: PMC11772360 DOI: 10.3389/fimmu.2024.1440830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/30/2024] [Accepted: 12/16/2024] [Indexed: 01/31/2025] Open
Abstract
Colorectal cancer (CRC) remains a significant cause of cancer-related mortality worldwide. Despite advancements in surgery, chemotherapy, and radiotherapy, the effectiveness of these conventional treatments is limited, particularly in advanced cases. Therefore, transition to novel treatment is urgently needed. Immunotherapy, especially immune checkpoint inhibitors (ICIs), has shown promise in improving outcomes for CRC patients. Notably, patients with deficient mismatch repair (dMMR) or microsatellite instability-high (MSI-H) tumors often benefit from ICIs, while the majority of CRC cases, which exhibit proficient mismatch repair (pMMR) or microsatellite-stable (MSS) status, generally show resistance to this approach. It is assumed that the MSI phenotype cause some changes in the tumor microenvironment (TME), thus triggering antitumor immunity and leading to response to immunotherapy. Understanding these differences in the TME relative to MSI status is essential for developing more effective therapeutic strategies. This review provides an overview of the TME components in CRC and explores current approaches aimed at enhancing ICI efficacy in MSS CRC.
Collapse
Affiliation(s)
- Qingzhe Wang
- Department of Targeting Therapy and Immunology, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Min Yu
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Shuang Zhang
- Department of Targeting Therapy and Immunology, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
2
|
González-Montero J, Rojas CI, Burotto M. Predictors of response to immunotherapy in colorectal cancer. Oncologist 2024; 29:824-832. [PMID: 38920285 PMCID: PMC11449076 DOI: 10.1093/oncolo/oyae152] [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] [Academic Contribution Register] [Received: 03/19/2024] [Accepted: 05/24/2024] [Indexed: 06/27/2024] Open
Abstract
Colorectal cancer (CRC) is a major cause of cancer-related deaths globally. While treatment advancements have improved survival rates, primarily through targeted therapies based on KRAS, NRAS, and BRAF mutations, personalized treatment strategies for CRC remain limited. Immunotherapy, mainly immune checkpoint blockade, has shown efficacy in various cancers but is effective in only a small subset of patients with CRC with deficient mismatch repair (dMMR) proteins or high microsatellite instability (MSI). Recent research has challenged the notion that CRC is immunologically inert, revealing subsets with high immunogenicity and diverse lymphocytic infiltration. Identifying precise biomarkers beyond dMMR and MSI is crucial to expanding immunotherapy benefits. Hence, exploration has extended to various biomarker sources, such as the tumor microenvironment, genomic markers, and gut microbiota. Recent studies have introduced a novel classification system, consensus molecular subtypes, that aids in identifying patients with CRC with an immunogenic profile. These findings underscore the necessity of moving beyond single biomarkers and toward a comprehensive understanding of the immunological landscape in CRC, facilitating the development of more effective, personalized therapies.
Collapse
Affiliation(s)
- Jaime González-Montero
- Bradford Hill Clinical Research Center, Santiago 8420383, Chile
- Basic and Clinical Oncology Department, University of Chile, Santiago 838045, Chile
| | - Carlos I Rojas
- Bradford Hill Clinical Research Center, Santiago 8420383, Chile
| | | |
Collapse
|
3
|
Rustamadji P, Wiyarta E, Pramono M, Maulanisa SC. Response to Neoadjuvant Chemotherapy in Invasive Breast Cancer Predicted by CD4+, CD8+, and FOXP3+ Tumor-Infiltrating Lymphocytes. Asian Pac J Cancer Prev 2024; 25:1607-1613. [PMID: 38809632 PMCID: PMC11318808 DOI: 10.31557/apjcp.2024.25.5.1607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 11/22/2023] [Accepted: 05/04/2023] [Indexed: 05/31/2024] Open
Abstract
BACKGROUND Response to neoadjuvant chemotherapy (NC) in individuals with invasive breast cancer (IBC) must be monitored, and biomarkers are needed. NC can activate an anti-tumour immune response in its microenvironment, known as Tumor-infiltrating Lymphocytes (TIL). TIL components believed to have great potential as predictors are CD4+, CD8+, and FOXP3+ TIL. This study aims to explore TIL components that can potentially be predictive biomarkers of NC pathological responses. METHODS A sample size of 40 were analyzed based on the relationship between CD4+, CD8+, and FOXP3+ TIL expression with the Miller-Payne (MP) grading system. Age, tumour grade, PR, ER, Ki-67, and HER2 were also evaluated. CD4+, CD8+, and FOXP3+ TIL expressions were analayzed by IHC staining, while other data were collected from archives. Data was analyzed using univariate and multivariate analysis. RESULTS Univariate analysis showed a significant relationship between CD4+ TIL and MP (p<0.001), CD8+ and MP (p=0.004), and FOXP3 with MP (p<0.001). The simultaneous integration of the three biomarkers in one model was not good enough to be a predictive model. Therefore, an exploratory analysis was conducted by testing several alternative models that combined two of the three existing biomarkers. It turned out that CD4+ TIL in model 2 (CD4+CD8+) and FOXP3+ TIL in model 4 (CD8+FOXP3+) showed significant coefficient values. Moreover, all of the threshold coefficients in model 4 are significant. CONCLUSION This study shows that CD4+, CD8+, and FOXP3+ TIL have promising potential as predictive biomarkers. In particular, FOXP3+ is dominant in predictive models of pathological response in patients with IBC.
Collapse
MESH Headings
- Humans
- Lymphocytes, Tumor-Infiltrating/immunology
- Lymphocytes, Tumor-Infiltrating/metabolism
- Female
- Forkhead Transcription Factors/metabolism
- Breast Neoplasms/drug therapy
- Breast Neoplasms/pathology
- Breast Neoplasms/immunology
- Breast Neoplasms/metabolism
- Neoadjuvant Therapy/methods
- Middle Aged
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/metabolism
- Biomarkers, Tumor/metabolism
- Prognosis
- CD4-Positive T-Lymphocytes/immunology
- CD4-Positive T-Lymphocytes/metabolism
- Adult
- Follow-Up Studies
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Tumor Microenvironment/immunology
- Neoplasm Invasiveness
- Aged
- Carcinoma, Ductal, Breast/drug therapy
- Carcinoma, Ductal, Breast/pathology
- Carcinoma, Ductal, Breast/immunology
- Carcinoma, Ductal, Breast/metabolism
Collapse
Affiliation(s)
- Primariadewi Rustamadji
- Department of Anatomic Pathology, Faculty of Medicine Universitas Indonesia-Dr. Cipto Mangunkusumo National Hospital, Jakarta, Indonesia.
| | | | | | | |
Collapse
|
4
|
Pan B, Wang Z, Chen R, Zhang X, Qiu J, Wu X, Yao Y, Luo Y, Wang X, Tang N. Single-cell atlas reveals characteristic changes in intrahepatic HBV-specific leukocytes. Microbiol Spectr 2024; 12:e0286023. [PMID: 38032223 PMCID: PMC10782979 DOI: 10.1128/spectrum.02860-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/17/2023] [Accepted: 11/01/2023] [Indexed: 12/01/2023] Open
Abstract
IMPORTANCE Hepatitis B virus (HBV)-specific CD8+ T cells play a central role in the clearance of virus and HBV-related liver injury. Acute infection with HBV induces a vigorous, multifunctional CD8+ T cell response, whereas chronic one exhibits a weaker response. Our study elucidated HBV-specific T cell responses in terms of viral abundance rather than the timing of infection. We showed that in the premalignant stage, the degree of impaired T cell function was not synchronized with the viral surface antigen, which was attributed the liver's tolerance to the virus. However, after the development of hepatocellular carcinoma, T cell exhaustion was inevitable, and it was marked by the exhaustion of the signature transcription factor TOX.
Collapse
Affiliation(s)
- Banglun Pan
- Department of Hepatobiliary Surgery, Fujian Institute of Hepatobiliary Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Zengbin Wang
- Department of Immunology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Rui Chen
- Department of Hepatobiliary Surgery, Fujian Institute of Hepatobiliary Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Xiaoxia Zhang
- Department of Hepatobiliary Surgery, Fujian Institute of Hepatobiliary Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Jiacheng Qiu
- Department of Hepatobiliary Surgery, Fujian Institute of Hepatobiliary Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Xiaoxuan Wu
- Department of Hepatobiliary Surgery, Fujian Institute of Hepatobiliary Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Yuxin Yao
- Department of Hepatobiliary Surgery, Fujian Institute of Hepatobiliary Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Yue Luo
- Department of Hepatobiliary Surgery, Fujian Institute of Hepatobiliary Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Xiaoqian Wang
- Department of Hepatobiliary Surgery, Fujian Institute of Hepatobiliary Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Cancer Center of Fujian Medical University, Fujian Medical University Union Hospital, Fuzhou, China
| | - Nanhong Tang
- Department of Hepatobiliary Surgery, Fujian Institute of Hepatobiliary Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Cancer Center of Fujian Medical University, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
| |
Collapse
|
5
|
Tamari H, Kitadai Y, Takigawa H, Yuge R, Urabe Y, Shimamoto F, Oka S. Investigating the Role of Tumor-Infiltrating Lymphocytes as Predictors of Lymph Node Metastasis in Deep Submucosal Invasive Colorectal Cancer: A Retrospective Cross-Sectional Study. Cancers (Basel) 2023; 15:5238. [PMID: 37958412 PMCID: PMC10649548 DOI: 10.3390/cancers15215238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/18/2023] [Revised: 09/25/2023] [Accepted: 10/30/2023] [Indexed: 11/15/2023] Open
Abstract
The role of tumor-infiltrating T cells (TILs) in colorectal cancer (CRC) and their significance in early-stage CRC remain unknown. We investigated the role of TILs in early-stage CRC, particularly in deep submucosal invasive (T1b) CRC. Sixty patients with CRC (20 each with intramucosal [IM group], submucosal invasive [SM group], and advanced cancer [AD group]) were randomly selected. We examined changes in TILs with tumor invasion and the relationship between TILs and LN metastasis risk. Eighty-four patients with T1b CRC who underwent initial surgical resection with LN dissection or additional surgical resection with LN dissection after endoscopic resection were then selected. TIL phenotype and number were evaluated using triple immunofluorescence for CD4, CD8, and Foxp3. All subtypes were more numerous according to the degree of CRC invasion and more abundant at the invasive front of the tumor (IF) than in the center of the tumor (CT) in the SM and AD groups. The increased Foxp3 cells at the IF and high ratios of Foxp3/CD4 and Foxp3/CD8 positively correlated with LN metastasis. In conclusion, tumor invasion positively correlated with the number of TILs in CRC. The number and ratio of Foxp3 cells at the IF may predict LN metastasis in T1b CRC.
Collapse
Affiliation(s)
- Hirosato Tamari
- Department of Gastroenterology, Hiroshima University Hospital, Hiroshima 734-8551, Japan; (H.T.); (H.T.); (R.Y.); (S.O.)
| | - Yasuhiko Kitadai
- Department of Health Sciences, Faculty of Human Culture and Science, Prefectural University of Hiroshima, Hiroshima 734-8558, Japan
| | - Hidehiko Takigawa
- Department of Gastroenterology, Hiroshima University Hospital, Hiroshima 734-8551, Japan; (H.T.); (H.T.); (R.Y.); (S.O.)
| | - Ryo Yuge
- Department of Gastroenterology, Hiroshima University Hospital, Hiroshima 734-8551, Japan; (H.T.); (H.T.); (R.Y.); (S.O.)
| | - Yuji Urabe
- Department of Gastrointestinal Endoscopy and Medicine, Hiroshima University Hospital, Hiroshima 734-8551, Japan;
| | - Fumio Shimamoto
- Faculty of Health Sciences, Hiroshima Cosmopolitan University, Hiroshima 734-0014, Japan;
| | - Shiro Oka
- Department of Gastroenterology, Hiroshima University Hospital, Hiroshima 734-8551, Japan; (H.T.); (H.T.); (R.Y.); (S.O.)
| |
Collapse
|
6
|
Hu W, Pei Y, Ning R, Li P, Zhang Z, Hong Z, Bao C, Guo X, Sun Y, Zhang Q. Immunomodulatory effects of carbon ion radiotherapy in patients with localized prostate cancer. J Cancer Res Clin Oncol 2023; 149:4533-4545. [PMID: 36138265 PMCID: PMC10349746 DOI: 10.1007/s00432-022-04194-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/12/2022] [Accepted: 07/06/2022] [Indexed: 10/14/2022]
Abstract
PURPOSE Radiotherapy is one of the main local treatment modalities for prostate cancer, while immunosuppressive effect induced by radiotherapy is an important factor of radiation resistance and treatment failure. Carbon ion radiotherapy (CIRT) is a novel radiotherapy technique and the immunomodulatory effect of CIRT provides the possibility of overcoming radioresistance and improving efficacy. The aim of this study was to assess the immune response evoked by CIRT in localized prostate cancer patients. METHODS Thirty-two patients were treated by CIRT combined with or without hormone therapy and peripheral blood samples were collected before and after CIRT. Investigation of peripheral immune cell frequency, proliferation, and cytokine expression was conducted by flow cytometry, real-time quantitative PCR and ELISA. RESULTS There were no significant differences in the frequencies of CD3 + , CD4 + , CD8 + T cells and NK cells after CIRT. CD4/CD8 ratio increased whereas B cells decreased. All lymphocyte subsets except regulatory T cells (Tregs) displayed increased proliferation and T cells exhibited increased functionality after CIRT, characterized by modestly increased cytokine secretion of TNF. Moreover, higher frequencies of Tregs were shown. Neither monocytic myeloid-derived suppressor cells (MDSCs) nor early MDSCs changed after CIRT. TGF-β1 gene expression decreased while IL-6 showed a non-significant trend towards a decrease. Both IL-10 gene expression and plasma TGF-β1 level were unchanged. CONCLUSION CIRT demonstrates the potential to elicit immune activation in localized prostate cancer patients, based on sparing lymphocytes, increased lymphocyte proliferation, enhanced T-cell functionality, together with limited induction of immunosuppressive cells and reduced expression of immunosuppressive cytokines.
Collapse
Affiliation(s)
- Wei Hu
- Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Fudan University Cancer Hospital, Shanghai, 201321, China
- Shanghai Key Laboratory of Radiation Oncology (20dz2261000), Shanghai, China
- Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, Shanghai, 201321, China
| | - Yulei Pei
- Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Fudan University Cancer Hospital, Shanghai, 201321, China
- Shanghai Key Laboratory of Radiation Oncology (20dz2261000), Shanghai, China
- Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, Shanghai, 201321, China
| | - Renli Ning
- Shanghai Key Laboratory of Radiation Oncology (20dz2261000), Shanghai, China
- Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, Shanghai, 201321, China
- Department of Research and Development, Shanghai Proton and Heavy Ion Center, Fudan University Cancer Hospital, Shanghai, 201321, China
| | - Ping Li
- Shanghai Key Laboratory of Radiation Oncology (20dz2261000), Shanghai, China
- Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, Shanghai, 201321, China
- Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Shanghai, 201321, China
| | - Zhenshan Zhang
- Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Fudan University Cancer Hospital, Shanghai, 201321, China
- Shanghai Key Laboratory of Radiation Oncology (20dz2261000), Shanghai, China
- Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, Shanghai, 201321, China
| | - Zhengshan Hong
- Shanghai Key Laboratory of Radiation Oncology (20dz2261000), Shanghai, China
- Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, Shanghai, 201321, China
- Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Shanghai, 201321, China
| | - Cihang Bao
- Shanghai Key Laboratory of Radiation Oncology (20dz2261000), Shanghai, China
- Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, Shanghai, 201321, China
- Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Shanghai, 201321, China
| | - Xiaomao Guo
- Shanghai Key Laboratory of Radiation Oncology (20dz2261000), Shanghai, China.
- Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, Shanghai, 201321, China.
- Department of Research and Development, Shanghai Proton and Heavy Ion Center, Fudan University Cancer Hospital, Shanghai, 201321, China.
| | - Yun Sun
- Shanghai Key Laboratory of Radiation Oncology (20dz2261000), Shanghai, China.
- Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, Shanghai, 201321, China.
- Department of Research and Development, Shanghai Proton and Heavy Ion Center, Fudan University Cancer Hospital, Shanghai, 201321, China.
| | - Qing Zhang
- Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Fudan University Cancer Hospital, Shanghai, 201321, China.
- Shanghai Key Laboratory of Radiation Oncology (20dz2261000), Shanghai, China.
- Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, Shanghai, 201321, China.
| |
Collapse
|
7
|
Lao J, Xu H, Liang Z, Luo C, Shu L, Xie Y, Wu Y, Hao Y, Yuan Y. Peripheral changes in T cells predict efficacy of anti-PD-1 immunotherapy in non-small cell lung cancer. Immunobiology 2023; 228:152391. [PMID: 37167681 DOI: 10.1016/j.imbio.2023.152391] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/26/2023] [Revised: 03/29/2023] [Accepted: 04/25/2023] [Indexed: 05/13/2023]
Abstract
The application of programmed cell death protein 1 (PD-1) antibodies has brought great benefits to non-small cell lung cancer (NSCLC) patients. Nevertheless, not all patients respond to anti-PD-1 immunotherapy. This study aimed to find response markers to predict efficacy of anti-PD-1 immunotherapy in NSCLC patients. 80 patients with NSCLC who would accept anti-PD-1 immunotherapy were recruited, and peripheral blood was obtained before and after treatment. Flow cytometry was used to detect proportions of circulating cell subsets and expression of co-stimulatory molecules, co-inhibitory molecules and cytokines in T cells from pre- and post-treatment patients. Results showed that proportions of CD4+ and CD8+ T cells, NK, γδT and mucosal-associated invariant T (MAIT) cells were higher and regulatory T cells (Tregs) were lower in responders (n = 50) after treatment but no obvious difference was found in non-responders (n = 30). After treatment, responders showed an increase in the frequency of co-stimulatory and co-inhibitory molecules, as well as the production of cytokines in T cells. This study indicates that monitoring the alterations of immune markers in circulating cells from NSCLC patients may be helpful to discriminate responders and non-responders, which provides a potential novel way to assess efficacy of anti-PD-1 immunotherapy.
Collapse
Affiliation(s)
- Juanfeng Lao
- Department of Laboratory Medicine, The People's Hospital of Guangxi Zhuang Autonomous Region, Guangxi Academy of Medical Sciences, Nanning, Guangxi Zhuang Autonomous Region 530021, China
| | - Huiting Xu
- Center for Infection and Immunity, the Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China
| | - Zibin Liang
- Department of Thoracic Oncology, The Cancer Center of The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong Province 519020, China
| | - Changliang Luo
- Department of Laboratory Medicine, The People's Hospital of Guangxi Zhuang Autonomous Region, Guangxi Academy of Medical Sciences, Nanning, Guangxi Zhuang Autonomous Region 530021, China
| | - Liuyang Shu
- Department of Medical Oncology I, The People's Hospital of Guangxi Zhuang Autonomous Region & Research Center of Oncology, Guangxi Academy of Medical Sciences, Nanning, Guangxi Zhuang Autonomous Region 530021, China
| | - Yuping Xie
- Department of Thoracic Oncology, The Cancer Center of The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong Province 519020, China
| | - Yongjian Wu
- Center for Infection and Immunity, the Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China
| | - Yanrong Hao
- Department of Medical Oncology I, The People's Hospital of Guangxi Zhuang Autonomous Region & Research Center of Oncology, Guangxi Academy of Medical Sciences, Nanning, Guangxi Zhuang Autonomous Region 530021, China.
| | - Yulin Yuan
- Department of Laboratory Medicine, The People's Hospital of Guangxi Zhuang Autonomous Region, Guangxi Academy of Medical Sciences, Nanning, Guangxi Zhuang Autonomous Region 530021, China.
| |
Collapse
|
8
|
Műzes G, Sipos F. Autoimmunity and Carcinogenesis: Their Relationship under the Umbrella of Autophagy. Biomedicines 2023; 11:biomedicines11041130. [PMID: 37189748 DOI: 10.3390/biomedicines11041130] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 03/05/2023] [Revised: 04/04/2023] [Accepted: 04/06/2023] [Indexed: 04/11/2023] Open
Abstract
The immune system and autophagy share a functional relationship. Both innate and adaptive immune responses involve autophagy and, depending on the disease’s origin and pathophysiology, it may have a detrimental or positive role on autoimmune disorders. As a “double-edged sword” in tumors, autophagy can either facilitate or impede tumor growth. The autophagy regulatory network that influences tumor progression and treatment resistance is dependent on cell and tissue types and tumor stages. The connection between autoimmunity and carcinogenesis has not been sufficiently explored in past studies. As a crucial mechanism between the two phenomena, autophagy may play a substantial role, though the specifics remain unclear. Several autophagy modifiers have demonstrated beneficial effects in models of autoimmune disease, emphasizing their therapeutic potential as treatments for autoimmune disorders. The function of autophagy in the tumor microenvironment and immune cells is the subject of intensive study. The objective of this review is to investigate the role of autophagy in the simultaneous genesis of autoimmunity and malignancy, shedding light on both sides of the issue. We believe our work will assist in the organization of current understanding in the field and promote additional research on this urgent and crucial topic.
Collapse
Affiliation(s)
- Györgyi Műzes
- Immunology Division, Department of Internal Medicine and Hematology, Semmelweis University, 1088 Budapest, Hungary
| | - Ferenc Sipos
- Immunology Division, Department of Internal Medicine and Hematology, Semmelweis University, 1088 Budapest, Hungary
| |
Collapse
|
9
|
Fortis SP, Sofopoulos M, Goulielmaki M, Arnogiannaki N, Ardavanis A, Perez SA, Gritzapis AD, Baxevanis CN. Association between Intratumoral CD8+ T Cells with FoxP3+ and CD163+ Cells: A Potential Immune Intrinsic Negative Feedback Mechanism for Acquired Immune Resistance. Cancers (Basel) 2022; 14:cancers14246208. [PMID: 36551693 PMCID: PMC9777444 DOI: 10.3390/cancers14246208] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 11/11/2022] [Revised: 12/08/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022] Open
Abstract
Acquired immune resistance (AIR) describes a situation in which cancer patients who initially responded clinically to immunotherapies, after a certain period of time, progress with their disease. Considering that AIR represents a feedback response of the tumor against the immune attack generated during the course of immunotherapies, it is conceivable that AIR may also occur before treatment initiation as a mechanism to escape endogenous adaptive antitumor immunity (EAAI). In the present study, we assessed the EAAI in paraffin-embedded breast primary tumor tissue samples and drew correlations with the clinical outcomes. In particular, we analyzed densities of CD8+ cells as elements mediating antitumor cytotoxicity, and of CD163+ and FoxP3+ cells as suppressor elements. We found a direct correlation between the densities of CD8+ cells and of CD163+ and/or FoxP3+ cells in the vast majority of patients' tumors. Importantly, the vast majority of patients whose tumors were overpopulated by CD8+ cells developed AIR, which was characterized by high intratumoral CD163+ and/or FoxP3+ cell densities and reduced overall survival (OS). We also showed that AIR depends on the levels of CD8+ cell-ratios in the tumor center to the invasive margin. Our data suggest that tumors develop AIR only when under a robust endogenous immune pressure.
Collapse
Affiliation(s)
- Sotirios P. Fortis
- Cancer Immunology and Immunotherapy Center, Cancer Research Center, Saint Savas Cancer Hospital, 11522 Athens, Greece
| | | | - Maria Goulielmaki
- Cancer Immunology and Immunotherapy Center, Cancer Research Center, Saint Savas Cancer Hospital, 11522 Athens, Greece
| | - Niki Arnogiannaki
- Pathology Department, Saint Savas Cancer Hospital, 11522 Athens, Greece
| | - Alexandros Ardavanis
- First Medical Oncology Clinic, Saint Savas Cancer Hospital, 11522 Athens, Greece
| | - Sonia A. Perez
- Cancer Immunology and Immunotherapy Center, Cancer Research Center, Saint Savas Cancer Hospital, 11522 Athens, Greece
| | - Angelos D. Gritzapis
- Cancer Immunology and Immunotherapy Center, Cancer Research Center, Saint Savas Cancer Hospital, 11522 Athens, Greece
| | - Constantin N. Baxevanis
- Cancer Immunology and Immunotherapy Center, Cancer Research Center, Saint Savas Cancer Hospital, 11522 Athens, Greece
- Correspondence: ; Tel.: +30-21064-09380
| |
Collapse
|
10
|
TP53 and LRP1B Co-Wild Predicts Improved Survival for Patients with LUSC Receiving Anti-PD-L1 Immunotherapy. Cancers (Basel) 2022; 14:cancers14143382. [PMID: 35884443 PMCID: PMC9320428 DOI: 10.3390/cancers14143382] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/13/2022] [Revised: 07/02/2022] [Accepted: 07/06/2022] [Indexed: 02/04/2023] Open
Abstract
Immunotherapy brought long-term benefits for partial patients with lung squamous cell carcinoma (LUSC). The predictor of anti-PD-L1 therapy was controversial and limited in LUSC. We aimed to explore novel biomarker for LUSC immunotherapy and the potential mechanism. Five hundred and twenty-five Chinese patients (Geneplus cohort) with LUSC underwent targeted sequencing and were involved to explore the genomic profiling. TP53 and LRP1B were the most frequently recurrent genes and correlated to higher tumor mutational burden (TMB). We observed that LUSC patients with TP53 and LRP1B co-wild (co-wild type) were associated with better survival of anti-PD-L1 therapy compared with TP53 mutant or LRP1B mutant (mutant type) in POPAR/OAK cohort. Copy-number variation (CNV) and whole genome doubling (WGD) data from TCGA LUSC cohort were obtained to assess the CNV events. There were fewer CNV alterations and lower chromosome instability in patients with TP53/LRP1B co-wild compared with those with TP53/LRP1B mutant. RNA expression data from the TCGA LUSC cohort were collected to explore the differences in RNA expression and tumor immune microenvironment (TIME) between mutant and co-wild groups. The TP53/LRP1B co-wild type had a significantly increased proportion of multiple tumor-infiltrating lymphocytes (TILs), including activated CD8 T cell, activated dendritic cell (DC), and effector memory CD8 T cell. Immune-related gene sets including checkpoint, chemokine, immunostimulatory, MHC and receptors were enriched in the co-wild type. In conclusion, TP53/LRP1B co-wild LUSC conferred an elevated response rate in anti-PD-L1 therapy and improved survival, which was associated with a chromosome-stable phenotype and an activated immune microenvironment.
Collapse
|
11
|
Al-Mterin MA, Murshed K, Alsalman A, Abu-Dayeh A, Elkord E. Associations of different immune checkpoints-expressing CD4+ Treg/ T cell subsets with disease-free survival in colorectal cancer patients. BMC Cancer 2022; 22:601. [PMID: 35655158 PMCID: PMC9161559 DOI: 10.1186/s12885-022-09710-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 03/10/2022] [Accepted: 05/26/2022] [Indexed: 12/24/2022] Open
Abstract
There are different subsets of T regulatory cells (Tregs), orchestrating critical roles in the regulation of anti-tumor immunity in colorectal cancer (CRC). In this study, we report that a high frequency of circulating CD4+FoxP3+ Tregs was associated with poorer disease-free survival (DFS), while their higher frequencies in tumor-infiltrating CD4+ Tregs was associated with better DFS. We further investigated such associations with four Tregs/T cells expressing or lacking FoxP3 and Helios (FoxP3±Helios±). For the first time, we report that a high frequency of circulating CD4+FoxP3+Helios+ Tregs was associated with poorer DFS, while a high frequency of tumor-infiltrating CD4+FoxP3−Helios− T cells was associated with poorer DFS. In the four FoxP3±Helios± T cell subsets expressing any of the immune checkpoints (ICs) investigated, we found that a high frequency of CD4+FoxP3+Helios−PD-1+ Tregs in circulation was associated with worse DFS. We also found that high frequencies of FoxP3+Helios+CTLA-4+ Tregs, FoxP3+Helios−CTLA-4+ Tregs, and FoxP3−Helios+CTLA-4+ CD4+ T cells in circulation were associated with worse DFS. In contrast, high frequencies of CD4+TIM-3+ T cells, FoxP3+Helios+TIM-3+ Tregs, and FoxP3−Helios+TIM-3+ CD4+ T cells in circulation were associated with longer DFS. Our data show that certain CD4+ Treg/T cell subsets could serve as independent predictive biomarkers in CRC patients. Identification of the exact subpopulations contributing to clinical outcomes is critical for prognoses and therapeutic targeting.
Collapse
|
12
|
Stachtea X, Loughrey MB, Salvucci M, Lindner AU, Cho S, McDonough E, Sood A, Graf J, Santamaria-Pang A, Corwin A, Laurent-Puig P, Dasgupta S, Shia J, Owens JR, Abate S, Van Schaeybroeck S, Lawler M, Prehn JHM, Ginty F, Longley DB. Stratification of chemotherapy-treated stage III colorectal cancer patients using multiplexed imaging and single-cell analysis of T-cell populations. Mod Pathol 2022; 35:564-576. [PMID: 34732839 PMCID: PMC8964416 DOI: 10.1038/s41379-021-00953-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 02/16/2021] [Revised: 10/06/2021] [Accepted: 10/11/2021] [Indexed: 11/08/2022]
Abstract
Colorectal cancer (CRC) has one of the highest cancer incidences and mortality rates. In stage III, postoperative chemotherapy benefits <20% of patients, while more than 50% will develop distant metastases. Biomarkers for identification of patients at increased risk of disease recurrence following adjuvant chemotherapy are currently lacking. In this study, we assessed immune signatures in the tumor and tumor microenvironment (TME) using an in situ multiplexed immunofluorescence imaging and single-cell analysis technology (Cell DIVETM) and evaluated their correlations with patient outcomes. Tissue microarrays (TMAs) with up to three 1 mm diameter cores per patient were prepared from 117 stage III CRC patients treated with adjuvant fluoropyrimidine/oxaliplatin (FOLFOX) chemotherapy. Single sections underwent multiplexed immunofluorescence staining for immune cell markers (CD45, CD3, CD4, CD8, FOXP3, PD1) and tumor/cell segmentation markers (DAPI, pan-cytokeratin, AE1, NaKATPase, and S6). We used annotations and a probabilistic classification algorithm to build statistical models of immune cell types. Images were also qualitatively assessed independently by a Pathologist as 'high', 'moderate' or 'low', for stromal and total immune cell content. Excellent agreement was found between manual assessment and total automated scores (p < 0.0001). Moreover, compared to single markers, a multi-marker classification of regulatory T cells (Tregs: CD3+/CD4+FOXP3+/PD1-) was significantly associated with disease-free survival (DFS) and overall survival (OS) (p = 0.049 and 0.032) of FOLFOX-treated patients. Our results also showed that PD1- Tregs rather than PD1+ Tregs were associated with improved survival. These findings were supported by results from an independent FOLFOX-treated cohort of 191 stage III CRC patients, where higher PD1- Tregs were associated with an increase overall survival (p = 0.015) for CD3+/CD4+/FOXP3+/PD1-. Overall, compared to single markers, multi-marker classification provided more accurate quantitation of immune cell types with stronger correlations with outcomes.
Collapse
Affiliation(s)
- Xanthi Stachtea
- Patrick G. Johnston Centre for Cancer Research, School of Medicine, Dentistry and Biomedical Science, Queen's University Belfast, Northern Ireland, UK
| | - Maurice B Loughrey
- Patrick G. Johnston Centre for Cancer Research, School of Medicine, Dentistry and Biomedical Science, Queen's University Belfast, Northern Ireland, UK
- Department of Cellular Pathology, Royal Victoria Hospital, Belfast Health and Social Care trust, Belfast, UK
| | - Manuela Salvucci
- Department of Physiology and Medical Physics and Centre for Systems Medicine, Royal College of Surgeons in Ireland (RCSI) University of Medicine and Health Sciences, 123 St. Stephen's Green, Dublin 2, Ireland
- GE Research Center, 1 Research Circle, Niskayuna, NY, 12309, USA
| | - Andreas U Lindner
- Department of Physiology and Medical Physics and Centre for Systems Medicine, Royal College of Surgeons in Ireland (RCSI) University of Medicine and Health Sciences, 123 St. Stephen's Green, Dublin 2, Ireland
- GE Research Center, 1 Research Circle, Niskayuna, NY, 12309, USA
| | - Sanghee Cho
- GE Research Center, 1 Research Circle, Niskayuna, NY, 12309, USA
| | | | - Anup Sood
- GE Research Center, 1 Research Circle, Niskayuna, NY, 12309, USA
| | - John Graf
- GE Research Center, 1 Research Circle, Niskayuna, NY, 12309, USA
| | | | - Alex Corwin
- GE Research Center, 1 Research Circle, Niskayuna, NY, 12309, USA
| | | | | | - Jinru Shia
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jonathan R Owens
- GE Research Center, 1 Research Circle, Niskayuna, NY, 12309, USA
| | - Samantha Abate
- GE Research Center, 1 Research Circle, Niskayuna, NY, 12309, USA
| | - Sandra Van Schaeybroeck
- Patrick G. Johnston Centre for Cancer Research, School of Medicine, Dentistry and Biomedical Science, Queen's University Belfast, Northern Ireland, UK
| | - Mark Lawler
- Patrick G. Johnston Centre for Cancer Research, School of Medicine, Dentistry and Biomedical Science, Queen's University Belfast, Northern Ireland, UK
| | - Jochen H M Prehn
- Department of Physiology and Medical Physics and Centre for Systems Medicine, Royal College of Surgeons in Ireland (RCSI) University of Medicine and Health Sciences, 123 St. Stephen's Green, Dublin 2, Ireland
- GE Research Center, 1 Research Circle, Niskayuna, NY, 12309, USA
| | - Fiona Ginty
- GE Research Center, 1 Research Circle, Niskayuna, NY, 12309, USA
| | - Daniel B Longley
- Patrick G. Johnston Centre for Cancer Research, School of Medicine, Dentistry and Biomedical Science, Queen's University Belfast, Northern Ireland, UK.
| |
Collapse
|
13
|
Bandaru SS, Boyilla R, Merchant N, Nagaraju GP, El-Rayes B. Targeting T regulatory cells: their role in colorectal carcinoma progression and current clinical trials. Pharmacol Res 2022; 178:106197. [DOI: 10.1016/j.phrs.2022.106197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 02/21/2022] [Revised: 03/24/2022] [Accepted: 03/25/2022] [Indexed: 10/18/2022]
|
14
|
Immunotherapy for Colorectal Cancer: Mechanisms and Predictive Biomarkers. Cancers (Basel) 2022; 14:cancers14041028. [PMID: 35205776 PMCID: PMC8869923 DOI: 10.3390/cancers14041028] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/30/2021] [Revised: 02/07/2022] [Accepted: 02/09/2022] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Late-stage colorectal cancer treatment often involves chemotherapy and radiation that can cause dose-limiting toxicity, and therefore there is great interest in developing targeted therapies for this disease. Immunotherapy is a targeted therapy that uses peptides, cells, antibodies, viruses, or small molecules to engage or train the immune system to kill cancer. Here, we discuss the preclinical and clinical development of immunotherapy for treatment of colorectal cancer and provide an overview of predictive biomarkers for such treatments. We also consider open questions including optimal combination treatments and sensitization of colorectal cancer patients with proficient mismatch repair enzymes. Abstract Though early-stage colorectal cancer has a high 5 year survival rate of 65–92% depending on the specific stage, this probability drops to 13% after the cancer metastasizes. Frontline treatments for colorectal cancer such as chemotherapy and radiation often produce dose-limiting toxicities in patients and acquired resistance in cancer cells. Additional targeted treatments are needed to improve patient outcomes and quality of life. Immunotherapy involves treatment with peptides, cells, antibodies, viruses, or small molecules to engage or train the immune system to kill cancer cells. Preclinical and clinical investigations of immunotherapy for treatment of colorectal cancer including immune checkpoint blockade, adoptive cell therapy, monoclonal antibodies, oncolytic viruses, anti-cancer vaccines, and immune system modulators have been promising, but demonstrate limitations for patients with proficient mismatch repair enzymes. In this review, we discuss preclinical and clinical studies investigating immunotherapy for treatment of colorectal cancer and predictive biomarkers for response to these treatments. We also consider open questions including optimal combination treatments to maximize efficacy, minimize toxicity, and prevent acquired resistance and approaches to sensitize mismatch repair-proficient patients to immunotherapy.
Collapse
|
15
|
Schnellhardt S, Hirneth J, Büttner-Herold M, Daniel C, Haderlein M, Hartmann A, Fietkau R, Distel L. The Prognostic Value of FoxP3+ Tumour-Infiltrating Lymphocytes in Rectal Cancer Depends on Immune Phenotypes Defined by CD8+ Cytotoxic T Cell Density. Front Immunol 2022; 13:781222. [PMID: 35140715 PMCID: PMC8818710 DOI: 10.3389/fimmu.2022.781222] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 09/22/2021] [Accepted: 01/04/2022] [Indexed: 12/14/2022] Open
Abstract
Tumour-infiltrating FoxP3+ regulatory T cells have been identified as both positive and negative prognostic factors in colorectal cancer (CRC) and rectal cancer (RC). In this study we investigated whether immune phenotypes, defined by CD8+ cytotoxic T cell density, may influence the prognostic association of FoxP3+ T cell densities in RC. Tissue microarrays from 154 rectal cancer resections were immunohistochemically double stained for CD8 and FoxP3. CD8+ and FoxP3+ cell densities were measured in the stromal and intraepithelial compartment. Stromal FoxP3+ cell densities were not associated with 10-year overall survival (OS). In the “immune-desert” phenotype, defined by very low stromal CD8+ cell density, a high density of stromal FoxP3+ T cells displayed a tendency towards an association with decreased 10-year OS (p = 0.179). In “inflamed” tumours, defined by high intraepithelial CD8+ T cell infiltration, the opposite was the case and high stromal FoxP3+ T cell densities were a positive prognostic factor (p = 0.048). Additionally, patients with an increased FoxP3/CD8 cell density ratio demonstrated a strong trend towards decreased 10-year OS (p = 0.066). These contrasting findings suggest functional heterogeneity within the group of FoxP3+ T cells. They are consistent with experimental studies which reported suppressive and non-suppressive populations of FoxP3+ T cells in CRC. Furthermore, our study demonstrates that CD8 immunohistochemistry may act as an instrument to identify tumours infiltrated by possibly functionally differing FoxP3+ T cell subtypes.
Collapse
Affiliation(s)
- Sören Schnellhardt
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Comprehensive Cancer Center Erlangen-Europäische Metropolregion Nürnberg (CCC ER-EMN), Erlangen, Germany
| | - Johannes Hirneth
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Comprehensive Cancer Center Erlangen-Europäische Metropolregion Nürnberg (CCC ER-EMN), Erlangen, Germany
| | - Maike Büttner-Herold
- Comprehensive Cancer Center Erlangen-Europäische Metropolregion Nürnberg (CCC ER-EMN), Erlangen, Germany
- Department of Nephropathology, Institute of Pathology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Christoph Daniel
- Comprehensive Cancer Center Erlangen-Europäische Metropolregion Nürnberg (CCC ER-EMN), Erlangen, Germany
- Department of Nephropathology, Institute of Pathology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Marlen Haderlein
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Comprehensive Cancer Center Erlangen-Europäische Metropolregion Nürnberg (CCC ER-EMN), Erlangen, Germany
| | - Arndt Hartmann
- Comprehensive Cancer Center Erlangen-Europäische Metropolregion Nürnberg (CCC ER-EMN), Erlangen, Germany
- Institute of Pathology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Rainer Fietkau
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Comprehensive Cancer Center Erlangen-Europäische Metropolregion Nürnberg (CCC ER-EMN), Erlangen, Germany
| | - Luitpold Distel
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Comprehensive Cancer Center Erlangen-Europäische Metropolregion Nürnberg (CCC ER-EMN), Erlangen, Germany
- *Correspondence: Luitpold Distel,
| |
Collapse
|
16
|
Moatti A, Cohen JL. The TNF-α/TNFR2 Pathway: Targeting a Brake to Release the Anti-tumor Immune Response. Front Cell Dev Biol 2021; 9:725473. [PMID: 34712661 PMCID: PMC8546260 DOI: 10.3389/fcell.2021.725473] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/15/2021] [Accepted: 09/24/2021] [Indexed: 12/15/2022] Open
Abstract
Newly discovered anti-cancer immunotherapies, such as immune checkpoint inhibitors and chimeric antigen receptor T cells, focus on spurring the anti-tumor effector T cell (Teff) response. Although such strategies have already demonstrated a sustained beneficial effect in certain malignancies, a substantial proportion of treated patients does not respond. CD4+FOXP3+ regulatory T cells (Tregs), a suppressive subset of T cells, can impair anti-tumor responses and reduce the efficacy of currently available immunotherapies. An alternative view that has emerged over the last decade proposes to tackle this immune brake by targeting the suppressive action of Tregs on the anti-tumoral response. It was recently demonstrated that the tumor necrosis factor alpha (TNF-α) tumor necrosis factor receptor 2 (TNFR2) is critical for the phenotypic stabilization and suppressive function of human and mouse Tregs. The broad non-specific effects of TNF-α infusion in patients initially led clinicians to abandon this signaling pathway as first-line therapy against neoplasms. Previously unrecognized, TNFR2 has emerged recently as a legitimate target for anti-cancer immune checkpoint therapy. Considering the accumulation of pre-clinical data on the role of TNFR2 and clinical reports of TNFR2+ Tregs and tumor cells in cancer patients, it is now clear that a TNFR2-centered approach could be a viable strategy, once again making the TNF-α pathway a promising anti-cancer target. Here, we review the role of the TNFR2 signaling pathway in tolerance and the equilibrium of T cell responses and its connections with oncogenesis. We analyze recent discoveries concerning the targeting of TNFR2 in cancer, as well as the advantages, limitations, and perspectives of such a strategy.
Collapse
Affiliation(s)
- Audrey Moatti
- Université Paris-Est Créteil Val de Marne, INSERM, IMRB, Créteil, France.,AP-HP, Groupe Hospitalo-Universitaire Chenevier Mondor, Centre d'Investigation Clinique Biothérapie, Créteil, France
| | - José L Cohen
- Université Paris-Est Créteil Val de Marne, INSERM, IMRB, Créteil, France.,AP-HP, Groupe Hospitalo-Universitaire Chenevier Mondor, Centre d'Investigation Clinique Biothérapie, Créteil, France
| |
Collapse
|
17
|
DiToro D, Basu R. Emerging Complexity in CD4 +T Lineage Programming and Its Implications in Colorectal Cancer. Front Immunol 2021; 12:694833. [PMID: 34489941 PMCID: PMC8417887 DOI: 10.3389/fimmu.2021.694833] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/13/2021] [Accepted: 08/04/2021] [Indexed: 12/17/2022] Open
Abstract
The intestinal immune system has the difficult task of protecting a large environmentally exposed single layer of epithelium from pathogens without allowing inappropriate inflammatory responses. Unmitigated inflammation drives multiple pathologies, including the development of colorectal cancer. CD4+T cells mediate both the suppression and promotion of intestinal inflammation. They comprise an array of phenotypically and functionally distinct subsets tailored to a specific inflammatory context. This diversity of form and function is relevant to a broad array of pathologic and physiologic processes. The heterogeneity underlying both effector and regulatory T helper cell responses to colorectal cancer, and its impact on disease progression, is reviewed herein. Importantly, T cell responses are dynamic; they exhibit both quantitative and qualitative changes as the inflammatory context shifts. Recent evidence outlines the role of CD4+T cells in colorectal cancer responses and suggests possible mechanisms driving qualitative alterations in anti-cancer immune responses. The heterogeneity of T cells in colorectal cancer, as well as the manner and mechanism by which they change, offer an abundance of opportunities for more specific, and likely effective, interventional strategies.
Collapse
Affiliation(s)
- Daniel DiToro
- Brigham and Women's Hospital, Boston, MA, United States.,Harvard Medical School, Boston, MA, United States.,Ragon Institute of MGH MIT and Harvard, Cambridge, MA, United States
| | - Rajatava Basu
- Division of Molecular and Cellular Pathology, Department of Pathology, University of Alabama at Birmingham (UAB), Birmingham, AL, United States
| |
Collapse
|
18
|
Wu D, Hacking S, Vitkovski T, Nasim M. Superpixel image segmentation of VISTA expression in colorectal cancer and its relationship to the tumoral microenvironment. Sci Rep 2021; 11:17426. [PMID: 34465822 PMCID: PMC8408240 DOI: 10.1038/s41598-021-96417-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/03/2020] [Accepted: 07/19/2021] [Indexed: 01/22/2023] Open
Abstract
Colorectal cancer (CRC) is the third most common cause of cancer related death in the United States (Jasperson et al. in Gastroenterology 138:2044–2058, 10.1053/j.gastro.2010.01.054, 2010). Many studies have explored prognostic factors in CRC. Today, much focus has been placed on the tumor microenvironment, including different immune cells and the extracellular matrix (ECM). The present study aims to evaluate the role of V-domain immunoglobulin suppressor of T cell activation (VISTA). We utilized QuPath for whole slides image analysis, performing superpixel image segmentation (SIS) on a 226 patient-cohort. High VISTA expression correlated with better disease-free survival (DFS), high tumor infiltrative lymphocyte, microsatellite instability, BRAF mutational status as well as lower tumor stage. High VISTA expression was also associated with mature stromal differentiation (SD). When cohorts were separated based on SD and MMR, only patients with immature SD and microsatellite stability were found to correlate VISTA expression with DFS. Considering raised VISTA expression is associated with improved survival, TILs, mature SD, and MMR in CRC; careful, well-designed clinical trials should be pursued which incorporate the underlying tumoral microenvironment.
Collapse
Affiliation(s)
- Dongling Wu
- Department of Pathology and Laboratory Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA
| | - Sean Hacking
- Department of Pathology and Laboratory Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA
| | - Taisia Vitkovski
- Department of Pathology and Laboratory Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA
| | - Mansoor Nasim
- Department of Pathology and Laboratory Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA.
| |
Collapse
|
19
|
Wang DK, Zuo Q, He QY, Li B. Targeted Immunotherapies in Gastrointestinal Cancer: From Molecular Mechanisms to Implications. Front Immunol 2021; 12:705999. [PMID: 34447376 PMCID: PMC8383067 DOI: 10.3389/fimmu.2021.705999] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/06/2021] [Accepted: 07/26/2021] [Indexed: 12/12/2022] Open
Abstract
Gastrointestinal cancer is a leading cause of cancer-related mortality and remains a major challenge for cancer treatment. Despite the combined administration of modern surgical techniques and chemoradiotherapy (CRT), the overall 5-year survival rate of gastrointestinal cancer patients in advanced stage disease is less than 15%, due to rapid disease progression, metastasis, and CRT resistance. A better understanding of the mechanisms underlying cancer progression and optimized treatment strategies for gastrointestinal cancer are urgently needed. With increasing evidence highlighting the protective role of immune responses in cancer initiation and progression, immunotherapy has become a hot research topic in the integrative management of gastrointestinal cancer. Here, an overview of the molecular understanding of colorectal cancer, esophageal cancer and gastric cancer is provided. Subsequently, recently developed immunotherapy strategies, including immune checkpoint inhibitors, chimeric antigen receptor T cell therapies, tumor vaccines and therapies targeting other immune cells, have been described. Finally, the underlying mechanisms, fundamental research and clinical trials of each agent are discussed. Overall, this review summarizes recent advances and future directions for immunotherapy for patients with gastrointestinal malignancies.
Collapse
Affiliation(s)
| | | | | | - Bin Li
- Ministry of Education (MOE), Key Laboratory of Tumor Molecular Biology and Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Institute of Life and Health Engineering, Jinan University, Guangzhou, China
| |
Collapse
|
20
|
Yang Z, Gao A, Shi W, Wang J, Zhang X, Xu Z, Xu T, Zheng Y, Sun Y, Yang F. ILT4 in Colorectal Cancer Cells Induces Suppressive T Cell Contexture and Disease Progression. Onco Targets Ther 2021; 14:4239-4254. [PMID: 34321889 PMCID: PMC8312509 DOI: 10.2147/ott.s290348] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 11/09/2020] [Accepted: 06/16/2021] [Indexed: 12/30/2022] Open
Abstract
Purpose Immune checkpoint blockade (ICB) therapy shows little or no clinical benefit in most colorectal cancer (CRC) patients, due to the immunosuppressive T cell contexture in the tumor microenvironment (TME). Immunoglobulin-like transcript (ILT) 4 is an immunosuppressive molecule in myeloid cells. ILT4 is enriched in solid tumor cells, facilitating their proliferation, invasion, and metastasis. However, the regulatory role of ILT4 in T cell immunity of CRC is still undetermined. Here, we aimed to explore how tumor cell-derived ILT4 orchestrates T cell infiltration, subset distribution, and function in CRC. Methods A total of 145 paraffin-embedded cancer tissues and the corresponding clinicopathological information were collected from CRC patients. Immunohistochemical (IHC) staining and public database analyses determined the correlation of ILT4 expression with different T cell subset densities, IFN-γ levels, and patient outcomes. Paired Ig-like receptor B (PIR-B, ILT4 mouse ortholog)-overexpressing/-downregulated MC38 cells were subcutaneously injected into C57BL/6 mice as a CRC transplantation model. The frequencies, subsets, and IFN-γ levels of T cells in mouse blood and spleens were determined using flow cytometry and immunohistochemistry, respectively. Results High ILT4 expression in CRC cells was associated with decreased T cell infiltration, disease progression, and poor patient survival. T cell subset analyses indicated that ILT4-high patients showed reduced CD8+ T cell but elevated FOXP3+ regulatory T (Treg) cell frequencies in the TME. High ILT4 levels predicted lower IFN-γ production by tumor-infiltrating lymphocytes (TILs), especially by CD8+T cells in human CRC tissues. Moreover, PIR-B overexpression accelerated MC38 growth in mice, decreased CD3+/CD8+/IFN-γ+ T cell densities, and elevated Treg infiltration in the TME, blood, and spleens. PIR-B knockdown had the opposite effects. Conclusion ILT4 in CRC cells induced immunosuppressive T cell subset infiltration and impaired IFN-γ production in TILs, suggesting that ILT4 might be a potential immunotherapeutic target and prognostic biomarker.
Collapse
Affiliation(s)
- Zijiang Yang
- Department of Oncology, Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250013, People's Republic of China
| | - Aiqin Gao
- Department of Thoracic Radiation Oncology, Shandong Cancer Hospital and Institute, Jinan, Shandong, 250117, People's Republic of China
| | - Wenjing Shi
- Department of Oncology, Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250013, People's Republic of China
| | - Jingnan Wang
- Department of Oncology, Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250013, People's Republic of China
| | - Xianchao Zhang
- Department of Pathology, Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250013, People's Republic of China
| | - Zhengyan Xu
- Department of Pathology, Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250013, People's Republic of China
| | - Tingting Xu
- Department of Pathology, Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250013, People's Republic of China
| | - Yan Zheng
- Research Center of Translational Medicine, Jinan Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250013, People's Republic of China
| | - Yuping Sun
- Proton Center, Shandong Cancer Hospital and Institute, Jinan, Shandong, 250117, People's Republic of China
| | - Fei Yang
- Department of Pathology, Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250013, People's Republic of China
| |
Collapse
|
21
|
Jiang M, Wu C, Zhang L, Sun C, Wang H, Xu Y, Sun H, Zhu J, Zhao W, Fang Q, Yu J, Chen P, Wu S, Zheng Z, He Y, Zhou C. FOXP3-based immune risk model for recurrence prediction in small-cell lung cancer at stages I-III. J Immunother Cancer 2021; 9:jitc-2021-002339. [PMID: 34006632 PMCID: PMC8137193 DOI: 10.1136/jitc-2021-002339] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Accepted: 04/20/2021] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Immunotherapies may prolong the survival of patients with small-cell lung cancer (SCLC) to some extent. The role of forkhead box protein P3 (FOXP3) in tumor microenvironment (TME) remains controversial. We aimed to examine FOXP3-related expression characteristics and prognostic values and to develop a clinically relevant predictive system for SCLC. METHODS We enrolled 102 patients with histologically confirmed SCLC at stages I-III. Through immunohistochemistry, we determined the expression pattern of FOXP3 and its association with other immune biomarkers. By machine learning and statistical analysis, we constructed effective immune risk score models. Furthermore, we examined FOXP3-related enrichment pathways and TME traits in distinct cohorts. RESULTS In SCLC, FOXP3 level was significantly associated with status of programmed death-ligand 1 (PD-L1), programmed cell death protein 1 (PD-1), CD4, CD8, and CD3 (p=0.002, p=0.001, p=0.002, p=0.030, and p<0.001). High FOXP3 expression showed longer relapse-free survival (RFS) than the low-level group (41.200 months, 95% CI 26.937 to 55.463, vs 14.000 months, 95% CI 8.133 to 19.867; p=0.008). For tumor-infiltrating lymphocytes (TILs), subgroup analysis demonstrated FOXP3 and PD-1, PD-L1, lymphocyte activation gene-3, CD3, CD4, or CD8 double positive were significantly correlated with longer RFS. We further performed importance evaluation for immune biomarkers, constructed an immune risk score incorporating the top three important biomarkers, FOXP3, TIL PD-L1, and CD8, and found their independently prognostic role to predict SCLC relapse. Better predictive performance was achieved in this immune risk model compared with single-indicator-based or two-indicator-based prediction systems (area under the curve 0.715 vs 0.312-0.711). Then, relapse prediction system integrating clinical staging and immune risk score was established, which performed well in different cohorts. High FOXP3-related genes were enriched in several immune-related pathways, and the close relationships of interleukin-2, CD28, basic excision repair genes MUTYH, POLD1, POLD2, and oxidative phosphorylation related gene cytochrome c oxidase subunit 8A with FOXP3 expression were revealed. Moreover, we found low-immune risk score group had statistically higher activated CD4+ memory T cells (p=0.014) and plasma cells (p=0.049) than the high-risk group. The heterogeneity of tumor-infiltrating immune cells might represent a promising feature for risk prediction in SCLC. CONCLUSION FOXP3 interacts closely with immune biomarkers on tumor-infiltrating cells in TME. This study highlighted the crucial prognostic value and promising clinical applications of FOXP3 in SCLC.
Collapse
Affiliation(s)
- Minlin Jiang
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, No 507 Zhengmin Road, Shanghai 200433, China.,Tongji University, No 1239 Siping Road, Shanghai 200433, China
| | - Chunyan Wu
- Department of Pathology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, No 507 Zhengmin Road, Shanghai 200433, China
| | - Liping Zhang
- Department of Pathology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, No 507 Zhengmin Road, Shanghai 200433, China
| | - Chenglong Sun
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, No 507 Zhengmin Road, Shanghai 200433, China.,Tongji University, No 1239 Siping Road, Shanghai 200433, China
| | - Hao Wang
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, No 507 Zhengmin Road, Shanghai 200433, China.,Tongji University, No 1239 Siping Road, Shanghai 200433, China
| | - Yi Xu
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, No 507 Zhengmin Road, Shanghai 200433, China.,Tongji University, No 1239 Siping Road, Shanghai 200433, China
| | - Hui Sun
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, No 507 Zhengmin Road, Shanghai 200433, China
| | - Jun Zhu
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, No 507 Zhengmin Road, Shanghai 200433, China
| | - Wencheng Zhao
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, No 507 Zhengmin Road, Shanghai 200433, China
| | - Qiyu Fang
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, No 507 Zhengmin Road, Shanghai 200433, China
| | - Jia Yu
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, No 507 Zhengmin Road, Shanghai 200433, China
| | - Peixin Chen
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, No 507 Zhengmin Road, Shanghai 200433, China.,Tongji University, No 1239 Siping Road, Shanghai 200433, China
| | - Shengyu Wu
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, No 507 Zhengmin Road, Shanghai 200433, China.,Tongji University, No 1239 Siping Road, Shanghai 200433, China
| | - Zixuan Zheng
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, No 507 Zhengmin Road, Shanghai 200433, China.,Tongji University, No 1239 Siping Road, Shanghai 200433, China
| | - Yayi He
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, No 507 Zhengmin Road, Shanghai 200433, China
| | - Caicun Zhou
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, No 507 Zhengmin Road, Shanghai 200433, China
| |
Collapse
|
22
|
Wilkinson K, Ng W, Roberts TL, Becker TM, Lim SHS, Chua W, Lee CS. Tumour immune microenvironment biomarkers predicting cytotoxic chemotherapy efficacy in colorectal cancer. J Clin Pathol 2021; 74:625-634. [PMID: 33753562 PMCID: PMC8461409 DOI: 10.1136/jclinpath-2020-207309] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/01/2020] [Accepted: 12/22/2020] [Indexed: 12/22/2022]
Abstract
The role of the local tumour and stromal immune landscape is increasingly recognised to be important in cancer development, progression and response to therapy. The composition, function, spatial orientation and gene expression profile of the infiltrate of the innate and adaptive immune system at the tumour and surrounding tissue has an established prognostic role in colorectal cancer (CRC). Multiple studies have confirmed that a tumour immune microenvironment (TIME) reflective of a type 1 adaptive immune response is associated with improved prognosis. There have been significant efforts to evolve these observations into validated, histopathology-based prognostic biomarkers, such as the Immunoscore. However, the clinical need lies much more in the development of predictive, not prognostic, biomarkers which have the potential to improve patient outcomes. This is particularly pertinent to help guide cytotoxic chemotherapy use in CRC, which remains the standard of care. Cytotoxic chemotherapy has recognised immunomodulatory activity distinct from its antimitotic effects, including mechanisms such as immunogenic cell death (ICD) and induction/inhibition of key immune players. Response to chemotherapy may differ with regard to molecular subtype of CRC, which are strongly associated with immune phenotypes. Thus, immune markers are potentially useful, though under-reported, predictive biomarkers. In this review, we discuss the impact of the TIME on response to cytotoxic chemotherapy in CRC, with a focus on baseline immune markers, and associated genomic and transcriptomic signatures.
Collapse
Affiliation(s)
- Kate Wilkinson
- Liverpool Cancer Therapy Centre, Liverpool Hospital, Liverpool, New South Wales, Australia .,School of Medicine, Western Sydney University, Liverpool, New South Wales, Australia
| | - Weng Ng
- Liverpool Cancer Therapy Centre, Liverpool Hospital, Liverpool, New South Wales, Australia.,School of Medicine, Western Sydney University, Liverpool, New South Wales, Australia
| | - Tara Laurine Roberts
- School of Medicine, Western Sydney University, Liverpool, New South Wales, Australia.,Ingham Institute for Applied Medical Research, Liverpool, New South Wales, Australia
| | - Therese M Becker
- School of Medicine, Western Sydney University, Liverpool, New South Wales, Australia.,Ingham Institute for Applied Medical Research, Liverpool, New South Wales, Australia
| | - Stephanie Hui-Su Lim
- School of Medicine, Western Sydney University, Liverpool, New South Wales, Australia.,Ingham Institute for Applied Medical Research, Liverpool, New South Wales, Australia.,Macarthur Cancer Therapy Centre, Campbelltown Hospital, Campbelltown, New South Wales, Australia
| | - Wei Chua
- Liverpool Cancer Therapy Centre, Liverpool Hospital, Liverpool, New South Wales, Australia.,School of Medicine, Western Sydney University, Liverpool, New South Wales, Australia.,Ingham Institute for Applied Medical Research, Liverpool, New South Wales, Australia
| | - Cheok Soon Lee
- School of Medicine, Western Sydney University, Liverpool, New South Wales, Australia.,Ingham Institute for Applied Medical Research, Liverpool, New South Wales, Australia.,Department of Anatomical Pathology, Liverpool Hospital, Liverpool, New South Wales, Australia
| |
Collapse
|
23
|
Correale P, Rotundo MS, Botta C, Del Vecchio MT, Tassone P, Tagliaferri P. Tumor infiltration by chemokine receptor 7 (CCR7)(+) T-lymphocytes is a favorable prognostic factor in metastatic colorectal cancer. Oncoimmunology 2021; 1:531-532. [PMID: 22754775 PMCID: PMC3382880 DOI: 10.4161/onci.19404] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/15/2022] Open
Abstract
The immune interactions occurring within the tumor microenvironment have a critical role in determining the outcome of colorectal cancer patients. We carried-out an immunohistochemical analysis of tumor infiltrating T-lymphocytes expressing chemokine receptor 7 (CCR7) in a series of colorectal cancer patients enrolled in a prospective clinical trial. We demonstrated that a high tumor infiltration score of this lymphocyte subset is predictive of longer progression free survival and overall survival.
Collapse
Affiliation(s)
- Pierpaolo Correale
- Medical Oncology Unit; Oncology Department; Azienda Ospedaliera Universitaria Senese; Siena, Italy
| | | | | | | | | | | |
Collapse
|
24
|
Senovilla L, Vacchelli E, Galon J, Adjemian S, Eggermont A, Fridman WH, Sautès-Fridman C, Ma Y, Tartour E, Zitvogel L, Kroemer G, Galluzzi L. Trial watch: Prognostic and predictive value of the immune infiltrate in cancer. Oncoimmunology 2021; 1:1323-1343. [PMID: 23243596 PMCID: PMC3518505 DOI: 10.4161/onci.22009] [Citation(s) in RCA: 188] [Impact Index Per Article: 47.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/12/2022] Open
Abstract
Solid tumors are constituted of a variety of cellular components, including bona fide malignant cells as well as endothelial, structural and immune cells. On one hand, the tumor stroma exerts major pro-tumorigenic and immunosuppressive functions, reflecting the capacity of cancer cells to shape the microenvironment to satisfy their own metabolic and immunological needs. On the other hand, there is a component of tumor-infiltrating leucocytes (TILs) that has been specifically recruited in the attempt to control tumor growth. Along with the recognition of the critical role played by the immune system in oncogenesis, tumor progression and response to therapy, increasing attention has been attracted by the potential prognostic and/or predictive role of the immune infiltrate in this setting. Data from large clinical studies demonstrate indeed that a robust infiltration of neoplastic lesions by specific immune cell populations, including (but not limited to) CD8+ cytotoxic T lymphocytes, Th1 and Th17 CD4+ T cells, natural killer cells, dendritic cells, and M1 macrophages constitutes an independent prognostic indicator in several types of cancer. Conversely, high levels of intratumoral CD4+CD25+FOXP3+ regulatory T cells, Th2 CD4+ T cells, myeloid-derived suppressor cells, M2 macrophages and neutrophils have frequently been associated with dismal prognosis. So far, only a few studies have addressed the true predictive potential of TILs in cancer patients, generally comforting the notion that—at least in some clinical settings—the immune infiltrate can reliably predict if a specific patient will respond to therapy or not. In this Trial Watch, we will summarize the results of clinical trials that have evaluated/are evaluating the prognostic and predictive value of the immune infiltrate in the context of solid malignancies.
Collapse
Affiliation(s)
- Laura Senovilla
- Institut Gustave Roussy; Villejuif, France ; Université Paris-Sud/Paris XI; Orsay, France ; INSERM, U848; Villejuif, France
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Lança T, Silva-Santos B. The split nature of tumor-infiltrating leukocytes: Implications for cancer surveillance and immunotherapy. Oncoimmunology 2021; 1:717-725. [PMID: 22934263 PMCID: PMC3429575 DOI: 10.4161/onci.20068] [Citation(s) in RCA: 111] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/18/2022] Open
Abstract
An important development in tumor immunology was the identification of highly diverse tumor-infiltrating leukocyte subsets that can play strikingly antagonistic functions. Namely, “anti-tumor” vs. “pro-tumor” roles have been suggested for Th1 and Th17 subsets of CD4+ T cells, Type I or Type II NKT cells, M1 and M2 macrophages, or N1 and N2 neutrophils, respectively. While these findings are being validated in cancer patients, it is also clear that the balance between infiltrating CD8+ cytotoxic and Foxp3+ regulatory T cells has prognostic value. Here we review the pre-clinical and clinical data that have shaped our current understanding of tumor-infiltrating leukocytes.
Collapse
Affiliation(s)
- Telma Lança
- Instituto de Medicina Molecular; Faculdade de Medicina; Universidade de Lisboa; Lisbon, Portugal
| | | |
Collapse
|
26
|
Tsujimoto H, Kobayashi M, Sugasawa H, Ono S, Kishi Y, Ueno H. Potential mechanisms of tumor progression associated with postoperative infectious complications. Cancer Metastasis Rev 2021; 40:285-296. [PMID: 33389285 DOI: 10.1007/s10555-020-09945-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 11/03/2020] [Accepted: 11/24/2020] [Indexed: 02/06/2023]
Abstract
There is increasing evidence that postoperative infectious complications (PICs) are associated with poor prognosis after potentially curative surgery. However, the role that PICs play in tumor development remains unclear. In this article, we reviewed the literature for novel insights on the mechanisms of cancer progression associated with PICs. The Medline and EMBASE databases were searched for publications regarding the role of suppression of antitumor immunity by PIC in tumor progression and selected 916 manuscripts were selected for this review. In addition, a summary of the authors' own experimental data from this field was set in the context of current knowledge regarding cancer progression under septic conditions. Initially, sepsis/microbial infection dramatically activates the systemic immune system with increases in pro-inflammatory mediators, which results in the development of systemic inflammatory response syndrome; however, when sepsis persists in septic patients, a shift toward an anti-inflammatory immunosuppressive state, characterized by macrophage deactivation, reduced antigen presentation, T cell anergy, and a shift in the T helper cell pattern to a predominantly TH2-type response, occurs. Thus, various cytokine reactions and the immune status dynamically change during microbial infection, including PIC. We proposed three possible mechanisms for the tumor progression associated with PIC: first, a mechanism in which microbes and/or microbial PAMPs may be directly involved in cancer growth; second, a mechanism in which factors released from immunocompetent cells during infections may affect tumor progression; and third, a mechanism in which factors suppress host tumor immunity during infections, which may result in tumor progression. A more detailed understanding by surgeons of the immunological features in cancer patients with PIC can subsequently open new avenues for improving unfavorable long-term oncological outcomes associated with PICs.
Collapse
Affiliation(s)
- Hironori Tsujimoto
- Department of Surgery, National Defense Medical College, 3-2 Namiki, Tokorozawa, 359-8513, Japan.
| | - Minako Kobayashi
- Department of Surgery, National Defense Medical College, 3-2 Namiki, Tokorozawa, 359-8513, Japan
| | - Hidekazu Sugasawa
- Department of Surgery, National Defense Medical College, 3-2 Namiki, Tokorozawa, 359-8513, Japan
| | - Satoshi Ono
- Department of Surgery, National Defense Medical College, 3-2 Namiki, Tokorozawa, 359-8513, Japan
| | - Yoji Kishi
- Department of Surgery, National Defense Medical College, 3-2 Namiki, Tokorozawa, 359-8513, Japan
| | - Hideki Ueno
- Department of Surgery, National Defense Medical College, 3-2 Namiki, Tokorozawa, 359-8513, Japan
| |
Collapse
|
27
|
Farc O, Cristea V. An overview of the tumor microenvironment, from cells to complex networks (Review). Exp Ther Med 2021; 21:96. [PMID: 33363607 PMCID: PMC7725019 DOI: 10.3892/etm.2020.9528] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/28/2020] [Accepted: 09/29/2020] [Indexed: 01/13/2023] Open
Abstract
For a long period, cancer has been believed to be a gene disease, in which oncogenic and suppressor mutations accumulate gradually, finally leading to the malignant transformation of cells. This vision has changed in the last few years, the involvement of the tumor microenvironment, the non-malignant part of the tumors, as an important contributor to the malignant growth being now largely recognized. There is a consensus according to which the understanding of the tumor microenvironment is important as a means to develop new approaches in the therapy of cancer. In this context, the present study is a review of the different types of non-malignant cells that can be found in tumors, with their pro or antitumoral actions, presence in tumors and therapeutic targeting. These cells establish complex relations between them, through cytokines, exosomes, cell adhesion, co-stimulation and co-inhibition; these relations will also be examined in the present work.
Collapse
Affiliation(s)
- Ovidiu Farc
- Immunology Department, ‘Iuliu Hatieganu’ University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
| | - Victor Cristea
- Immunology Department, ‘Iuliu Hatieganu’ University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
| |
Collapse
|
28
|
Regulatory (FoxP3 +) T cells and TGF-β predict the response to anti-PD-1 immunotherapy in patients with non-small cell lung cancer. Sci Rep 2020; 10:18994. [PMID: 33149213 PMCID: PMC7642363 DOI: 10.1038/s41598-020-76130-1] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/08/2020] [Accepted: 10/01/2020] [Indexed: 01/28/2023] Open
Abstract
Antitumor immune responses induced by immune checkpoint inhibitors anti-PD-1 or anti-PD-L1 have been used as therapeutic strategies in advanced non-small cell lung cancer (NSCLC) patients over the last decade. Favorable antitumor activity to immune checkpoint inhibitors is correlated with high PD-L1 expression, increased tumor-infiltrating lymphocytes, and decreased suppressive immune cells including Treg cells, myeloid-derived suppressor cells, or tumor-associated macrophages in various cancer types. In this study, we investigated the potential correlation between clinical outcomes and peripheral blood immune cell profiles, specifically focused on FoxP3+ Treg cells, collected at baseline and one week after anti-PD-1 therapy in two independent cohorts of patients with NSCLC: a discovery cohort of 83 patients and a validation cohort of 49 patients. High frequencies of circulating Treg cells one week after anti-PD-1 therapy were correlated with a high response rate, longer progression-free survival, and overall survival. Furthermore, high levels of TGF-β and Treg cells were associated with favorable clinical outcomes. Our results suggest that higher levels of FoxP3+ Treg cells and TGF-β can predict a favorable response to anti-PD-1 immunotherapy in patients with advanced NSCLC.
Collapse
|
29
|
Oshi M, Asaoka M, Tokumaru Y, Angarita FA, Yan L, Matsuyama R, Zsiros E, Ishikawa T, Endo I, Takabe K. Abundance of Regulatory T Cell (Treg) as a Predictive Biomarker for Neoadjuvant Chemotherapy in Triple-Negative Breast Cancer. Cancers (Basel) 2020; 12:E3038. [PMID: 33086518 PMCID: PMC7603157 DOI: 10.3390/cancers12103038] [Citation(s) in RCA: 87] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 09/20/2020] [Revised: 10/12/2020] [Accepted: 10/16/2020] [Indexed: 02/07/2023] Open
Abstract
Regulatory CD4+ T cell (Treg), a subset of tumor-infiltrating lymphocytes (TILs), are known to suppress anticancer immunity but its clinical relevance in human breast cancer remains unclear. In this study, we estimated the relative abundance of Tregs in breast cancer of multiple patient cohorts by using the xCell algorithm on bulk tumor gene expression data. In total, 5177 breast cancer patients from five independent cohorts (TCGA-BRCA, GSE96058, GSE25066, GSE20194, and GSE110590) were analyzed. Treg abundance was not associated with cancer aggressiveness, patient survival, or immune activity markers, but it was lower in metastatic tumors when compared to matched primary tumors. Treg was associated with a high mutation rate of TP53 genes and copy number mutations as well as with increased tumor infiltration of M2 macrophages and decreased infiltration of T helper type 1 (Th1) cells. Pathological complete response (pCR) after neoadjuvant chemotherapy (NAC) was significantly associated with low Treg abundance in triple negative breast cancer (TNBC) but not in ER-positive/Her2-negative subtype. High Treg abundance was significantly associated with high tumor expression of multiple immune checkpoint inhibitor genes. In conclusion, Treg abundance may have potential as a predictive biomarker of pCR after NAC in TNBC.
Collapse
Affiliation(s)
- Masanori Oshi
- Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA; (M.O.); (M.A.); (Y.T.); (F.A.A.)
- Department of Gastroenterological Surgery, Yokohama City University Graduate School of Medicine, Yokohama 236-0004, Japan; (R.M.); (I.E.)
| | - Mariko Asaoka
- Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA; (M.O.); (M.A.); (Y.T.); (F.A.A.)
- Department of Breast Surgery and Oncology, Tokyo Medical University, Tokyo 160-8402, Japan;
| | - Yoshihisa Tokumaru
- Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA; (M.O.); (M.A.); (Y.T.); (F.A.A.)
- Department of Surgical Oncology, Graduate School of Medicine, Gifu University, 1-1 Yanagido, Gifu 501-1194, Japan
| | - Fernando A. Angarita
- Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA; (M.O.); (M.A.); (Y.T.); (F.A.A.)
| | - Li Yan
- Department of Biostatistics & Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA;
| | - Ryusei Matsuyama
- Department of Gastroenterological Surgery, Yokohama City University Graduate School of Medicine, Yokohama 236-0004, Japan; (R.M.); (I.E.)
| | - Emese Zsiros
- Center for Immunotherapy, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA;
- Department of Gynecologic Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Takashi Ishikawa
- Department of Breast Surgery and Oncology, Tokyo Medical University, Tokyo 160-8402, Japan;
| | - Itaru Endo
- Department of Gastroenterological Surgery, Yokohama City University Graduate School of Medicine, Yokohama 236-0004, Japan; (R.M.); (I.E.)
| | - Kazuaki Takabe
- Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA; (M.O.); (M.A.); (Y.T.); (F.A.A.)
- Department of Gastroenterological Surgery, Yokohama City University Graduate School of Medicine, Yokohama 236-0004, Japan; (R.M.); (I.E.)
- Department of Breast Surgery and Oncology, Tokyo Medical University, Tokyo 160-8402, Japan;
- Department of Surgery, Jacobs School of Medicine and Biomedical Sciences, State University of New York, Buffalo, NY 14263, USA
- Division of Digestive and General Surgery, Niigata University Graduate School of Medical and Dental Sciences, Niigata 951-8520, Japan
- Department of Breast Surgery, Fukushima Medical University School of Medicine, Fukushima 960-1295, Japan
| |
Collapse
|
30
|
Grumish EL, Armstrong AR, Voigt RM, Forsyth CB, Bishehsari F. Alcohol-Induced Immune Dysregulation in the Colon Is Diurnally Variable. Visc Med 2020; 36:212-219. [PMID: 32775352 DOI: 10.1159/000507124] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 11/20/2019] [Accepted: 03/11/2020] [Indexed: 01/17/2023] Open
Abstract
Introduction Alcohol increases the risk of colon cancer. Colonic inflammation mediates the effects of alcohol on colon carcinogenesis. Circadian rhythm disruption enhances the alcohol's effect on colonic inflammation and cancer. Objective Here, we investigate the diurnal variation of lymphocyte infiltration in the colonic mucosa in response to alcohol. Methods Sixty C57BL6/J mice were fed a chow diet, and gavaged with alcohol at a specific time once per day for 3 consecutive days. Immunohistochemistry and immunofluorescence staining were used to quantify total, effector, and regulatory T cells in the colon. Student's t test, one-way ANOVA, and two-way ANOVA were used to determine significance. Results Following the alcohol binge, the composition of immune T cell subsets in the mouse colon was time-dependent. Alcohol did not alter the total number of CD3+ T cells. However, upon alcohol treatment, T-bet+ T helper 1 (Th1) cells appeared to dominate the T cell population following a reduction in Foxp3+ regulatory T cell (Treg) numbers. Depletion of Tregs was time-dependent, and their numbers were dramatically reduced when alcohol was administered during the rest phase. A reduction in Tregs significantly increased the Th1/Treg ratio, resulting in a more proinflammatory milieu. Conclusions Alcohol enhanced the proinflammatory profile in the colon mucosa, as demonstrated by a higher T-bet+/Foxp3+ ratio, especially during the rest phase. These findings may partly account for the interaction of circadian rhythm disruption with alcohol in colon inflammation and cancer.
Collapse
Affiliation(s)
- Eve Lauren Grumish
- Division of Gastroenterology, Department of Internal Medicine, Rush University Medical Center, Chicago, Illinois, USA
| | - Andrew R Armstrong
- Division of Gastroenterology, Department of Internal Medicine, Rush University Medical Center, Chicago, Illinois, USA
| | - Robin M Voigt
- Division of Gastroenterology, Department of Internal Medicine, Rush University Medical Center, Chicago, Illinois, USA
| | - Christopher B Forsyth
- Division of Gastroenterology, Department of Internal Medicine, Rush University Medical Center, Chicago, Illinois, USA
| | - Faraz Bishehsari
- Division of Gastroenterology, Department of Internal Medicine, Rush University Medical Center, Chicago, Illinois, USA
| |
Collapse
|
31
|
FoxP3 + T regulatory cells in cancer: Prognostic biomarkers and therapeutic targets. Cancer Lett 2020; 490:174-185. [PMID: 32721551 DOI: 10.1016/j.canlet.2020.07.022] [Citation(s) in RCA: 193] [Impact Index Per Article: 38.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/21/2020] [Revised: 06/28/2020] [Accepted: 07/16/2020] [Indexed: 12/19/2022]
Abstract
T Regulatory cells (Tregs) can have both protective and pathological roles. They maintain immune homeostasis and inhibit immune responses in various diseases, including cancer. Proportions of Tregs in the peripheral blood of some cancer patients increase by approximately two-fold, compared to those in healthy individuals. Tregs contribute to cancer development and progression by suppressing T effector cell functions, thereby compromising tumor killing and promoting tumor growth. Highly immunosuppressive Tregs express upregulated levels of the transcription factor, Forkhead box protein P3 (FoxP3). Elevated levels of FoxP3+ Tregs within the tumor microenvironment (TME) showed a positive correlation with poor prognosis in various cancer patients. Despite the success of immunotherapy, including the use of immune checkpoint inhibitors, a significant proportion of patients show low response rates as a result of primary or acquired resistance against therapy. Some of the mechanisms which underlie the development of therapy resistance are associated with Treg suppressive function. In this review, we describe Treg contribution to cancer development/progression, and the mechanisms of Treg-mediated immunosuppression. We discuss the prognostic significance of FoxP3+ Tregs in different cancers and their potential use as prognostic biomarkers. We also describe potential therapeutic strategies to target Tregs in combination with other types of immunotherapies aiming to overcome tumor resistance and improve clinical outcomes in cancer patients. Overall, understanding the prognostic significance of FoxP3+ Tregs in various cancers and their contribution to therapy resistance could help in the development of more effective targeted therapeutic strategies to enhance the clinical outcomes in cancer patients.
Collapse
|
32
|
Khadge S, Sharp JG, Thiele GM, McGuire TR, Talmadge JE. Fatty Acid Mediators in the Tumor Microenvironment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1259:125-153. [PMID: 32578175 DOI: 10.1007/978-3-030-43093-1_8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Academic Contribution Register] [Indexed: 02/06/2023]
Abstract
Patients with cancer frequently overexpress inflammatory cytokines with an associated neutrophilia both of which may be downregulated by diets with high omega-3 polyunsaturated fatty acids (ω-3 PUFA). The anti-inflammatory activity of dietary ω-3 PUFA has been suggested to have anticancer properties and to improve survival of cancer patients. Currently, the majority of dietary research efforts do not differentiate between obesity and dietary fatty acid consumption as mediators of inflammatory cell expansion and tumor microenvironmental infiltration, initiation, and progression. In this chapter, we discuss the relationships between dietary lipids, inflammation, neoplasia and strategies to regulate these relationships. We posit that dietary composition, notably the ratio of ω-3 vs. ω-6 PUFA, regulates tumor initiation and progression and the frequency and sites of metastasis that, together, impact overall survival (OS). We focus on three broad topics: first, the role of dietary lipids in chronic inflammation and tumor initiation, progression, and regression; second, lipid mediators linking inflammation and cancer; and third, dietary lipid regulation of murine and human tumor initiation, progression, and metastasis.
Collapse
Affiliation(s)
- Saraswoti Khadge
- Department of Pathology and Microbiology and Immunology, University of Nebraska Medical Center, Omaha, NE, USA.,Vanderbilt University, Nashville, TN, USA
| | - John Graham Sharp
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE, USA
| | - Geoffrey M Thiele
- Department of Pathology and Microbiology and Immunology, University of Nebraska Medical Center, Omaha, NE, USA.,Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA.,Veteran Affairs Nebraska-Western Iowa Health Care System, Omaha, NE, USA
| | - Timothy R McGuire
- Department of Pharmacy Practice, University of Nebraska Medical Center, Omaha, NE, USA
| | - James E Talmadge
- Department of Pathology and Microbiology and Immunology, University of Nebraska Medical Center, Omaha, NE, USA. .,Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA.
| |
Collapse
|
33
|
Li W, Wang W, Liao P, Song K, Zhu Z, Wang K, Liu Z, Zhang W, Xie S, He Y, Mcleod HL, Chen L. High levels of tumor-infiltrating lymphocytes showed better clinical outcomes in FOLFOX-treated gastric cancer patients. Pharmacogenomics 2020; 21:751-759. [PMID: 32615909 DOI: 10.2217/pgs-2019-0102] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/22/2022] Open
Abstract
Background: Tumor-infiltrating lymphocytes (TILs) and postoperative chemotherapeutics interact in the tumor micro-environment. This interaction has not been well investigated in gastric cancer. Materials & methods: A total of 129 patients were divided into high or low TILs based on the median number of positive CD3+ and FoxP3+ T cells, which was assessed by immunocytochemistry. Results: Cox regression analysis showed that the stage III disease with shorter overall survival was significant. The analysis showed that high numbers of CD3+ or FoxP3+ T cells have better clinical outcomes in FOLFOX-treated patients. Conclusion: High CD3+ and FoxP3+ T-cell infiltration was associated with better clinical outcomes in patients with gastric cancer treated with FOLFOX, suggesting TILs incorporated into algorithms to improve the therapeutic efficacy of optimal chemotherapy.
Collapse
Affiliation(s)
- Wei Li
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, 110 Xiangya Road, Changsha, Hunan, 410078, PR China.,Department of Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, 110 Xiangya Road, Changsha, Hunan, 410078, PR China.,Institude of National Clinical Research Center for Geriatric Disorders, 87 Xiangya Road, Changsha, Hunan, 410008, PR China
| | - Weili Wang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, 110 Xiangya Road, Changsha, Hunan, 410078, PR China.,Department of Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, 110 Xiangya Road, Changsha, Hunan, 410078, PR China.,Institude of National Clinical Research Center for Geriatric Disorders, 87 Xiangya Road, Changsha, Hunan, 410008, PR China
| | - Ping Liao
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, 110 Xiangya Road, Changsha, Hunan, 410078, PR China.,Department of Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, 110 Xiangya Road, Changsha, Hunan, 410078, PR China.,Institude of National Clinical Research Center for Geriatric Disorders, 87 Xiangya Road, Changsha, Hunan, 410008, PR China
| | - Kun Song
- Department of Gastrointestinal Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, 410078, PR China
| | - Zhanwei Zhu
- Institute of Medical Sciences, Xiangya Hospital, Central South University, Changsha, Hunan, 410078, PR China
| | - Kuansong Wang
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan, 410078, PR China
| | - Zhaoqian Liu
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, 110 Xiangya Road, Changsha, Hunan, 410078, PR China.,Department of Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, 110 Xiangya Road, Changsha, Hunan, 410078, PR China.,Institude of National Clinical Research Center for Geriatric Disorders, 87 Xiangya Road, Changsha, Hunan, 410008, PR China
| | - Wei Zhang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, 110 Xiangya Road, Changsha, Hunan, 410078, PR China.,Department of Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, 110 Xiangya Road, Changsha, Hunan, 410078, PR China.,Institude of National Clinical Research Center for Geriatric Disorders, 87 Xiangya Road, Changsha, Hunan, 410008, PR China
| | - Shangchen Xie
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, 110 Xiangya Road, Changsha, Hunan, 410078, PR China.,Department of Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, 110 Xiangya Road, Changsha, Hunan, 410078, PR China.,Institude of National Clinical Research Center for Geriatric Disorders, 87 Xiangya Road, Changsha, Hunan, 410008, PR China
| | - Yijing He
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, 110 Xiangya Road, Changsha, Hunan, 410078, PR China.,Department of Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, 110 Xiangya Road, Changsha, Hunan, 410078, PR China.,Institude of National Clinical Research Center for Geriatric Disorders, 87 Xiangya Road, Changsha, Hunan, 410008, PR China
| | - Howard L Mcleod
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, 110 Xiangya Road, Changsha, Hunan, 410078, PR China.,Department of Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, 110 Xiangya Road, Changsha, Hunan, 410078, PR China.,Institude of National Clinical Research Center for Geriatric Disorders, 87 Xiangya Road, Changsha, Hunan, 410008, PR China
| | - Ling Chen
- Department of Gastrointestinal Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, 410078, PR China
| |
Collapse
|
34
|
Chan LF, Sadahiro S, Suzuki T, Okada K, Miyakita H, Yamamoto S, Kajiwara H. Tissue-Infiltrating Lymphocytes as a Predictive Factor for Recurrence in Patients with Curatively Resected Colon Cancer: A Propensity Score Matching Analysis. Oncology 2020; 98:680-688. [PMID: 32526753 DOI: 10.1159/000506706] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/28/2020] [Accepted: 02/19/2020] [Indexed: 12/15/2022]
Abstract
BACKGROUND In patients with colorectal cancer, the rate of recurrence increases as the histologic stage progresses. However, the prediction of recurrence in individual patients is difficult. Many studies have reported on the relation between outcomes and tissue-infiltrating lymphocytes (TILs). The aim of our study was to clarify the relation between TILs and oncologic outcomes in patients with colon cancer using propensity score matching analysis. METHODS The study group comprised 513 patients with colon cancer who received curative resection. By using propensity score matching for sex, age, tumor location, T stage, N stage, histologic type, and adjuvant therapy as conventional prognostic factors, 61 patients with recurrence and 61 patients with no recurrence were selected. Hematoxylin-eosin staining and immunohistochemical staining using CD3, CD8, CD4, and FoxP3 were performed for lymphocytes in the primary tissue. The results were evaluated separately in the whole tumor, the central part, and the invasive margin. RESULTS The median follow-up period was 53 months. Among the 513 patients, 70 had recurrence and 443 had no recurrence. In the comparison of outcomes between the 61 patients with recurrence and the 61 patients with no recurrence, univariate analysis showed that the disease-free survival rate was significantly higher among the patients with positive TILs in the whole tumor and in the invasive margin (p = 0.016 and p = 0.012, respectively) and with CD8+ cells in the central part (p = 0.039) than among those with negative results. A multivariate analysis showed that TILs in the invasive margin (hazard ratio 1.81; 95% confidence interval, 1.03-3.05; p = 0.037) and CD8+ cell density in the central part (hazard ratio 1.76; 95% confidence interval, 1.07-2.93; p = 0.023) were prognostic factors that were independent from conventional prognostic factors. CONCLUSIONS In patients with curatively resected colon cancer, TILs in the invasive margin and CD8+ cell density in the central part may be prognostic factors suggesting host antitumor immune response.
Collapse
Affiliation(s)
- Lin Fung Chan
- Department of Surgery, School of Medicine, Tokai University, Isehara, Japan
| | - Sotaro Sadahiro
- Department of Surgery, School of Medicine, Tokai University, Isehara, Japan,
| | - Toshiyuki Suzuki
- Department of Surgery, School of Medicine, Tokai University, Isehara, Japan
| | - Kazutake Okada
- Department of Surgery, School of Medicine, Tokai University, Isehara, Japan
| | - Hiroshi Miyakita
- Department of Surgery, School of Medicine, Tokai University, Isehara, Japan
| | - Seiichiro Yamamoto
- Department of Surgery, School of Medicine, Tokai University, Isehara, Japan
| | - Hiroshi Kajiwara
- Department of Pathology, School of Medicine, Tokai University, Isehara, Japan
| |
Collapse
|
35
|
Xu W, Wu X, Wang X, Yu S, Xu G, Xiong J, Zhang J, Sang X, Zheng Y, Liu W. Prognostic Significance of the Preoperative Lymphocyte to Monocyte Ratio in Patients with Gallbladder Carcinoma. Cancer Manag Res 2020; 12:3271-3283. [PMID: 32494191 PMCID: PMC7227785 DOI: 10.2147/cmar.s243326] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/22/2019] [Accepted: 04/23/2020] [Indexed: 12/26/2022] Open
Abstract
Background This study was designed to investigate the prognostic value of the lymphocyte to monocyte ratio (LMR) in patients with gallbladder carcinoma (GBC). Patients and Methods We retrospectively enrolled 154 consecutive GBC patients from 2005 to 2017 in this study. The LMR of preoperative blood samples was calculated by dividing the lymphocyte count by the monocyte count. A receiver operating characteristic (ROC) curve was employed to identify the optimal cut-off value of the LMR in the determination of overall survival (OS). The Kaplan–Meier method was utilized to assess OS, and the Log rank test was employed to compare survival differences. Univariate and multivariate Cox regression analyses were conducted to detect independent prognostic indicators. Results The optimal cut-off point for the LMR was 4.76 according to the ROC curve. Patients ≤60 years old with an LMR ≤4.76 experienced significantly worse OS than those with an LMR >4.76 (hazard ratio (HR): 0.399, 95% confidence interval (CI): 0.265–0.602, P<0.001); however, the prognostic value of the LMR was not determined in patients >60 years old or among the entire study cohort (both P>0.05). Significantly poorer OS was observed in patients >60 years with an LMR ≤4.21 compared to those with an LMR >4.21 (HR: 1.830, 95% CI: 1.129–2.967, P=0.014). Multivariate Cox regression analysis indicated that both the high and low LMR cut-off values were independent risk factors for OS (HR: 0.272, 95% CI: 0.105–0.704, P=0.007; HR: 0.544, 95% CI: 0.330–0.895, P=0.017). Conclusion The LMR is an independent prognostic indicator for GBC patients, the cut-off value of which is age dependent.
Collapse
Affiliation(s)
- Weiyu Xu
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, People's Republic of China
| | - Xiaoqian Wu
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), Beijing 100730, People's Republic of China
| | - Xuezhu Wang
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), Beijing 100730, People's Republic of China
| | - Si Yu
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), Beijing 100730, People's Republic of China
| | - Gang Xu
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), Beijing 100730, People's Republic of China
| | - Jianping Xiong
- Department of Interventional Radiology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, People's Republic of China
| | - Junwei Zhang
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), Beijing 100730, People's Republic of China
| | - Xinting Sang
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), Beijing 100730, People's Republic of China
| | - Yongchang Zheng
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), Beijing 100730, People's Republic of China
| | - Wei Liu
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, People's Republic of China
| |
Collapse
|
36
|
Alternative macrophage polarisation associated with resistance to anti-PD1 blockade is possibly supported by the splicing of FKBP51 immunophilin in melanoma patients. Br J Cancer 2020; 122:1782-1790. [PMID: 32317723 PMCID: PMC7283486 DOI: 10.1038/s41416-020-0840-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 11/19/2019] [Revised: 02/25/2020] [Accepted: 03/24/2020] [Indexed: 02/08/2023] Open
Abstract
Background FKBP51 immunophilin is abundantly expressed by immune cells. Co-inhibitory immune receptor signalling generates the splicing isoform FKBP51s. Tregs stained by FKBP51s are increased in melanoma patients and their counts are associated with anti-CTLA-4 response. An expansion of FKBP51s+PD-L1+ monocytes was measured in a group of non-responding patients to anti-CTLA-4. The aim of this work was to confirm the predictive value of response of FKBP51s+Tregs in a cohort of patients undergoing anti-PD1 treatment and shed light on a monocyte subset co-expressing PD-L1/FKBP51s. Methods Co-cultures of organoids and autologous lymphocytes were used to confirm that tumour T-cell interaction can induce FKBP51s. PBMC immunophenotype and flow cytometry served to assess and monitor FKBP51s+Treg and FKBP51s+PD-L1+ monocytes in 22 advanced melanoma patients treated with anti-PD1. Silencing and overexpression of FKBP51s in human macrophages served to address the protein role in the tolerant macrophages’ behaviour. Results FKBP51s+Tregs count was increased in responders and had a prognostic value. Non-responders showed an early increase in FKBP51s+ PD-L1+ monocytes during anti-PD1 treatment. Manipulation of FKBP51s modulated the macrophage–phenotype, with forced protein expression promoting aspects associated with tolerance. Conclusions FKBP51s may guide in the selection and monitoring of melanoma patient candidates to immune-checkpoint-targeted therapy. Manipulation of FKBP51s may overcome resistance.
Collapse
|
37
|
Fedele V, Melisi D. Permissive State of EMT: The Role of Immune Cell Compartment. Front Oncol 2020; 10:587. [PMID: 32391271 PMCID: PMC7189417 DOI: 10.3389/fonc.2020.00587] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/14/2020] [Accepted: 03/31/2020] [Indexed: 12/13/2022] Open
Abstract
The Epithelial to Mesenchymal Transition (EMT) type 3 is a reversible dynamic process recognized as a major determinant of the metastatic event, although many questions regarding its role throughout this process remain unanswered. The ability of cancer cells to migrate and colonize distant organs is a key aspect of tumor progression and evolution, requiring constant tumor cells and tumor microenvironment (TME) changes, as well as constant changes affecting the cross-talk between the two aforementioned compartments. Alterations affecting tumor cells, such as transcription factors, trans-membrane receptors, chromatin remodeling complexes and metabolic pathways, leading to the disappearance of the epithelial phenotype and concomitant gaining of the undifferentiated mesenchymal phenotype are undoubtedly major players of the EMT process. However, several lines of evidence point out toward a more critical role of TME composition in creating an “EMT-permissive state.” The “EMT-permissive state” consists in changes affecting physical and biochemical properties (i.e., stiffness and/or hypoxia) as well as changes of the TME cellular component (i.e., immune-cell, blood vessel, lymphatic vessels, fibroblasts, and fat cells) that favor and induce the epithelial mesenchymal transition. In this mini review, we will discuss the role of the tumor microenvironment cellular component that are involved in supporting the EMT, with particular emphasis on the immune-inflammatory cells component.
Collapse
Affiliation(s)
- Vita Fedele
- Digestive Molecular Clinical Oncology Research Unit, Section of Medical Oncology, Università degli Studi di Verona, Verona, Italy
| | - Davide Melisi
- Digestive Molecular Clinical Oncology Research Unit, Section of Medical Oncology, Università degli Studi di Verona, Verona, Italy
| |
Collapse
|
38
|
Rudolf J, Büttner-Herold M, Erlenbach-Wünsch K, Posselt R, Jessberger J, Haderlein M, Hecht M, Hartmann A, Fietkau R, Distel L. Regulatory T cells and cytotoxic T cells close to the epithelial-stromal interface are associated with a favorable prognosis. Oncoimmunology 2020; 9:1746149. [PMID: 32363115 PMCID: PMC7185207 DOI: 10.1080/2162402x.2020.1746149] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 10/10/2019] [Revised: 01/09/2020] [Accepted: 02/29/2020] [Indexed: 01/09/2023] Open
Abstract
Cytotoxic T cells and regulatory T cells play a crucial role in the outcome of cancer patients. Besides the density of these cells, it was shown recently that the spatial distribution is equally important. Here, we specifically analyzed the spatial distribution of these T cell subtypes at the epithelial-stromal interface in a rectal cancer cohort and its relevance for prognosis. We studied a cohort of 191 patients with advanced rectal cancer treated by radiochemotherapy (RCT). Tissue microarrays were immunohistochemical double-stained by FoxP3+ and CD+. Cell densities were analyzed in the stromal and epithelial compartment. Additionally, an image analysis software calculated the distances of lymphocytes to the epithelial-stromal interface (ESI). CD8+ and FoxP3+ cell counts decreased clearly after RCT with the decrease of FoxP3+ being more pronounced than of CD8+ cells. In the invasive front, short distances of the ESI to CD8+ and to FoxP3+ cells were associated with improved overall survival. Cell counts in the stromal compartment had no influence on prognosis. No correlation between stromal and epithelial lymphocyte densities was observed. The distance of epithelial-stromal interface to CD8+ and FoxP3+ cells was more accurate in predicting prognosis in the stromal compartment of rectal cancer patients than mere cell counts and could thereby be means of better stratifying patients for therapy. This observation will have to be validated in future prospective studies with regard to other tumor entities and its implications for the responsiveness of tumors to new therapeutic modalities.
Collapse
Affiliation(s)
- Julian Rudolf
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Maike Büttner-Herold
- Department of Nephropathology, Institute of Pathology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Katharina Erlenbach-Wünsch
- Institute of Pathology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Rebecca Posselt
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Jonas Jessberger
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Marlen Haderlein
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Markus Hecht
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Arndt Hartmann
- Institute of Pathology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Rainer Fietkau
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Luitpold Distel
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
39
|
The Prognostic Implications of Tumor Infiltrating Lymphocytes in Colorectal Cancer: A Systematic Review and Meta-Analysis. Sci Rep 2020; 10:3360. [PMID: 32099066 PMCID: PMC7042281 DOI: 10.1038/s41598-020-60255-4] [Citation(s) in RCA: 181] [Impact Index Per Article: 36.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 03/05/2019] [Accepted: 02/03/2020] [Indexed: 12/17/2022] Open
Abstract
Tumor-infiltrating lymphocytes (TILs) are an important histopathologic feature of colorectal cancer that confer prognostic information. Previous clinical and epidemiologic studies have found that the presence and quantification of tumor-infiltrating lymphocytes are significantly associated with disease-specific and overall survival in colorectal cancer. We performed a systematic review and meta-analysis, establishing pooled estimates for survival outcomes based on the presence of TILs in colon cancer. PubMed (Medline), Embase, Cochrane Library, Web of Science, and ClinicalTrials.gov were searched from inception to April 2017. Studies were included, in which the prognostic significance of intratumoral tumor infiltrating lymphocytes, as well as subsets of CD3, CD8, FOXP3, CD45R0 lymphocytes, were determined within the solid tumor center, the invasive margin, and tumor stroma. Random-effects models were calculated to estimated summary effects using hazard ratios. Forty-three relevant studies describing 21,015 patients were included in our meta-analysis. The results demonstrate that high levels of generalized TILS as compared to low levels had an improved overall survival (OS) with a HR of 0.65 (p = <0.01). In addition, histologically localized CD3+ T-cells at the tumor center were significantly associated with better disease-free survival (HR = 0.46, 95% CI 0.36–0.61, p = 0.05), and CD3 + cells at the invasive margin were associated with improved disease-free survival (HR = 0.57, 95% CI 0.38–0.86, p = 0.05). CD8+ T-cells at the tumor center had statistically significant prognostic value on cancer-specific survival and overall survival with HRs of 0.65 (p = 0.02) and 0.71 (p < 0.01), respectively. Lastly, FOXP3+ T-cells at the tumor center were associated with improved prognosis for cancer-specific survival (HR = 0.65, p < 0.01) and overall survival (HR = 0.70, p < 0.01). These findings suggest that TILs and specific TIL subsets serve as prognostic biomarkers for colorectal cancer.
Collapse
|
40
|
Toor SM, Murshed K, Al-Dhaheri M, Khawar M, Abu Nada M, Elkord E. Immune Checkpoints in Circulating and Tumor-Infiltrating CD4 + T Cell Subsets in Colorectal Cancer Patients. Front Immunol 2019; 10:2936. [PMID: 31921188 PMCID: PMC6928042 DOI: 10.3389/fimmu.2019.02936] [Citation(s) in RCA: 105] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/20/2019] [Accepted: 11/29/2019] [Indexed: 12/22/2022] Open
Abstract
Blockade of inhibitory immune checkpoints (ICs) is a promising therapeutic approach; however, it has shown limited success in some cancers including colorectal cancer (CRC). The tumor microenvironment (TME) is largely responsible for response to therapy, and its constituents may provide robust biomarkers for successful immunotherapeutic approaches. In this study, we performed phenotypical characterization and critical analyses of key inhibitory ICs and T regulatory cell (Treg)-related markers on CD4+ T cell subsets in CRC patients, and compared with normal colon tissues and peripheral blood from the same patients. We also investigated correlations between the levels of different CD4+ T cell subsets and the clinicopathologic features including disease stage and tumor budding. We found a significant increase in the levels of CD4+FoxP3+Helios+ T cells, which represent potentially highly immunosuppressive Tregs, in the CRC TME. Additionally, tumor-infiltrating CD4+ T cells upregulated programmed cell death protein-1 (PD-1), cytotoxic T-lymphocyte-associated protein-4 (CTLA-4), T cell immunoglobulin and mucin domain-3 (TIM-3) and lymphocyte-activation gene 3 (LAG-3). We also characterized the expression of PD-1, CTLA-4, TIM-3, and LAG-3 on different CD4+FoxP3−/+Helios−/+ T cell subsets. Interestingly, we found that CTLA-4, TIM-3, and LAG-3 were mainly co-expressed on FoxP3+Helios+ Tregs in the TME. Additionally, FoxP3high Tregs expressed higher levels of Helios, CTLA-4 and TIM-3 than FoxP3low T cells. These results highlight the significance of Tregs in the CRC TME and suggest that Tregs may hamper response to IC blockade in CRC patients, but effects of different IC inhibition regimes on Treg levels or activity warrants further investigations. We also found that CD4+CTLA-4+ T cells in circulation are increased in patients with advanced disease stage. This study simultaneously provides important insights into the differential levels of CD4+ T cell subpopulations and IC expression in CRC TME, compared to periphery and associations with clinicopathologic features, which could be used as potential biomarkers for CRC progression and response to therapy.
Collapse
Affiliation(s)
- Salman M Toor
- Cancer Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar
| | - Khaled Murshed
- Department of Pathology, Hamad Medical Corporation, Doha, Qatar
| | | | - Mahwish Khawar
- Department of Surgery, Hamad Medical Corporation, Doha, Qatar
| | | | - Eyad Elkord
- Cancer Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar.,Biomedical Research Centre, School of Science, Engineering and Environment, University of Salford, Salford, United Kingdom
| |
Collapse
|
41
|
Alexander PG, McMillan DC, Park JH. The local inflammatory response in colorectal cancer - Type, location or density? A systematic review and meta-analysis. Cancer Treat Rev 2019; 83:101949. [PMID: 31869737 DOI: 10.1016/j.ctrv.2019.101949] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 11/01/2019] [Revised: 12/04/2019] [Accepted: 12/05/2019] [Indexed: 02/06/2023]
Abstract
INTRODUCTION The host anti-tumour inflammatory response is a strong prognostic indicator, and tumour infiltrating lymphocytes (TILs) are believed to have a complimentary role alongside TNM assessment in dictating future management. However, there is wide disagreement regarding the most efficacious and cost-effective method of assessment. METHODS A comprehensive literature search was performed of EMBASE, MedLine and PubMed as well as an assessment of references to identify all relevant studies relating to the assessment of the peri-tumoural inflammatory response or TILs and prognosis in colorectal cancer (CRC). A meta-analysis was performed of 67 studies meeting the REMARK criteria using RevMan software. RESULTS Intratumoural assessment of both CD3 and CD8 in CRC were significant for disease-free survival (DFS) (combined HRs 0.46; 95%CI: 0.39-0.54 and 0.54; 95%CI: 0.45-0.65), as well as overall survival (OS) and disease-specific survival (DSS). The same was true for assessment of CD3 and CD8 at the invasive margin (DFS: combined HRs 0.45; 95%CI: 0.33-0.61 and 0.51; 95%CI: 0.41-0.62). However, similar fixed effects summaries were also observed for H&E-based methods, like Klintrup-Makinen grade (DFS: HR 0.62; 95%CI: 0.43-0.88). Furthermore, inflammatory assessments were independent of MSI status. CONCLUSION The evidence suggests that it is the density of a co-ordinated local inflammatory infiltrate that confers survival benefit, rather than any individual immune cell subtype. Furthermore, the location of individual cells within the tumour microenvironment does not appear to influence survival. The authors advocate a standardised assessment of the local inflammatory response, but caution against emphasizing the importance of any individual immune cell subtype.
Collapse
Affiliation(s)
| | | | - James H Park
- School of Medicine, University of Glasgow, Glasgow, United Kingdom
| |
Collapse
|
42
|
The Gastrointestinal Tumor Microenvironment: An Updated Biological and Clinical Perspective. JOURNAL OF ONCOLOGY 2019; 2019:6240505. [PMID: 31885581 PMCID: PMC6893275 DOI: 10.1155/2019/6240505] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Academic Contribution Register] [Received: 05/31/2019] [Accepted: 10/30/2019] [Indexed: 12/24/2022]
Abstract
Gastrointestinal cancers are still responsible for high numbers of cancer-related deaths despite advances in therapy. Tumor-associated cells play a key role in tumor biology, by supporting or halting tumor development through the production of extracellular matrix, growth factors, cytokines, and extracellular vesicles. Here, we review the roles of these tumor-associated cells in the initiation, angiogenesis, immune modulation, and resistance to therapy of gastrointestinal cancers. We also discuss novel diagnostic and therapeutic strategies directed at tumor-associated cells and their potential benefits for the survival of these patients.
Collapse
|
43
|
Caraglia M, Correale P, Giannicola R, Staropoli N, Botta C, Pastina P, Nesci A, Caporlingua N, Francini E, Ridolfi L, Mini E, Roviello G, Ciliberto D, Agostino RM, Strangio A, Azzarello D, Nardone V, Falzea A, Cappabianca S, Bocchetti M, D'Arrigo G, Tripepi G, Tassone P, Addeo R, Giordano A, Pirtoli L, Francini G, Tagliaferri P. GOLFIG Chemo-Immunotherapy in Metastatic Colorectal Cancer Patients. A Critical Review on a Long-Lasting Follow-Up. Front Oncol 2019; 9:1102. [PMID: 31781481 PMCID: PMC6857002 DOI: 10.3389/fonc.2019.01102] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/21/2019] [Accepted: 10/07/2019] [Indexed: 01/19/2023] Open
Abstract
Background: GOLFIG is a chemo-immunotherapy regimen established in preclinical models that combines gemcitabine + FOLFOX (fluoropyrimidine backbone coupled to oxaliplatin) poly-chemotherapy with low-dose s. c. recombinant interleukin-2 (rIL-2) and granulocyte-macrophage colony stimulating factor (GM-CSF). Promising antitumor effects in metastatic colorectal cancer (mCRC) patients were obtained in previous phase II and III trials. Here we report the results of 15 years of follow-up. Methods: This is a multi-institutional retrospective analysis including 179 mCRC patients receiving GOLFIG regimen between June 2002 and June 2018. Sixty-two of them received the treatment as frontline (enrolled in the GOLFIG-2 phase III trial) and 117 as second/third line (49 enrolled in the GOLFIG-1 phase II trial and 68 as compassionate use). One hundred twelve patients showed a primary left side and 67 a primary right side; K/N-ras mutational status was available in 74 cases, and an activating mutation was detected in 33. Kaplan-Meier and Cox regression analyses were carried out to relate PFS and OS with different parameters. Results: Overall, we recorded a mean PFS and OS of 15.28 (95% CI: 10.36-20.20) and 24.6 (95% CI: 19.07-30.14) months, respectively, with 14 patients surviving free of progression for 10 years. This regimen, in our updated survey of the GOLFIG-2 trial, confirmed superiority over FOLFOX in terms of PFS (hazard ratio (HR) = 0.58, p = 0.006) with a trend to a longer OS (HR = 0.69, P = 0.06) in the first line. Our analysis also confirmed significant antitumor activity in pre-treated patients, reporting a mean PFS and OS of 12.55 (95% CI: 7.19-17.9) and 20.28 (95% CI: 14.4-26.13) months, respectively. Immune-related adverse events (irAEs) were recorded in 24% of the cases and were related to a longer survival (HR = 0.36; P = 0.0001). Finally, patients' outcome was not correlated to sex, sidedness, and MT-K/N-ras. Conclusions: The GOLFIG regimen is a reliable underestimated therapeutic option in pre-treated mCRC patients and offers a strong rationale to design further trials.
Collapse
Affiliation(s)
- Michele Caraglia
- Department of Precision Medicine, University of Campania "L. Vanvitelli", Naples, Italy.,Biogem Scarl, Institute of Genetic Research, Laboratory of Precision and Molecular Oncology, Ariano Irpino, Italy
| | - Pierpaolo Correale
- Medical Oncology Unit, "Bianchi-Melacrino-Morelli" Grand Metropolitan Hospital, Reggio Calabria, Italy
| | - Rocco Giannicola
- Medical Oncology Unit, "Bianchi-Melacrino-Morelli" Grand Metropolitan Hospital, Reggio Calabria, Italy
| | - Nicoletta Staropoli
- Medical Oncology Unit, Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy
| | - Cirino Botta
- Medical Oncology Unit, Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy
| | | | - Antonello Nesci
- Unit of Pharmacy, Section of Anti-blastic Drugs, "Bianchi-Melacrino-Morelli" Grand Metropolitan Hospital, Reggio Calabria, Italy
| | - Nadia Caporlingua
- Unit of Pharmacy, Section of Anti-blastic Drugs, "Bianchi-Melacrino-Morelli" Grand Metropolitan Hospital, Reggio Calabria, Italy
| | | | - Laura Ridolfi
- Immunotherapy, Cell Therapy and Biobank, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori IRCCS, Meldola, Italy
| | - Enrico Mini
- Section of Clinical Pharmacology and Oncology, Department of Health Sciences, School of Medicine/Translational Oncology Unit, Careggi University Hospital, University of Florence, Florence, Italy
| | - Giandomenico Roviello
- Section of Clinical Pharmacology and Oncology, Department of Health Sciences, School of Medicine/Translational Oncology Unit, Careggi University Hospital, University of Florence, Florence, Italy
| | - Domenico Ciliberto
- Medical Oncology Unit, Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy
| | - Rita Maria Agostino
- Medical Oncology Unit, "Bianchi-Melacrino-Morelli" Grand Metropolitan Hospital, Reggio Calabria, Italy
| | - Alessandra Strangio
- Medical Oncology Unit, "Bianchi-Melacrino-Morelli" Grand Metropolitan Hospital, Reggio Calabria, Italy
| | - Domenico Azzarello
- Medical Oncology Unit, "Bianchi-Melacrino-Morelli" Grand Metropolitan Hospital, Reggio Calabria, Italy
| | - Valerio Nardone
- Radiation Oncology Unit, Siena University Hospital, Siena, Italy
| | - Antonella Falzea
- Medical Oncology Unit, "Bianchi-Melacrino-Morelli" Grand Metropolitan Hospital, Reggio Calabria, Italy
| | - Salvatore Cappabianca
- Department of Precision Medicine, University of Campania "L. Vanvitelli", Naples, Italy.,Biogem Scarl, Institute of Genetic Research, Laboratory of Precision and Molecular Oncology, Ariano Irpino, Italy
| | - Marco Bocchetti
- Department of Precision Medicine, University of Campania "L. Vanvitelli", Naples, Italy.,Biogem Scarl, Institute of Genetic Research, Laboratory of Precision and Molecular Oncology, Ariano Irpino, Italy
| | - Graziella D'Arrigo
- Statistical Unit, IFC-CNR (CNR), Grand Metropolitan Hospital-IFC, Reggio Calabria, Italy
| | - Giovanni Tripepi
- Statistical Unit, IFC-CNR (CNR), Grand Metropolitan Hospital-IFC, Reggio Calabria, Italy
| | - Pierfrancesco Tassone
- Medical Oncology Unit, Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy
| | - Raffaele Addeo
- Oncology Unit, Day Hospital, San Giovanni di Dio Hospital, ASL Napoles 2 Nord, Frattamaggiore, Italy
| | - Antonio Giordano
- Sbarro Institute for Cancer Research and Molecular Medicine and Center of Biotechnology, College of Science and Technology, Temple University, Philadelphia, PA, United States.,Department of Medical Biotechnology, University of Siena, Siena, Italy
| | - Luigi Pirtoli
- Sbarro Institute for Cancer Research and Molecular Medicine and Center of Biotechnology, College of Science and Technology, Temple University, Philadelphia, PA, United States
| | - Guido Francini
- Medical Oncology Unit, Siena University Hospital, Siena, Italy
| | - Pierosandro Tagliaferri
- Medical Oncology Unit, Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy
| |
Collapse
|
44
|
Bishehsari F, Engen PA, Voigt RM, Swanson G, Shaikh M, Wilber S, Naqib A, Green SJ, Shetuni B, Forsyth CB, Saadalla A, Osman A, Hamaker BR, Keshavarzian A, Khazaie K. Abnormal Eating Patterns Cause Circadian Disruption and Promote Alcohol-Associated Colon Carcinogenesis. Cell Mol Gastroenterol Hepatol 2019; 9:219-237. [PMID: 31689559 PMCID: PMC6957855 DOI: 10.1016/j.jcmgh.2019.10.011] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 03/18/2019] [Revised: 10/25/2019] [Accepted: 10/28/2019] [Indexed: 12/13/2022]
Abstract
BACKGROUND & AIMS Alcohol intake with circadian rhythm disruption (CRD) increases colon cancer risk. We hypothesized that eating during or around physiologic rest time, a common habit in modern society, causes CRD and investigated the mechanisms by which it promotes alcohol-associated colon carcinogenesis. METHODS The effect of feeding time on CRD was assessed using B6 mice expressing a fusion protein of PERIOD2 and LUCIFERASE (PER2::LUC) were used to model colon polyposis and to assess the effects of feeding schedules, alcohol consumption, and prebiotic treatment on microbiota composition, short-chain fatty acid levels, colon inflammation, and cancer risk. The relationship between butyrate signaling and a proinflammatory profile was assessed by inactivating the butyrate receptor GPR109A. RESULTS Eating at rest (wrong-time eating [WTE]) shifted the phase of the colon rhythm in PER2::LUC mice. In TS4Cre × APClox468 mice, a combination of WTE and alcohol exposure (WTE + alcohol) decreased the levels of short-chain fatty acid-producing bacteria and of butyrate, reduced colonic densities of regulatory T cells, induced a proinflammatory profile characterized by hyperpermeability and an increased mucosal T-helper cell 17/regulatory T cell ratio, and promoted colorectal cancer. Prebiotic treatment improved the mucosal inflammatory profile and attenuated inflammation and cancer. WTE + alcohol-induced polyposis was associated with increased signal transducer and activator of transcription 3 expression. Decreased butyrate signaling activated the epithelial signal transducer and activator of transcription 3 in vitro. The relationship between butyrate signaling and a proinflammatory profile was confirmed in human colorectal cancers using The Cancer Genome Atlas. CONCLUSIONS Abnormal timing of food intake caused CRD and interacts with alcohol consumption to promote colon carcinogenesis by inducing a protumorigenic inflammatory profile driven by changes in the colon microbiota and butyrate signaling. Accession number of repository for microbiota sequence data: raw FASTQ data were deposited in the NCBI Sequence Read Archive under project PRJNA523141.
Collapse
Affiliation(s)
- Faraz Bishehsari
- Department of Internal Medicine, Division of Gastroenterology, Rush University Medical Center, Chicago, Illinois.
| | - Phillip A Engen
- Department of Internal Medicine, Division of Gastroenterology, Rush University Medical Center, Chicago, Illinois
| | - Robin M Voigt
- Department of Internal Medicine, Division of Gastroenterology, Rush University Medical Center, Chicago, Illinois
| | - Garth Swanson
- Department of Internal Medicine, Division of Gastroenterology, Rush University Medical Center, Chicago, Illinois
| | - Maliha Shaikh
- Department of Internal Medicine, Division of Gastroenterology, Rush University Medical Center, Chicago, Illinois
| | - Sherry Wilber
- Department of Internal Medicine, Division of Gastroenterology, Rush University Medical Center, Chicago, Illinois
| | - Ankur Naqib
- Department of Internal Medicine, Division of Gastroenterology, Rush University Medical Center, Chicago, Illinois; Sequencing Core, Research Resources Center, University of Illinois at Chicago, Chicago, Illinois
| | - Stefan J Green
- Sequencing Core, Research Resources Center, University of Illinois at Chicago, Chicago, Illinois; Department of Biological Sciences, University of Illinois at Chicago, Chicago, Illinois
| | - Brandon Shetuni
- Northwestern Medicine, Central DuPage Hospital, Winfield, Illinois
| | - Christopher B Forsyth
- Department of Internal Medicine, Division of Gastroenterology, Rush University Medical Center, Chicago, Illinois
| | | | - Abu Osman
- Department of Immunology, Mayo Clinic, Rochester, Minnesota
| | - Bruce R Hamaker
- Whistler Center for Carbohydrate Research, Department of Food Science, Purdue University, West Lafayette, Indiana
| | - Ali Keshavarzian
- Department of Internal Medicine, Division of Gastroenterology, Rush University Medical Center, Chicago, Illinois; Department of Physiology, Rush University Medical Center, Chicago, Illinois; Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | | |
Collapse
|
45
|
Franke AJ, Skelton WP, Starr JS, Parekh H, Lee JJ, Overman MJ, Allegra C, George TJ. Immunotherapy for Colorectal Cancer: A Review of Current and Novel Therapeutic Approaches. J Natl Cancer Inst 2019; 111:1131-1141. [PMID: 31322663 PMCID: PMC6855933 DOI: 10.1093/jnci/djz093] [Citation(s) in RCA: 102] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/01/2018] [Revised: 04/02/2019] [Accepted: 05/09/2019] [Indexed: 12/22/2022] Open
Abstract
Colorectal cancer (CRC) remains a leading cause of cancer-related deaths in the United States. Although immunotherapy has dramatically changed the landscape of treatment for many advanced cancers, the benefit in CRC has thus far been limited to patients with microsatellite instability high (MSI-H):DNA mismatch repair-deficient (dMMR) tumors. Recent studies in the refractory CRC setting have led to US Food and Drug Administration approvals for pembrolizumab as well as nivolumab (with or without ipilimumab) for tumors harboring an MSI-H:dMMR molecular profile. Several randomized controlled trials are underway to move immunotherapy into the frontline for metastatic cancer (with or without chemotherapy) and the adjuvant setting. Awareness of these studies is critical given the relatively low incidence (approximately 3%-5%) of MSI-H:dMMR in advanced or metastatic CRC to support study completion, because the results could be potentially practice changing. The real challenge in this disease is related to demonstrating the benefit of immunotherapy for the vast majority of patients with CRC not harboring MSI-H:dMMR. Given the rapid pace of scientific changes, this article provides a narrative review regarding the current landscape of immunotherapy for CRC. Particular attention is paid to the currently available data that inform today's clinical practice along with upcoming randomized controlled trials that may soon dramatically change the treatment landscape for CRC.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Thomas J George
- Correspondence to: Thomas J. George, MD, FACP, Department of Medicine, Division of Hematology & Oncology, University of Florida, PO Box 100278, 1600 SW Archer Rd, Gainesville, FL 32610–0278 (e-mail: )
| |
Collapse
|
46
|
Echarti A, Hecht M, Büttner-Herold M, Haderlein M, Hartmann A, Fietkau R, Distel L. CD8+ and Regulatory T cells Differentiate Tumor Immune Phenotypes and Predict Survival in Locally Advanced Head and Neck Cancer. Cancers (Basel) 2019; 11:cancers11091398. [PMID: 31546872 PMCID: PMC6769847 DOI: 10.3390/cancers11091398] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/01/2019] [Revised: 09/12/2019] [Accepted: 09/15/2019] [Indexed: 12/31/2022] Open
Abstract
Background: The tumor immune status “inflamed”, “immune excluded”, and “desert” might serve as a predictive parameter. We studied these three cancer immune phenotypes while using a simple immunohistochemical algorithm. Methods: Pre-treatment tissue samples of 280 patients with locally advanced HNSCC treated with radiochemotherapy were analyzed. A double staining of CD8+ cytotoxic T cells (CTL) and FoxP3+ (Treg) was performed and the cell density was evaluated in the intraepithelial and stromal compartment of the tumor. Results: The classification of tumors as “immune desert” when stromal CTL were ≤ 50 cells/mm2, “inflamed” when intraepithelial CTL were > 500 cells/mm2, and as “excluded” when neither of these definitions met these cut off values allowed the best discrimination regarding overall survival. These groups had median OS periods of 37, 61, and 85 months, respectively. In “immune desert” and “immune excluded” tumors high Treg tended to worsen OS, but in “inflamed” tumors high Treg clearly improved OS. Conclusions: We propose that, in locally advanced HNSCC, the tumor immune state “inflamed”, “immune excluded”, and “immune desert” can be defined by intraepithelial and stromal CTL. Tregs can further subdivide these groups. The opposing effects of Tregs in the different groups might be the reason for the inconsistency of Tregs prognostic values published earlier.
Collapse
Affiliation(s)
- Alessia Echarti
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, D-91054 Erlangen, Germany.
| | - Markus Hecht
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, D-91054 Erlangen, Germany.
| | - Maike Büttner-Herold
- Department of Nephropathology, Institute of Pathology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, D-91054 Erlangen, Germany.
| | - Marlen Haderlein
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, D-91054 Erlangen, Germany.
| | - Arndt Hartmann
- Institute of Pathology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany.
| | - Rainer Fietkau
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, D-91054 Erlangen, Germany.
| | - Luitpold Distel
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, D-91054 Erlangen, Germany.
| |
Collapse
|
47
|
Abolarinwa BA, Ibrahim RB, Huang YH. Conceptual Development of Immunotherapeutic Approaches to Gastrointestinal Cancer. Int J Mol Sci 2019; 20:E4624. [PMID: 31540435 PMCID: PMC6769557 DOI: 10.3390/ijms20184624] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/08/2019] [Revised: 09/14/2019] [Accepted: 09/16/2019] [Indexed: 02/07/2023] Open
Abstract
Gastrointestinal (GI) cancer is one of the common causes of cancer-related death worldwide. Chemotherapy and/or immunotherapy are the current treatments, but some patients do not derive clinical benefits. Recently, studies from cancer molecular subtyping have revealed that tumor molecular biomarkers may predict the immunotherapeutic response of GI cancer patients. However, the therapeutic response of patients selected by the predictive biomarkers is suboptimal. The tumor immune-microenvironment apparently plays a key role in modulating these molecular-determinant predictive biomarkers. Therefore, an understanding of the development and recent advances in immunotherapeutic pharmacological intervention targeting tumor immune-microenvironments and their potential predictive biomarkers will be helpful to strengthen patient immunotherapeutic efficacy. The current review focuses on an understanding of how the host-microenvironment interactions and the predictive biomarkers can determine the efficacy of immune checkpoint inhibitors. The contribution of environmental pathogens and host immunity to GI cancer is summarized. A discussion regarding the clinical evidence of predictive biomarkers for clinical trial therapy design, current immunotherapeutic strategies, and the outcomes to GI cancer patients are highlighted. An understanding of the underlying mechanism can predict the immunotherapeutic efficacy and facilitate the future development of personalized therapeutic strategies targeting GI cancers.
Collapse
Affiliation(s)
- Bilikis Aderonke Abolarinwa
- International PhD Program for Cell Therapy and Regeneration Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan.
| | - Ridwan Babatunde Ibrahim
- Institute of Brain Science, School of Medicine, National Yang-Ming University, Taipei 11221, Taiwan.
- Taiwan International Graduate Program (TIGP) in Interdisciplinary Neuroscience, National Yang-Ming University and Academia Sinica, Taipei 11529, Taiwan.
| | - Yen-Hua Huang
- International PhD Program for Cell Therapy and Regeneration Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan.
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan.
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan.
- TMU Research Center for Cell Therapy and Regeneration Medicine, Taipei Medical University, Taipei 11031, Taiwan.
- Center for Reproductive Medicine, Taipei Medical University Hospital, Taipei 11031, Taiwan.
- Comprehensive Cancer Center of Taipei Medical University, Taipei 11031, Taiwan.
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei 11031, Taiwan.
- Ph.D. Program for Translational Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan.
| |
Collapse
|
48
|
Papamichael D, Glynne-Jones R. Identifying patients who may benefit from oxaliplatin-containing perioperative chemo(radio)therapy for rectal cancer. Ann Oncol 2019; 29:1616-1618. [PMID: 29982296 DOI: 10.1093/annonc/mdy230] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/22/2022] Open
Affiliation(s)
- D Papamichael
- Department of Medical Oncology, Bank of Cyprus Oncology Centre, Nicosia, Cyprus.
| | - R Glynne-Jones
- Department of Radiotherapy, Mount Vernon Centre for Cancer Treatment, Northwood, UK
| |
Collapse
|
49
|
Zhang Y, Xu J, Zhang N, Chen M, Wang H, Zhu D. Targeting the tumour immune microenvironment for cancer therapy in human gastrointestinal malignancies. Cancer Lett 2019; 458:123-135. [DOI: 10.1016/j.canlet.2019.05.017] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/28/2019] [Revised: 05/11/2019] [Accepted: 05/14/2019] [Indexed: 12/12/2022]
|
50
|
Scientific reports concerning the impact of interleukin 4, interleukin 10 and transforming growth factor β on cancer cells. Cent Eur J Immunol 2019; 44:190-200. [PMID: 31530989 PMCID: PMC6745546 DOI: 10.5114/ceji.2018.76273] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 11/21/2017] [Accepted: 03/12/2018] [Indexed: 02/07/2023] Open
Abstract
Cytokines are signalling proteins generated in most part by immune cells that have critical functions in cellular lifespan. Here we present recent data on three selected anti-inflammatory cytokines: interleukin (IL)-10, IL-4 and transforming growth factor β (TGF-β). IL-10 inhibits the synthesis of major pro-inflammatory cytokines, chemokines, and mediates anti-inflammatory reactions. IL-4 is a multifunctional cytokine which plays a crucial role in the regulation of immune responses and is involved in processes associated with development and differentiation of lymphocytes and regulation of T cell survival. Transforming TGF-β, which in normal cells or pre-cancerous cells, promotes proliferation arrest which represses tumour growth. In this review, we focus on the influence of IL-10, IL-4 and TGF-β on various types of cancer as well as potential of these selected cytokines to serve as new biomarkers which can support effective therapies for cancer patients. This article is presented based on a review of the newest research results.
Collapse
|