1
|
Lin J, Wu Y, Liu G, Cui R, Xu Y. Advances of ultrasound in tumor immunotherapy. Int Immunopharmacol 2024; 134:112233. [PMID: 38735256 DOI: 10.1016/j.intimp.2024.112233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 04/11/2024] [Accepted: 05/07/2024] [Indexed: 05/14/2024]
Abstract
Immunotherapy has become a revolutionary method for treating tumors, offering new hope to cancer patients worldwide. Immunotherapy strategies such as checkpoint inhibitors, chimeric antigen receptor T-cell (CAR-T) therapy, and cancer vaccines have shown significant potential in clinical trials. Despite the promising results, there are still limitations that impede the overall effectiveness of immunotherapy; the response to immunotherapy is uneven, the response rate of patients is still low, and systemic immune toxicity accompanied with tumor cell immune evasion is common. Ultrasound technology has evolved rapidly in recent years and has become a significant player in tumor immunotherapy. The introductions of high intensity focused ultrasound and ultrasound-stimulated microbubbles have opened doors for new therapeutic strategies in the fight against tumor. This paper explores the revolutionary advancements of ultrasound combined with immunotherapy in this particular field.
Collapse
Affiliation(s)
- Jing Lin
- Department of Ultrasound, Guangdong Provincial Hospital of Chinese Medicine-Zhuhai Hospital, Zhuhai, PR China.
| | - Yuwei Wu
- Faculty of Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Taipa, Macao, PR China
| | - Guangde Liu
- Department of Ultrasound, Guangdong Provincial Hospital of Chinese Medicine-Zhuhai Hospital, Zhuhai, PR China
| | - Rui Cui
- Department of Ultrasonography, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510000, PR China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510000, PR China
| | - Youhua Xu
- Faculty of Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Taipa, Macao, PR China; Macau University of Science and Technology Zhuhai MUST Science and Technology Research Institute, Hengqin, Zhuhai, PR China.
| |
Collapse
|
2
|
Bell B, Flores-Lovon K, Cueva-Chicaña LA, Macedo R. Role of chemokine receptors in gastrointestinal mucosa. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2024; 388:20-52. [PMID: 39260937 DOI: 10.1016/bs.ircmb.2024.02.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/13/2024]
Abstract
Chemokine receptors are essential for the immune response in the oral and gut mucosa. The gastrointestinal mucosa is characterized by the presence of immune populations because it is susceptible to inflammatory and infectious diseases, necessitating immune surveillance. Chemokine receptors are expressed on immune cells and play a role in gastrointestinal tissue-homing, although other non-immune cells also express them for various biological functions. CCR9, CXCR3 and CXCR6 play an important role in the T cell response in inflammatory and neoplastic conditions of the gastrointestinal mucosa. However, CXCR6 could also be found in gastric cancer cells, highlighting the different roles of chemokine receptors in different pathologies. On the other hand, CCR4 and CCR8 are critical for Treg migration in gastrointestinal tissues, correlating with poor prognosis in mucosal cancers. Other chemokine receptors are also important in promoting myeloid infiltration with context-dependent roles. Further, CXCR4 and CXCR7 are also present in gastrointestinal tumor cells and are known to stimulate proliferation, migration, and invasion into other tissues, among other pro-tumorigenic functions. Determining the processes underlying mucosal immunity and creating tailored therapeutic approaches for gastrointestinal diseases requires an understanding of the complex interactions that occur between chemokine receptors and their ligands in these mucosal tissues.
Collapse
Affiliation(s)
- Brett Bell
- Albert Einstein College of Medicine, New York, NY, United States
| | - Kevin Flores-Lovon
- Universidad Nacional de San Agustín de Arequipa, Arequipa, Peru; Grupo de Investigación en Inmunología (GII), Arequipa, Peru
| | - Luis A Cueva-Chicaña
- Universidad Nacional de San Agustín de Arequipa, Arequipa, Peru; Grupo de Investigación en Inmunología (GII), Arequipa, Peru
| | - Rodney Macedo
- Albert Einstein College of Medicine, New York, NY, United States; Grupo de Investigación en Inmunología (GII), Arequipa, Peru; Columbia Center for Translational Immunology, Department of Medicine, Columbia University Medical Center, New York, United States.
| |
Collapse
|
3
|
Nikolouzakis TK, Chrysos E, Docea AO, Fragkiadaki P, Souglakos J, Tsiaoussis J, Tsatsakis A. Current and Future Trends of Colorectal Cancer Treatment: Exploring Advances in Immunotherapy. Cancers (Basel) 2024; 16:1995. [PMID: 38893120 PMCID: PMC11171065 DOI: 10.3390/cancers16111995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 05/15/2024] [Accepted: 05/20/2024] [Indexed: 06/21/2024] Open
Abstract
Cancer of the colon and rectum (CRC) has been identified among the three most prevalent types of cancer and cancer-related deaths for both sexes. Even though significant progress in surgical and chemotherapeutic techniques has markedly improved disease-free and overall survival rates in contrast to those three decades ago, recent years have seen a stagnation in these improvements. This underscores the need for new therapies aiming to augment patient outcomes. A number of emerging strategies, such as immune checkpoint inhibitors (ICIs) and adoptive cell therapy (ACT), have exhibited promising outcomes not only in preclinical but also in clinical settings. Additionally, a thorough appreciation of the underlying biology has expanded the scope of research into potential therapeutic interventions. For instance, the pivotal role of altered telomere length in early CRC carcinogenesis, leading to chromosomal instability and telomere dysfunction, presents a promising avenue for future treatments. Thus, this review explores the advancements in CRC immunotherapy and telomere-targeted therapies, examining potential synergies and how these novel treatment modalities intersect to potentially enhance each other's efficacy, paving the way for promising future therapeutic advancements.
Collapse
Affiliation(s)
| | - Emmanuel Chrysos
- Department of General Surgery, University General Hospital of Heraklion, 71110 Heraklion, Greece; (T.K.N.); (E.C.)
| | - Anca Oana Docea
- Department of Toxicology, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Persefoni Fragkiadaki
- Department of Forensic Sciences and Toxicology, Faculty of Medicine, University of Crete, 71003 Heraklion, Greece; (P.F.); (A.T.)
| | - John Souglakos
- Laboratory of Translational Oncology, Medical School, University of Crete, 70013 Heraklion, Greece;
| | - John Tsiaoussis
- Department of Anatomy, Medical School, University of Crete, 70013 Heraklion, Greece;
| | - Aristidis Tsatsakis
- Department of Forensic Sciences and Toxicology, Faculty of Medicine, University of Crete, 71003 Heraklion, Greece; (P.F.); (A.T.)
| |
Collapse
|
4
|
Ahmad A, Mahmood N, Raza MA, Mushtaq Z, Saeed F, Afzaal M, Hussain M, Amjad HW, Al-Awadi HM. Gut microbiota and their derivatives in the progression of colorectal cancer: Mechanisms of action, genome and epigenome contributions. Heliyon 2024; 10:e29495. [PMID: 38655310 PMCID: PMC11035079 DOI: 10.1016/j.heliyon.2024.e29495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 04/08/2024] [Accepted: 04/09/2024] [Indexed: 04/26/2024] Open
Abstract
Gut microbiota interacts with host epithelial cells and regulates many physiological functions such as genetics, epigenetics, metabolism of nutrients, and immune functions. Dietary factors may also be involved in the etiology of colorectal cancer (CRC), especially when an unhealthy diet is consumed with excess calorie intake and bad practices like smoking or consuming a great deal of alcohol. Bacteria including Fusobacterium nucleatum, Enterotoxigenic Bacteroides fragilis (ETBF), and Escherichia coli (E. coli) actively participate in the carcinogenesis of CRC. Gastrointestinal tract with chronic inflammation and immunocompromised patients are at high risk for CRC progression. Further, the gut microbiota is also involved in Geno-toxicity by producing toxins like colibactin and cytolethal distending toxin (CDT) which cause damage to double-stranded DNA. Specific microRNAs can act as either tumor suppressors or oncogenes depending on the cellular environment in which they are expressed. The current review mainly highlights the role of gut microbiota in CRC, the mechanisms of several factors in carcinogenesis, and the role of particular microbes in colorectal neoplasia.
Collapse
Affiliation(s)
- Awais Ahmad
- Department of Food Science, Government College University Faisalabad, Faisalabad, Pakistan
| | - Nasir Mahmood
- Department of Zoology, University of Central Punjab Bahawalpur, Bahawalpur, Pakistan
| | - Muhammad Ahtisham Raza
- Department of Food Science, Government College University Faisalabad, Faisalabad, Pakistan
| | - Zarina Mushtaq
- Department of Food Science, Government College University Faisalabad, Faisalabad, Pakistan
| | - Farhan Saeed
- Department of Food Science, Government College University Faisalabad, Faisalabad, Pakistan
| | - Muhammad Afzaal
- Department of Food Science, Government College University Faisalabad, Faisalabad, Pakistan
| | - Muzzamal Hussain
- Department of Food Science, Government College University Faisalabad, Faisalabad, Pakistan
| | - Hafiz Wasiqe Amjad
- International Medical School, Jinggangshan University, Ji'an, Jiangxi, China
| | | |
Collapse
|
5
|
Jiang J, Lin C, Chang T, Lo L, Lin C, Lu R, Yang C. Decreased interleukin-17RA expression is associated with good prognosis in patients with colorectal cancer and inhibits tumor growth and vascularity in mice. Cancer Med 2024; 13:e7059. [PMID: 38491831 PMCID: PMC10943367 DOI: 10.1002/cam4.7059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 01/18/2024] [Accepted: 02/16/2024] [Indexed: 03/18/2024] Open
Abstract
BACKGROUND Interleukin-17 (IL-17) is a pro-inflammatory cytokine that plays a vital role in the promotion of tumorigenesis in various cancers, including colorectal cancer (CRC). Based on current evidence, IL-17 binds to interleukin-17 receptor A (IL-17RA); however, the role of IL-17RA has not been elucidated in previous studies on CRC. In this study, we explored the role of IL-17RA in human CRC tissues and the progression of CRC in humans and mice. METHODS The expressions of IL-17RA and epithelial-mesenchymal transition (EMT)-related genes were examined in CRC cells and tissue samples by quantitative real-time polymerase chain reaction. The role of IL-17RA in pathogenesis and prognosis was evaluated using a Chi-squared test, Kaplan-Meier analysis, univariate, and multivariate Cox regression analysis in 133 CRC patients. A tumor-bearing mice model was executed to evaluate the role of IL-17RA in tumor growth, vascularity and population of infiltrating immune cells. RESULTS IL-17RA expression was found to be significantly higher in CRC tissues than in adjacent normal tissues. The expression of IL-17RA in Stage IV patients was significantly higher than that in Stages I and II patients. Patients with high IL-17RA expression exhibited significantly worse overall and CRC-specific survival than those with low IL-17RA expression. Functional assessment suggested that the knockdown of IL-17RA expression distinctly suppressed cellular proliferation, migration, invasion, and EMT-related gene expression. In a tumor-bearing mouse model, decreased IL-17RA expression significantly repressed tumor growth and vascularity and reduced the population of regulatory T cells (Tregs) and myeloid-derived suppressor cells (MDSCs). CONCLUSION Reduced IL-17RA expression also suppressed cellular proliferation, migration, and invasion, and the expression of EMT genes. Knockdown of IL-17RA inhibited tumor growth and vascularity and decreased the population of Tregs and MDSCs in mouse tumors. Overall, IL-17RA expression was identified to be independently associated with the prognosis of patients with CRC.
Collapse
Affiliation(s)
- Jeng‐Kai Jiang
- School of MedicineNational Yang Ming Chiao Tung UniversityTaipeiTaiwan
- Division of Colon and Rectal Surgery, Department of SurgeryTaipei Veterans General HospitalTaipeiTaiwan
| | - Chi‐Hung Lin
- Institute of Microbiology and ImmunologyNational Yang Ming Chiao Tung UniversityTaipeiTaiwan
- Department of Biological Science and TechnologyNational Yang Ming Chiao Tung UniversityHsinchuTaiwan
- Cancer Progression Research CenterNational Yang Ming Chiao Tung UniversityTaipeiTaiwan
| | - Ting‐An Chang
- Department of Pathology, Ren‐Ai BranchTaipei City HospitalTaipeiTaiwan
| | - Liang‐Chuan Lo
- National Genomics Center for Clinical and Biotechnological Applications, Cancer and Immunology Research CenterNational Yang Ming Chiao Tung UniversityTaipeiTaiwan
| | - Chien‐Ping Lin
- Division of Colon and Rectal Surgery, Department of SurgeryTaipei Veterans General HospitalTaipeiTaiwan
| | - Ruey‐Hwa Lu
- Department of Surgery, Zhongxing BranchTaipei City HospitalTaipeiTaiwan
| | - Chih‐Yung Yang
- Commission for General EducationNational United UniversityMiaoliTaiwan
- General Education CenterUniversity of TaipeiTaipeiTaiwan
- Department of Education and ResearchTaipei City HospitalTaipeiTaiwan
| |
Collapse
|
6
|
Meng Q, Zhao Y, Xu M, Wang P, Li J, Cui R, Fu W, Ding S. Increased circulating regulatory T cells and decreased follicular T helper cells are associated with colorectal carcinogenesis. Front Immunol 2024; 15:1287632. [PMID: 38343544 PMCID: PMC10853383 DOI: 10.3389/fimmu.2024.1287632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Accepted: 01/10/2024] [Indexed: 02/15/2024] Open
Abstract
Objective Colorectal cancer (CRC) is the third most prevalent cancer worldwide and is associated with high morbidity and mortality rates. Colorectal carcinogenesis occurs via the conventional adenoma-to-carcinoma and serrated pathways. Conventional T helper (Th) and innate lymphoid cells (ILCs) play vital roles in maintaining intestinal homeostasis. However, the contribution of these two major lymphoid cell populations and their associated cytokines to CRC development is unclear. Therefore, we aimed to analyze peripheral lymphocyte profiles during colorectal carcinogenesis. Methods We collected 86 blood samples concurrently, and pathologists confirmed the presence of various pathological conditions (i.e., HPs, adenoma, and carcinoma) using hematoxylin and eosin staining. Ten healthy donors were recruited as healthy controls (HCs) from the physical examination center. We performed flow cytometry on peripheral blood mononuclear cells collected from patients with various pathological conditions and the HCs, and cytokines (interleukin-2, interleukin-4, interleukin-5, interleukin-13, interleukin-17A, interleukin-17F, interleukin-22, interferon-γ, and tumor necrosis factor-α) were quantified. We also analyzed the published single-cell RNA sequence data derived from tissue samples from different stages of colorectal carcinogenesis. Results The cytokine response in peripheral CD4+ T cells was upregulated during the carcinoma process. The frequency of peripheral regulatory T cells (Tregs) increased in the adenoma and carcinoma stages. While the T follicular helper (Tfh) cell proportion was downregulated in the adenoma and carcinoma processes. Thus, Th cell subsets, especially Tregs and Tfh cells, were involved in colonic diseases. Moreover, the immunological profile characteristics in the HPs were clarified. Conclusion We comprehensively analyzed circulating ILCs and adaptive T-cell lymphocyte subtypes in colorectal carcinoma progression. Our results show the immunological profile characteristics and support the involvement of Th subsets, especially Treg and Tfh cell populations, in colonic diseases. These findings significantly enhance our understanding of the immune mechanisms underlying CRC and its precancerous lesions. Further investigation of the Treg and Tfh cells' function in colorectal disease development will provide potential therapeutic targets for monitoring and preventing CRC development.
Collapse
Affiliation(s)
- Qiao Meng
- Department of Gastroenterology, Peking University Third Hospital, Beijing, China
- Beijing Key Laboratory for Helicobacter Pylori Infection and Upper Gastrointestinal Diseases, Beijing, China
| | - Yang Zhao
- Department of Laboratory Medicine, Peking University Third Hospital, Beijing, China
| | - Miao Xu
- Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Pingzhang Wang
- Department of Immunology, School of Basic Medical Sciences, Peking University Health Science Center, NHC Key Laboratory of Medical Immunology (Peking University), Beijing, China
| | - Jun Li
- Department of Gastroenterology, Peking University Third Hospital, Beijing, China
| | - Rongli Cui
- Department of Gastroenterology, Peking University Third Hospital, Beijing, China
| | - Weiwei Fu
- Department of Gastroenterology, Peking University Third Hospital, Beijing, China
- Beijing Key Laboratory for Helicobacter Pylori Infection and Upper Gastrointestinal Diseases, Beijing, China
| | - Shigang Ding
- Department of Gastroenterology, Peking University Third Hospital, Beijing, China
- Beijing Key Laboratory for Helicobacter Pylori Infection and Upper Gastrointestinal Diseases, Beijing, China
| |
Collapse
|
7
|
Gu J, Deng S, Jiang Z, Mao F, Xue Y, Qin L, Shi J, Yang J, Li H, Yu J, Liu K, Wu K, Cao Y, Cai K. Modified Naples prognostic score for evaluating the prognosis of patients with obstructive colorectal cancer. BMC Cancer 2023; 23:941. [PMID: 37798689 PMCID: PMC10557152 DOI: 10.1186/s12885-023-11435-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 09/22/2023] [Indexed: 10/07/2023] Open
Abstract
BACKGROUND Inflammatory, immune, and nutritional status are key factors in obstructive colorectal cancer (OCRC). This study aims to investigate the value of modified Naples prognostic score (M-NPS) in evaluating OCRC prognosis. METHODS A total of 196 OCRC patients were retrospectively analyzed to construct M-NPS based on serum albumin (ALB), total cholesterol (CHOL), neutrophil:lymphocyte ratio (NLR), and lymphocyte:monocyte ratio (LMR), and then they were divided into three groups. The Kaplan-Meier (KM) method and Cox proportional hazard regression analysis were performed for overall survival (OS) and disease-free survival (DFS) of OCRC patients. RESULTS Patients with high M-NPS had worse OS and DFS (P = 0.0001, P = 0.0011). Multivariate COX analysis showed that M-NPS was an independent prognostic factor for OCRC patients. Patients in the M-NPS 2 group had significantly worse OS (hazard ratio [HR] = 4.930 (95% confidence interval [95% CI], 2.217-10.964), P < 0.001) and DFS (HR = 3.508 (95% CI, 1.691-7.277), P < 0.001) than those in the 0 group. CONCLUSION M-NPS was an independent prognostic factor for OCRC patients; it might provide a potential reference for immunonutritional intervention in patients with obstruction.
Collapse
Affiliation(s)
- Junnan Gu
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China
| | - Shenghe Deng
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China
| | - Zhenxing Jiang
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China
| | - Fuwei Mao
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China
| | - Yifan Xue
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China
| | - Le Qin
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China
| | - Jianguo Shi
- Department of Gastrointestinal Surgery, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China
| | - Jia Yang
- Department of Gastrointestinal Surgery, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China
| | - Huili Li
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China
| | - Jie Yu
- Department of Colorectal Anal Surgery, Jingzhou Central Hospital, The Second Clinical Medical College, Yangtze University, No. 60 Jingzhong Road, Jingzhou, 434020, Hubei Province, China
| | - Ke Liu
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China
| | - Ke Wu
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China
| | - Yinghao Cao
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China.
| | - Kailin Cai
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China.
| |
Collapse
|
8
|
Kumar A, Gautam V, Sandhu A, Rawat K, Sharma A, Saha L. Current and emerging therapeutic approaches for colorectal cancer: A comprehensive review. World J Gastrointest Surg 2023; 15:495-519. [PMID: 37206081 PMCID: PMC10190721 DOI: 10.4240/wjgs.v15.i4.495] [Citation(s) in RCA: 27] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 01/11/2023] [Accepted: 03/06/2023] [Indexed: 04/22/2023] Open
Abstract
Colorectal cancer (CRC) affects 1 in 23 males and 1 in 25 females, making it the third most common cancer. With roughly 608000 deaths worldwide, CRC accounts for 8% of all cancer-related deaths, making it the second most common cause of death due to cancer. Standard and conventional CRC treatments include surgical expurgation for resectable CRC and radiotherapy, chemotherapy, immunotherapy, and their combinational regimen for non-resectable CRC. Despite these tactics, nearly half of patients develop incurable recurring CRC. Cancer cells resist the effects of chemotherapeutic drugs in a variety of ways, including drug inactivation, drug influx and efflux modifications, and ATP-binding cassette transporter overexpression. These constraints necessitate the development of new target-specific therapeutic strategies. Emerging therapeutic approaches, such as targeted immune boosting therapies, non-coding RNA-based therapies, probiotics, natural products, oncolytic viral therapies, and biomarker-driven therapies, have shown promising results in preclinical and clinical studies. We tethered the entire evolutionary trends in the development of CRC treatments in this review and discussed the potential of new therapies and how they might be used in conjunction with conventional treatments as well as their advantages and drawbacks as future medicines.
Collapse
Affiliation(s)
- Anil Kumar
- Department of Pharmacology, Post Graduate Institute of Medical Education and Research, Chandigarh 160012, India
| | - Vipasha Gautam
- Department of Pharmacology, Post Graduate Institute of Medical Education and Research, Chandigarh 160012, India
| | - Arushi Sandhu
- Department of Pharmacology, Post Graduate Institute of Medical Education and Research, Chandigarh 160012, India
| | - Kajal Rawat
- Department of Pharmacology, Post Graduate Institute of Medical Education and Research, Chandigarh 160012, India
| | - Antika Sharma
- Department of Pharmacology, Post Graduate Institute of Medical Education and Research, Chandigarh 160012, India
| | - Lekha Saha
- Department of Pharmacology, Post Graduate Institute of Medical Education and Research, Chandigarh 160012, India
| |
Collapse
|
9
|
Bonnereau J, Courau T, Asesio N, Salfati D, Bouhidel F, Corte H, Hamoudi S, Hammoudi N, Lavolé J, Vivier-Chicoteau J, Chardiny V, Maggiori L, Blery M, Remark R, Bonnafous C, Cattan P, Toubert A, Bhat P, Allez M, Aparicio T, Le Bourhis L. Autologous T cell responses to primary human colorectal cancer spheroids are enhanced by ectonucleotidase inhibition. Gut 2023; 72:699-709. [PMID: 35803702 DOI: 10.1136/gutjnl-2021-326553] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 06/29/2022] [Indexed: 12/12/2022]
Abstract
OBJECTIVE T cells are major effectors of the antitumoural immune response. Their activation by tumour-associated antigens can unleash their proliferation and cytotoxic functions, leading to tumour cell elimination. However, tumour-related immunosuppressive mechanisms including the overexpression of immune checkpoints like programmed cell death protein-1 (PD-1), are also engaged, promoting immune escape. Current immunotherapies targeting these pathways have demonstrated weak efficacy in colorectal cancer (CRC). It is thus crucial to find new targets for immunotherapy in this cancer type. DESIGN In a prospective cohort of patients with CRC, we investigated the phenotype of tumour-related and non-tumour related intestinal T cells (n=44), particularly the adenosinergic pathway, correlating with clinical phenotype. An autologous coculture model was developed between patient-derived primary tumour spheroids and their autologous tumour-associated lymphocytes. We used this relevant model to assess the effects of CD39 blockade on the antitumour T cell response. RESULTS We show the increased expression of CD39, and its co-expression with PD-1, on tumour infiltrating T cells compared with mucosal lymphocytes. CD39 expression was higher in the right colon and early-stage tumours, thus defining a subset of patients potentially responsive to CD39 blockade. Finally, we demonstrate in autologous conditions that CD39 blockade triggers T cell infiltration and tumour spheroid destruction in cocultures. CONCLUSION In CRC, CD39 is strongly expressed on tumour infiltrating lymphocytes and its inhibition represents a promising therapeutic strategy for treating patients.
Collapse
Affiliation(s)
- Julie Bonnereau
- INSERM U1160, Institut de Recherche Saint-Louis, Saint Louis Hospital, Université de Paris, Paris, France
| | - Tristan Courau
- INSERM U1160, Institut de Recherche Saint-Louis, Saint Louis Hospital, Université de Paris, Paris, France
| | - Nicolas Asesio
- Department of Hepato-Gastroenterology, Hôpital Saint-Louis, Paris, France
| | - Delphine Salfati
- Department of Hepato-Gastroenterology, Hôpital Saint-Louis, Paris, France
| | - Fatiha Bouhidel
- Anatomopathology Department, Hôpital Saint-Louis, Paris, France
| | - Hélène Corte
- Digestive Surgery Department, Hôpital Saint-Louis, Paris, France
| | - Sarah Hamoudi
- INSERM U1160, Institut de Recherche Saint-Louis, Saint Louis Hospital, Université de Paris, Paris, France
| | - Nassim Hammoudi
- INSERM U1160, Institut de Recherche Saint-Louis, Saint Louis Hospital, Université de Paris, Paris, France.,Department of Hepato-Gastroenterology, Hôpital Saint-Louis, Paris, France
| | - Julie Lavolé
- INSERM U1160, Institut de Recherche Saint-Louis, Saint Louis Hospital, Université de Paris, Paris, France
| | - Justine Vivier-Chicoteau
- INSERM U1160, Institut de Recherche Saint-Louis, Saint Louis Hospital, Université de Paris, Paris, France.,Department of Hepato-Gastroenterology, Hôpital Saint-Louis, Paris, France
| | - Victor Chardiny
- INSERM U1160, Institut de Recherche Saint-Louis, Saint Louis Hospital, Université de Paris, Paris, France
| | - Leon Maggiori
- Digestive Surgery Department, Hôpital Saint-Louis, Paris, France
| | | | | | | | - Pierre Cattan
- Digestive Surgery Department, Hôpital Saint-Louis, Paris, France
| | - Antoine Toubert
- INSERM U1160, Institut de Recherche Saint-Louis, Saint Louis Hospital, Université de Paris, Paris, France
| | - Purnima Bhat
- Medical School, The Australian National University, Canberra, Australian Capital Territory, Australia.,Gastroenterology and Hepatology Unit, Canberra Hospital, Canberra, Australian Capital Territory, Australia
| | - Matthieu Allez
- INSERM U1160, Institut de Recherche Saint-Louis, Saint Louis Hospital, Université de Paris, Paris, France.,Department of Hepato-Gastroenterology, Hôpital Saint-Louis, Paris, France
| | - Thomas Aparicio
- INSERM U1160, Institut de Recherche Saint-Louis, Saint Louis Hospital, Université de Paris, Paris, France.,Department of Hepato-Gastroenterology, Hôpital Saint-Louis, Paris, France
| | - Lionel Le Bourhis
- INSERM U1160, Institut de Recherche Saint-Louis, Saint Louis Hospital, Université de Paris, Paris, France
| |
Collapse
|
10
|
Prakash A, Gates T, Zhao X, Wangmo D, Subramanian S. Tumor-derived extracellular vesicles in the colorectal cancer immune environment and immunotherapy. Pharmacol Ther 2023; 241:108332. [PMID: 36526013 DOI: 10.1016/j.pharmthera.2022.108332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 12/06/2022] [Accepted: 12/12/2022] [Indexed: 12/15/2022]
Abstract
Despite significant advances in the screening, diagnosis, and treatment of colorectal cancer (CRC) immune checkpoint inhibitors (ICIs) continue to have limited utility outside of microsatellite-high disease. Given the durable response to immunotherapy seen across malignancies, increasing CRC response rates to ICI therapy is an active area of clinical research. An increasing body of work has demonstrated that tumor-derived extracellular vesicles (TEVs) are key modulators in tumor signaling and the determinants of the tumor microenvironment. Pre-clinical models have shown that TEVs are directly involved in antigen presentation and are involved in radiation-induced DNA damage signaling. Both direct and indirect modifications of these TEVs can alter CRC immunogenicity and ICI treatment response, making them attractive targets for potential therapeutic development. In addition, modified TEVs can be developed using several different mechanisms, with varied cargo including micro-RNAs and small peptide molecules. Recent work has shown strong pre-clinical evidence of injected modified TEV-induced ICI activity, with knockdown of the micro-RNA miR-424 in TEVs improving CRC immunogenicity and increasing anti-PD-1 activity in mouse models. Clinical trials are ongoing in the evaluation of modified TEVs in cancer therapy, but they appear to be a promising therapeutic target in CRC.
Collapse
Affiliation(s)
- Ajay Prakash
- Masonic Cancer Center, University of Minnesota Medical School, Minneapolis, MN, United States of America.
| | - Travis Gates
- Department of Surgery, University of Minnesota Medical School, Minneapolis, MN, United States of America
| | - Xianda Zhao
- Department of Surgery, University of Minnesota Medical School, Minneapolis, MN, United States of America
| | - Dechen Wangmo
- Department of Surgery, University of Minnesota Medical School, Minneapolis, MN, United States of America
| | - Subbaya Subramanian
- Masonic Cancer Center, University of Minnesota Medical School, Minneapolis, MN, United States of America; Department of Surgery, University of Minnesota Medical School, Minneapolis, MN, United States of America; Center for Immunology, University of Minnesota Medical School, Minneapolis, MN, United States of America
| |
Collapse
|
11
|
Jie Y, Yang X, Chen W. Pulsatilla Decoction Combined with 5-Fluorouracil Triggers Immunogenic Cell Death in Colorectal Cancer Cells. Cancer Biother Radiopharm 2022; 37:945-954. [PMID: 34042519 DOI: 10.1089/cbr.2020.4369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Background: Our research is designed to explore the role of 5-FU and Pulsatilla decoction (PD) through modulation of Immunogenic cell death (ICD) for the co-treatment of Colorectal cancer (CRC). Materials and Methods: Cell viability was evaluated by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-2H-tetrazol-3-ium bromide (MTT) assays. Cell apoptosis was assessed using flow cytometry. Phosphorylation of STAT3 and expression of Mcl-1 and Bcl-xl were measured by Western blot assays. The levels of ATP and HMGB1 in the supernatants of the culture medium were analyzed by ATP assays and the HMGB1 enzyme linked immunosorbent assay kit. The cell surface levels of CRT were measured by immunofluorescence assays. The tumor growth was analyzed in mice. Results: PD increased 5-FU-induced ICD in CRC cells, as demonstrated by the extracellular levels of adenosine triphosphate (ATP) and high-mobility group box 1 (HMGB1), and the surface levels of calreticulin (CRT). Our mechanism study showed that PD promoted 5-FU-induced ICD by inactivating signal transducer and activator of transcription 3 (STAT3). Furthermore, the co-treatment of 5-FU and PD further promoted 5-FU-induced CRT expression and T cell infiltration in vivo. Tumorigenicity analysis revealed that 5-FU combined with PD notably reduced tumor growth. Conclusion: This study indicated that PD enhances 5-FU-induced ICD and anti-tumor effect in CRC by inactivating STAT3. The combined application of 5-FU with PD may improve the anti-tumor activity of 5-FU in CRC.
Collapse
Affiliation(s)
- Yanghua Jie
- Radiotherapy Center, Affiliated Hospital of Traditional Chinese Medicine, Xinjiang Medical University, Urumqi, China
| | - Xiaobei Yang
- Department of Anorectal, Urumqi City Hospital of Traditional Chinese Medicine, Urumqi, China
| | - Weidong Chen
- Department of Anorectal, Hospital (T.C.M) Affiliated to Southwest Medical University, Luzhou, China
| |
Collapse
|
12
|
Yu L, Zhang MM, Hou JG. Molecular and cellular pathways in colorectal cancer: apoptosis, autophagy and inflammation as key players. Scand J Gastroenterol 2022; 57:1279-1290. [PMID: 35732586 DOI: 10.1080/00365521.2022.2088247] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND Colorectal carcinogenesis (CRC) is one of the most aggressive forms of cancer, particularly in developing countries. It accounts for the second and third-highest reason for cancer-induced lethality in women and men respectively. CRC involves genetic and epigenetic modifications in colonic epithelium, leading to colon adenocarcinoma. The current review highlights the pathogenic mechanisms and multifactorial etiology of CRC, influenced by apoptosis, inflammation, and autophagy pathways. METHODS We have carried out a selective literature review on mechanisms contributing to the pathogenesis of CRC. RESULTS Resistance to senescence and apoptosis of the mesenchymal cells, which play a key role in intestinal organogenesis, morphogenesis and homeostasis, appears important for sporadic CRC. Additionally, inflammation-associated tumorigenesis is a key incident in CRC, supported by immune disruptors, adaptive and innate immune traits, environmental factors, etc. involving oxidative stress, DNA damage and epigenetic modulations. The self-digesting mechanism, autophagy, also plays a twin role in CRC through the participation of LC3/LC3-II, Beclin-1, ATG5, other autophagy proteins, and Inflammatory Bowel Disease (IBD) susceptibility genes. It facilitates the promotion of effective surveillance pathways and stimulates the generation of malignant tumor cells. The autophagy and apoptotic pathways undergo synergistic or antagonistic interactions in CRC and bear a critical association with IBD that results from the pro-neoplastic effects of persistent intestinal inflammation. Conversely, pro-inflammatory factors stimulate tumor growth and angiogenesis and inhibit apoptosis, suppressing anti-tumor activities. CONCLUSION Hence, research attempts for the development of potential therapies for CRC are in progress, primarily based on combinatorial approaches targeting apoptosis, inflammation, and autophagy.
Collapse
Affiliation(s)
- Lei Yu
- Department of Radiotherapy, The Second Hospital of Jilin University, Changchun, China
| | - Miao-Miao Zhang
- Department of Radiotherapy, The Second Hospital of Jilin University, Changchun, China
| | - Ji-Guang Hou
- Department of Radiotherapy, The Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
13
|
Liu K, Mao X, Li T, Xu Z, An R. Immunotherapy and immunobiomarker in breast cancer: current practice and future perspectives. Am J Cancer Res 2022; 12:3532-3547. [PMID: 36119833 PMCID: PMC9442024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 06/28/2022] [Indexed: 06/15/2023] Open
Abstract
Among the new cancer cases and resulting deaths among women worldwide, breast cancer is the most significant threat to women's health. In recent years, immunotherapy was initially used to treat patients with metastatic breast cancer, where it demonstrated its unique value by providing a novel way to improve therapeutic effects and prolong survival time. With the development of clinical trials related to immunotherapy for breast cancer, tumour vaccines, such as DNA vaccines, have been observed to improve the disease-free survival (DFS) and overall survival (OS) of patients. Monoclonal antibodies have also shown good efficacy, and adoptive cell therapies, such as CAR-T, exhibit strong tumour killing ability and good safety, and thus, these therapies may comprise a new strategy for the treatment of breast cancer. These breakthrough successes have promoted the achievement of "individualized" breast cancer treatment. Moreover, a recent study showed that patients with various cancer types with a higher tumour mutational burden (TMB) are more likely to benefit from immunotherapy. As research progresses, TMB may also demonstrate a certain clinical significance in the treatment of breast cancer. This paper reviews the latest research progress on breast cancer immunotherapy and the predictive value and application status of TMB in immunotherapy regimens for breast cancer patients to provide a reference for further in-depth studies of breast cancer immunotherapy.
Collapse
Affiliation(s)
- Kangsheng Liu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xi’an Jiaotong UniversityXi’an 710061, Shannxi, P. R. China
- Department of Clinical Laboratory, Women’s Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care HospitalNanjing 210029, Jiangsu, P. R. China
| | - Xiaodong Mao
- Department of Endocrinology, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese MedicineNanjing 210028, Jiangsu, P. R. China
| | - Taiping Li
- Department of Neuro-Psychiatric Institute, The Affiliated Brain Hospital of Nanjing Medical UniversityNanjing 210029, Jiangsu, P. R. China
| | - Zhirong Xu
- Department of Clinical Laboratory, The First Affiliated Hospital of Soochow UniversitySuzhou 215006, Jiangsu, P. R. China
| | - Ruifang An
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xi’an Jiaotong UniversityXi’an 710061, Shannxi, P. R. China
| |
Collapse
|
14
|
Rodrigues MC, Morais JAV, Ganassin R, Oliveira GRT, Costa FC, Morais AAC, Silveira AP, Silva VCM, Longo JPF, Muehlmann LA. An Overview on Immunogenic Cell Death in Cancer Biology and Therapy. Pharmaceutics 2022; 14:pharmaceutics14081564. [PMID: 36015189 PMCID: PMC9413301 DOI: 10.3390/pharmaceutics14081564] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 07/22/2022] [Accepted: 07/25/2022] [Indexed: 11/28/2022] Open
Abstract
Immunogenic cell death (ICD) is a modality of regulated cell death that is sufficient to promote an adaptive immune response against antigens of the dying cell in an immunocompetent host. An important characteristic of ICD is the release and exposure of damage-associated molecular patterns, which are potent endogenous immune adjuvants. As the induction of ICD can be achieved with conventional cytotoxic agents, it represents a potential approach for the immunotherapy of cancer. Here, different aspects of ICD in cancer biology and treatment are reviewed.
Collapse
Affiliation(s)
- Mosar Corrêa Rodrigues
- Faculty of Ceilandia, University of Brasilia, Brasilia 72220-275, Brazil; (M.C.R.); (J.A.V.M.); (R.G.); (G.R.T.O.); (F.C.C.)
- Laboratory of Nanobiotechnology, Department of Genetics and Morphology, Institute of Biological Sciences, University of Brasilia, Brasilia 70910-900, Brazil; (A.A.C.M.); (A.P.S.); (V.C.M.S.); (J.P.F.L.)
| | - José Athayde Vasconcelos Morais
- Faculty of Ceilandia, University of Brasilia, Brasilia 72220-275, Brazil; (M.C.R.); (J.A.V.M.); (R.G.); (G.R.T.O.); (F.C.C.)
- Laboratory of Nanobiotechnology, Department of Genetics and Morphology, Institute of Biological Sciences, University of Brasilia, Brasilia 70910-900, Brazil; (A.A.C.M.); (A.P.S.); (V.C.M.S.); (J.P.F.L.)
| | - Rayane Ganassin
- Faculty of Ceilandia, University of Brasilia, Brasilia 72220-275, Brazil; (M.C.R.); (J.A.V.M.); (R.G.); (G.R.T.O.); (F.C.C.)
- Laboratory of Nanobiotechnology, Department of Genetics and Morphology, Institute of Biological Sciences, University of Brasilia, Brasilia 70910-900, Brazil; (A.A.C.M.); (A.P.S.); (V.C.M.S.); (J.P.F.L.)
| | - Giulia Rosa Tavares Oliveira
- Faculty of Ceilandia, University of Brasilia, Brasilia 72220-275, Brazil; (M.C.R.); (J.A.V.M.); (R.G.); (G.R.T.O.); (F.C.C.)
- Laboratory of Nanobiotechnology, Department of Genetics and Morphology, Institute of Biological Sciences, University of Brasilia, Brasilia 70910-900, Brazil; (A.A.C.M.); (A.P.S.); (V.C.M.S.); (J.P.F.L.)
| | - Fabiana Chagas Costa
- Faculty of Ceilandia, University of Brasilia, Brasilia 72220-275, Brazil; (M.C.R.); (J.A.V.M.); (R.G.); (G.R.T.O.); (F.C.C.)
- Laboratory of Nanobiotechnology, Department of Genetics and Morphology, Institute of Biological Sciences, University of Brasilia, Brasilia 70910-900, Brazil; (A.A.C.M.); (A.P.S.); (V.C.M.S.); (J.P.F.L.)
| | - Amanda Alencar Cabral Morais
- Laboratory of Nanobiotechnology, Department of Genetics and Morphology, Institute of Biological Sciences, University of Brasilia, Brasilia 70910-900, Brazil; (A.A.C.M.); (A.P.S.); (V.C.M.S.); (J.P.F.L.)
| | - Ariane Pandolfo Silveira
- Laboratory of Nanobiotechnology, Department of Genetics and Morphology, Institute of Biological Sciences, University of Brasilia, Brasilia 70910-900, Brazil; (A.A.C.M.); (A.P.S.); (V.C.M.S.); (J.P.F.L.)
| | - Victor Carlos Mello Silva
- Laboratory of Nanobiotechnology, Department of Genetics and Morphology, Institute of Biological Sciences, University of Brasilia, Brasilia 70910-900, Brazil; (A.A.C.M.); (A.P.S.); (V.C.M.S.); (J.P.F.L.)
| | - João Paulo Figueiró Longo
- Laboratory of Nanobiotechnology, Department of Genetics and Morphology, Institute of Biological Sciences, University of Brasilia, Brasilia 70910-900, Brazil; (A.A.C.M.); (A.P.S.); (V.C.M.S.); (J.P.F.L.)
| | - Luis Alexandre Muehlmann
- Faculty of Ceilandia, University of Brasilia, Brasilia 72220-275, Brazil; (M.C.R.); (J.A.V.M.); (R.G.); (G.R.T.O.); (F.C.C.)
- Laboratory of Nanobiotechnology, Department of Genetics and Morphology, Institute of Biological Sciences, University of Brasilia, Brasilia 70910-900, Brazil; (A.A.C.M.); (A.P.S.); (V.C.M.S.); (J.P.F.L.)
- Correspondence:
| |
Collapse
|
15
|
The HER family as therapeutic targets in colorectal cancer. Crit Rev Oncol Hematol 2022; 174:103681. [PMID: 35462030 DOI: 10.1016/j.critrevonc.2022.103681] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 03/30/2022] [Accepted: 04/07/2022] [Indexed: 12/23/2022] Open
Abstract
The human epidermal growth factor receptor (HER, ErbB) family has four members, epidermal growth factor receptor (EGFR), HER2, HER3, and HER4. Although distinct in ligands and functions, all of the HER family members are receptor tyrosine kinases playing important roles in the pathogenesis of cancers. In the era of precision medicine, the HER family is one of the most important and successful cancer therapeutic targets, hallmarked by the approval of anti-EGFR therapies for the treatment of colorectal cancer and non-small cell lung cancer, and anti-HER2 therapies for the treatment of breast cancer and gastric cancer. This review briefly discusses how HER family members were discovered, their functions and roles in cancer, and most importantly, the developmental history and recent updates of therapies targeting HER family members, with colorectal cancer as a focus. We also discussed the patient selection and drug resistance to anti-EGFR therapies in the treatment of colorectal cancer.
Collapse
|
16
|
Berle M, Hestetun KE, Vethe H, Chera S, Paulo JA, Dahl O, Myklebust MP. Mapping Proteome Changes in Microsatellite Stable, Recurrent Colon Cancer Reveals a Significant Immune System Signature. Cancer Genomics Proteomics 2022; 19:130-144. [PMID: 35181583 DOI: 10.21873/cgp.20309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 01/10/2022] [Accepted: 01/12/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND/AIM Better stratification of the risk of relapse will help select the right patients for adjuvant treatment and improve targeted therapies for patients with colon cancer. MATERIALS AND METHODS To understand why a subset of tumors relapse, we compared the proteome of two groups of patients with colon cancer with similar stage, stratified based on the presence or absence of recurrence. RESULTS Using tumor biopsies from the primary operation, we identified dissimilarity between recurrent and nonrecurrent mismatch satellite stable colon cancer and found that signaling related to immune activation and inflammation was associated with relapse. CONCLUSION Immune modulation may have an effect on mismatch satellite stable colon cancer. At present, immune therapy is offered primarily to microsatellite instable colon cancer. Hopefully, immune therapy in mismatch satellite stable colon cancer beyond PD-1 and PD-L1 inhibitors can be implemented.
Collapse
Affiliation(s)
- Magnus Berle
- Department of Clinical Medicine, University of Bergen, Bergen, Norway; .,Department of Surgery, Haukeland University Hospital, Bergen, Norway.,Department of Surgery, Haraldsplass Deaconess Hospital, Bergen, Norway
| | - Kjersti E Hestetun
- Department of Clinical Science, University of Bergen, Bergen, Norway.,Department of Oncology and Medical Physics, Haukeland University Hospital, Bergen, Norway
| | - Heidrun Vethe
- Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | - Simona Chera
- Department of Clinical Science, University of Bergen, Bergen, Norway.,Department of Medicine, Division of Endocrinology, Diabetes, Nutrition and Patient Education, University Hospital of Geneva, Geneva, Switzerland
| | - Joao A Paulo
- Department of Cell Biology, Harvard Medical School, Boston, MA, U.S.A
| | - Olav Dahl
- Department of Clinical Science, University of Bergen, Bergen, Norway.,Department of Oncology and Medical Physics, Haukeland University Hospital, Bergen, Norway
| | | |
Collapse
|
17
|
Luo C, Liu F, Su W, Long P, Liang J, Hou W, Jiang H, Long X, Su G. Prognostic value of LINC02560 in colorectal cancer correlates with tumor microenvironment immunity. J Cancer 2022; 12:7507-7517. [PMID: 35003370 PMCID: PMC8734400 DOI: 10.7150/jca.64940] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 10/23/2021] [Indexed: 12/24/2022] Open
Abstract
Background: LINC02560 is a new 477 bp long non-coding RNA located in 19q13.43. However, the expression of LINC02560 in colorectal cancer (CRC) has not been reported, and its correlation with tumor development and function is still unclear. Methods: The expression of LINC02560 in CRC was first analyzed in the cancer genome atlas (TCGA) combined with The Genotype-Tissue Expression(GTEx) databases and then validated by clinical CRC samples and cell lines. The association between LINC02560 expression and clinicopathologic variables was analyzed by the Wilcoxon Rank SUM test. Cox regression analysis and Kaplan-Meier plots were used to assess the prognostic value of LINC02560 in CRC. The correlation between the expression level of LINC02560 and the 24 immune cells in tumor microenvironment (TME) was analyzed by single sample gene set enrichment analysis (ssGSEA). Gene set enrichment analysis (GSEA) was conducted to detect potential biological processes associated with LINC02560 in CRC. Results: LINC02560 was significantly up-regulated in CRC in comparison to normal samples. There are significant differences in the expression of LINC02560 in different subgroups of N stage, M stage, carcinoembryonic antigen (CEA) level, residual tumor, TP53 status and pathological stage. The high LINC02560 expression indicated poor overall survival (OS) and progress free interval (PFI) in patients with CRC. Moreover, the multivariate Cox analysis demonstrated that the expression of LINC02560 was an independent prognosis-predicting factor for OS in CRC patients. GSEA indicated that high expression of LINC02560 was involved in MAPK, Wnt, and PPAR signaling pathways and participated in humoral immune processes. We also identified that LINC02560 expression had a negative correlation with 4 kinds of immune cells. Conclusions: In summary, our research results indicate that LINC02560 may be a potential prognostic biomarker. It is involved in the occurrence and development of CRC and may affect the prognosis of CRC patients by regulating immune cells in the TME.
Collapse
Affiliation(s)
- Chunying Luo
- Department of Cell Biology, Medical College of Guangxi University, Nanning 530004, Guangxi, PR China.,Department of Pathology, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise 533000, Guangxi, PR China
| | - Fahui Liu
- Department of Pathology, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise 533000, Guangxi, PR China
| | - Weichao Su
- Department of Gastrointestinal Surgery III, Xiamen Cancer Hospital, First Affiliated Hospital of Xiamen University, 55 Zhenhai Road, Xiamen 361003, China
| | - Puze Long
- Department of Pathology, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise 533000, Guangxi, PR China
| | - Jiadong Liang
- Department of Pathology, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise 533000, Guangxi, PR China
| | - Wanyun Hou
- Department of Pathology, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise 533000, Guangxi, PR China
| | - Haifeng Jiang
- Department of Gastrointestinal Surgery III, Xiamen Cancer Hospital, First Affiliated Hospital of Xiamen University, 55 Zhenhai Road, Xiamen 361003, China
| | - Xidai Long
- Department of Pathology, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise 533000, Guangxi, PR China
| | - Guoqiang Su
- Department of Cell Biology, Medical College of Guangxi University, Nanning 530004, Guangxi, PR China.,Department of Gastrointestinal Surgery III, Xiamen Cancer Hospital, First Affiliated Hospital of Xiamen University, 55 Zhenhai Road, Xiamen 361003, China
| |
Collapse
|
18
|
Zhang Y, Zhang L, Sun H, Liu Y, Xu J, Huang H, Fu J, Zhang D, Tian T, Zhao Y, Wang G. Inhibitory immune checkpoints PDCD-1 and LAG-3 hypermethylation may reduce the risk of colorectal cancer. Mol Med 2021; 27:114. [PMID: 34544358 PMCID: PMC8454079 DOI: 10.1186/s10020-021-00373-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 09/05/2021] [Indexed: 12/24/2022] Open
Abstract
Background Changes in DNA methylation of immunosuppressive checkpoints may impact express and consequently affect antigen processing and presentation by tumor cells and facilitates evasion of immunosurveillance and lead to colorectal cancer (CRC). This study is to investigate the effect of PDCD-1, LAG-3 methylation statuses in peripheral blood leukocytes on CRC risk. Methods GSE51032 dataset from Gene Expression Omnibus comprised of 166 CRC patients and 424 normal samples was used to identify significantly differentially methylated CpG sites of the two genes. A case–control study with 390 CRC patients and 397 cancer-free controls was carried out to validate the relationship between the methylation levels of the two genes and CRC susceptibility and then estimated their interactions with environmental factors on CRC risk. Results In the GSE51032 dataset, cg06291111 (PDCD-1) and cg10191002 (LAG-3) were screened as the candidate CpG sites for the following study. There were significant associations between hypermethylation of PDCD-1 and LAG-3 and lower risk of CRC (ORadj = 0.322, 95% CI 0.197–0.528; ORadj = 0.666, 95% CI 0.446–0.5996, respectively). Moreover, the results in case–control study showed similar trend, that hypermethylation of PDCD-1 and LAG-3 were associated with lower CRC risk (ORadj = 0.448, 95% CI 0.322–0.622; ORadj = 0.417, 95% CI 0.301–0.578, respectively). A synergistic interaction between LAG-3 hypermethylation and intake of eggs on CRC risk was observed. There were combination effects between hypermethylation of PDCD-1 and LAG-3 and environmental factors on CRC risk. Conclusions PDCD-1 and LAG-3 may potentially serve as blood-based predictive biomarkers for CRC risk. Supplementary Information The online version contains supplementary material available at 10.1186/s10020-021-00373-5.
Collapse
Affiliation(s)
- Yuanyuan Zhang
- Department of Epidemiology, Public Health College of Harbin Medical University, 157 Baojian Street, Nangang District, Harbin, 150081, Heilongjiang, People's Republic of China
| | - Lei Zhang
- Department of Epidemiology, Public Health College of Harbin Medical University, 157 Baojian Street, Nangang District, Harbin, 150081, Heilongjiang, People's Republic of China
| | - Hongru Sun
- Department of Epidemiology, Public Health College of Harbin Medical University, 157 Baojian Street, Nangang District, Harbin, 150081, Heilongjiang, People's Republic of China
| | - Ying Liu
- Department of Epidemiology, Public Health College of Harbin Medical University, 157 Baojian Street, Nangang District, Harbin, 150081, Heilongjiang, People's Republic of China
| | - Jing Xu
- Department of Epidemiology, Public Health College of Harbin Medical University, 157 Baojian Street, Nangang District, Harbin, 150081, Heilongjiang, People's Republic of China
| | - Hao Huang
- Department of Epidemiology, Public Health College of Harbin Medical University, 157 Baojian Street, Nangang District, Harbin, 150081, Heilongjiang, People's Republic of China
| | - Jinming Fu
- Department of Epidemiology, Public Health College of Harbin Medical University, 157 Baojian Street, Nangang District, Harbin, 150081, Heilongjiang, People's Republic of China
| | - Ding Zhang
- Department of Epidemiology, Public Health College of Harbin Medical University, 157 Baojian Street, Nangang District, Harbin, 150081, Heilongjiang, People's Republic of China
| | - Tian Tian
- Department of Epidemiology, Public Health College of Harbin Medical University, 157 Baojian Street, Nangang District, Harbin, 150081, Heilongjiang, People's Republic of China.
| | - Yashuang Zhao
- Department of Epidemiology, Public Health College of Harbin Medical University, 157 Baojian Street, Nangang District, Harbin, 150081, Heilongjiang, People's Republic of China.
| | - Guiyu Wang
- Department of Colorectal Cancer Surgery, The Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Street, Nangang District, Harbin, 150001, Heilongjiang, People's Republic of China.
| |
Collapse
|
19
|
VAN Nguyen S, Shamoun L, Landerholm K, Andersson RE, Wagsater D, Dimberg J. Cytotoxic T-lymphocyte Antigen-4 ( CTLA-4) Gene Polymorphism (rs3087243) Is Related to Risk and Survival in Patients With Colorectal Cancer. In Vivo 2021; 35:969-975. [PMID: 33622891 DOI: 10.21873/invivo.12339] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 01/12/2021] [Accepted: 01/13/2021] [Indexed: 12/15/2022]
Abstract
BACKGROUND/AIM Cytotoxic T-lymphocyte antigen-4 (CTLA-4), transiently expressed on T cells, plays a pivotal role in the negative feedback regulation of T-cell activation and proliferation. The aim of the present study was to examine the influence of CTLA-4 gene polymorphism rs3087243 on CRC susceptibility and long-term survival in Swedish patients with CRC. PATIENTS AND METHODS Genotypes of 491 patients and 433 healthy controls were determined, using TaqMan single nucleotide polymorphism (SNP) assays based on polymerase chain reaction. RESULTS Patients carrying allele A were found to be at a higher risk of CRC and this allele was found to be more common in patients with disseminated disease compared to localized disease in the right colon. Kaplan-Meier analysis of cancer-specific survival showed that carriers of allele A had the highest risk of CRC-related death. CONCLUSION The SNP rs3087243 of the CTLA-4 gene was associated with CRC risk and, therefore, it could be a prognostic marker for Swedish patients with CRC.
Collapse
Affiliation(s)
- Song VAN Nguyen
- Department of Medical Laboratory, Danang University of Medical Technology and Pharmacy, Danang, Vietnam
| | - Levar Shamoun
- Department of Laboratory Medicine and Pathology, Region Jönköping County, Jönköping, Sweden.,Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Kalle Landerholm
- Department of Surgery, Region Jönköping County, Jönköping, Sweden; Department of Biomedical and Clinical Science, Linköping University, Linköping, Sweden
| | - Roland E Andersson
- Department of Surgery, Region Jönköping County, Jönköping, Sweden; Department of Biomedical and Clinical Science, Linköping University, Linköping, Sweden
| | - Dick Wagsater
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Jan Dimberg
- Department of Natural Science and Biomedicine, School of Health and Welfare, Jönköping University, Jönköping, Sweden
| |
Collapse
|
20
|
Pecci F, Cantini L, Bittoni A, Lenci E, Lupi A, Crocetti S, Giglio E, Giampieri R, Berardi R. Beyond Microsatellite Instability: Evolving Strategies Integrating Immunotherapy for Microsatellite Stable Colorectal Cancer. Curr Treat Options Oncol 2021; 22:69. [PMID: 34110510 PMCID: PMC8192371 DOI: 10.1007/s11864-021-00870-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/30/2021] [Indexed: 12/19/2022]
Abstract
OPINION STATEMENT Advanced colorectal cancer (CRC) is a heterogeneous disease, characterized by several subtypes with distinctive genetic and epigenetic patterns. During the last years, immune checkpoint inhibitors (ICIs) have revamped the standard of care of several tumors such as non-small cell lung cancer and melanoma, highlighting the role of immune cells in tumor microenvironment (TME) and their impact on cancer progression and treatment efficacy. An "immunoscore," based on the percentage of two lymphocyte populations both at tumor core and invasive margin, has been shown to improve prediction of treatment outcome when added to UICC-TNM classification. To date, pembrolizumab, an anti-programmed death protein 1 (PD1) inhibitor, has gained approval as first-line therapy for mismatch-repair-deficient (dMMR) and microsatellite instability-high (MSI-H) advanced CRC. On the other hand, no reports of efficacy have been presented in mismatch-repair-proficient (pMMR) and microsatellite instability-low (MSI-L) or microsatellite stable (MSS) CRC. This group includes roughly 95% of all advanced CRC, and standard chemotherapy, in addition to anti-EGFR or anti-angiogenesis drugs, still represents first treatment choice. Hopefully, deeper understanding of CRC immune landscape and of the impact of specific genetic and epigenetic alterations on tumor immunogenicity might lead to the development of new drug combination strategies to overcome ICIs resistance in pMMR CRC, thus paving the way for immunotherapy even in this subgroup.
Collapse
Affiliation(s)
- Federica Pecci
- Clinical Oncology, Università Politecnica delle Marche, AOU Ospedali Riuniti, Via Conca 71, 60126 Ancona, Italy
| | - Luca Cantini
- Clinical Oncology, Università Politecnica delle Marche, AOU Ospedali Riuniti, Via Conca 71, 60126 Ancona, Italy
| | - Alessandro Bittoni
- Clinical Oncology, Università Politecnica delle Marche, AOU Ospedali Riuniti, Via Conca 71, 60126 Ancona, Italy
| | - Edoardo Lenci
- Clinical Oncology, Università Politecnica delle Marche, AOU Ospedali Riuniti, Via Conca 71, 60126 Ancona, Italy
| | - Alessio Lupi
- Clinical Oncology, Università Politecnica delle Marche, AOU Ospedali Riuniti, Via Conca 71, 60126 Ancona, Italy
| | - Sonia Crocetti
- Clinical Oncology, Università Politecnica delle Marche, AOU Ospedali Riuniti, Via Conca 71, 60126 Ancona, Italy
| | - Enrica Giglio
- Clinical Oncology, Università Politecnica delle Marche, AOU Ospedali Riuniti, Via Conca 71, 60126 Ancona, Italy
| | - Riccardo Giampieri
- Clinical Oncology, Università Politecnica delle Marche, AOU Ospedali Riuniti, Via Conca 71, 60126 Ancona, Italy
| | - Rossana Berardi
- Clinical Oncology, Università Politecnica delle Marche, AOU Ospedali Riuniti, Via Conca 71, 60126 Ancona, Italy
| |
Collapse
|
21
|
Sun Z, Xia W, Lyu Y, Song Y, Wang M, Zhang R, Sui G, Li Z, Song L, Wu C, Liew CC, Yu L, Cheng G, Cheng C. Immune-related gene expression signatures in colorectal cancer. Oncol Lett 2021; 22:543. [PMID: 34079596 PMCID: PMC8157333 DOI: 10.3892/ol.2021.12804] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Accepted: 03/11/2021] [Indexed: 12/24/2022] Open
Abstract
The immune system is crucial in regulating colorectal cancer (CRC) tumorigenesis. Identification of immune-related transcriptomic signatures derived from the peripheral blood of patients with CRC would provide insights into CRC pathogenesis, and suggest novel clues to potential immunotherapy strategies for the disease. The present study collected blood samples from 59 patients with CRC and 62 healthy control patients and performed whole blood gene expression profiling using microarray hybridization. Immune-related gene expression signatures for CRC were identified from immune gene datasets, and an algorithmic predictive model was constructed for distinguishing CRC from controls. Model performance was characterized using an area under the receiver operating characteristic curve (ROC AUC). Functional categories for CRC-specific gene expression signatures were determined using gene set enrichment analyses. A Kaplan-Meier plotter survival analysis was also performed for CRC-specific immune genes in order to characterize the association between gene expression and CRC prognosis. The present study identified five CRC-specific immune genes [protein phosphatase 3 regulatory subunit Bα (PPP3R1), amyloid β precursor protein, cathepsin H, proteasome activator subunit 4 and DEAD-Box Helicase 3 X-Linked]. A predictive model based on this five-gene panel showed good discriminatory power (independent test set sensitivity, 83.3%; specificity, 94.7%, accuracy, 89.2%; ROC AUC, 0.96). The candidate genes were involved in pathways associated with ‘adaptive immune responses’, ‘innate immune responses’ and ‘cytokine signaling’. The survival analysis found that a high level of PPP3R1 expression was associated with a poor CRC prognosis. The present study identified five CRC-specific immune genes that were potential diagnostic biomarkers for CRC. The biological function analysis indicated a close association between CRC pathogenesis and the immune system, and may reveal more information about the immunogenic and pathogenic mechanisms driving CRC in the future. Overall, the association between PPP3R1 expression and survival of patients with CRC revealed potential new targets for CRC immunotherapy.
Collapse
Affiliation(s)
- Zhenqing Sun
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266000, P.R. China
| | - Wei Xia
- Department of Nuclear Medicine, The Seventh People's Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200137, P.R. China
| | - Yali Lyu
- R&D Department, Huaxia Bangfu Technology Incorporated, Beijing 100000, P.R. China
| | - Yanan Song
- Department of Nuclear Medicine, The Seventh People's Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200137, P.R. China
| | - Min Wang
- R&D Department, Huaxia Bangfu Technology Incorporated, Beijing 100000, P.R. China
| | - Ruirui Zhang
- R&D Department, Huaxia Bangfu Technology Incorporated, Beijing 100000, P.R. China
| | - Guode Sui
- Department of General Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266000, P.R. China
| | - Zhenlu Li
- Department of General Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266000, P.R. China
| | - Li Song
- Department of General Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266000, P.R. China
| | - Changliang Wu
- Department of General Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266000, P.R. China
| | - Choong-Chin Liew
- Golden Health Diagnostics Inc., Yan Cheng, Jiangsu 224000, P.R. China.,Department of Clinical Pathology and Laboratory Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada.,Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Lei Yu
- R&D Department, Huaxia Bangfu Technology Incorporated, Beijing 100000, P.R. China
| | - Guang Cheng
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266000, P.R. China
| | - Changming Cheng
- R&D Department, Huaxia Bangfu Technology Incorporated, Beijing 100000, P.R. China
| |
Collapse
|
22
|
Molecularly targeted therapy for advanced gastrointestinal noncolorectal cancer treatment: how to choose? Past, present, future. Anticancer Drugs 2021; 32:593-601. [PMID: 33929995 DOI: 10.1097/cad.0000000000001071] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Gastrointestinal cancer is a leading cause of death worldwide. Conventional cytotoxic chemotherapy has been the backbone of advanced gastrointestinal cancer treatment for decades and still represents a key element of the therapeutic armamentarium. However, only small increments in survival outcomes have been reached. New clinical trials are designed, including classic chemotherapy in association with either small-molecule inhibitors or mAb. During the past few years, remarkable progress in molecular biology of gastrointestinal noncolorectal cancers, the discovery of specific targets and the resulting development of systemic drugs that block critical kinases and several molecular pathways have all contributed to progress. New biological agents with molecularly targeted therapies are now available or currently included in clinical trials (EGFR inhibitors (i), antiangiogenic agents, c-METi, IDHi, FGFR2i, BRAFi, Pi3Ki/AKTi/mTORi, NTRKi). When we focus on the current state of precision medicine for gastrointestinal malignancies, it becomes apparent that there is a mixed history of success and failure. The aim of this review is to focus on the studies that have been completed to date with target therapies and to understand which of these are currently the accepted choice in clinical practice and which need further confirmation and approval for inclusion in guidelines. All these findings will enable to guide clinical practice for oncologists in the design of the next round of clinical trials.
Collapse
|
23
|
Taïeb J, André T, El Hajbi F, Barbier E, Toullec C, Kim S, Bouche O, Di Fiore F, Chauvenet M, Perrier H, Evesque L, Laurent-Puig P, Emile JF, Bez J, Lepage C, Tougeron D. Avelumab versus standard second line treatment chemotherapy in metastatic colorectal cancer patients with microsatellite instability: The SAMCO-PRODIGE 54 randomised phase II trial. Dig Liver Dis 2021; 53:318-323. [PMID: 33359404 DOI: 10.1016/j.dld.2020.11.031] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 11/26/2020] [Indexed: 12/11/2022]
Abstract
Immune checkpoint inhibitors have failed in treating metastatic colorectal cancer (mCRC) patients except those with dMMR/MSI tumors. However, until very recently we had only non-comparative promising data in this population with anti-programmed cell death 1/ programmed cell death ligand 1 (PD1/PD-L1) antibodies alone or combined with anti- cytotoxic T-lymphocyte-associated protein 4 (CTLA4) antibodies. This comparative phase II trial (NCT03186326), conducted in more than 100 centers in France, will include dMMR/MSI mCRC patients with progression after a first-line treatment with chemotherapy ± targeted therapies, to evaluate efficacy and safety of the anti-PDL1 Avelumab versus a standard second-line treatment. Main inclusion criteria were patients aged 18 to 75 years, ECOG performance status ≤2, dMMR/MSI mCRC and failure of a standard first-line regimen. Patient will be randomised to receive Avelumab 10 mg/kg versus standard second-line doublet chemotherapy plus a targeted agent according to tumor RAS status. Patients will be followed for 4 years. A gain of 5 months in median PFS is expected in favour of the Avelumab arm (12 vs 7 months; HR=0.58). Secondary endpoints include objective response rate, overall survival, quality of life and toxicity. In addition, circulating tumour DNA and microbiota will be explored to test their potential prognostic and predictive values. The study was opened in March 2018.
Collapse
Affiliation(s)
- Julien Taïeb
- Assistance Publique - Hôpitaux de Paris, European Georges Pompidou hospital, Department of oncology, Paris France; Centre de Recherche des Cordeliers, INSERM, CNRS, Sorbonne Université, USPC, Université de Paris, Equipe labellisée Ligue Nationale contre le cancer, Paris, France.
| | - Thierry André
- Sorbonne Université and Hôpital Saint Antoine, Paris, France
| | - Farid El Hajbi
- Service de cancérologie Digestive et Urologique, centre Oscar Lambret, Lille, France
| | - Emilie Barbier
- Fédération Francophone de Cancérologie Digestive, EPICAD INSERM LNC-UMR 1231, University of Burgundy and Franche Comté, Dijon, France
| | | | - Stefano Kim
- Service d'oncologie CHU Jean Minjoz, Besancon, France
| | - Olivier Bouche
- Unité de Médecine Ambulatoire-Cancérologie, CHU Robert Debre, Reims, France
| | | | - Marion Chauvenet
- Service d'hépato-gastro-entérologie, CHU Lyon Sud, Hospices civils de Lyon, France
| | - Hervé Perrier
- Service d'Oncologie, Hôpital Saint Joseph, Marseille, France
| | - Ludovic Evesque
- Service d'Oncologie, Centre Antoine Lacassagne, Nice, France
| | - Pierre Laurent-Puig
- Centre de Recherche des Cordeliers, INSERM, CNRS, Sorbonne Université, USPC, Université de Paris, Equipe labellisée Ligue Nationale contre le cancer, Paris, France; Assistance Publique - Hôpitaux de Paris, Department of Biology, European George Pompidou hospital
| | - Jean-François Emile
- EA4340, Pathology department and INSERM, Ambroise Pare Hospital, Boulogne, France
| | - Jérémie Bez
- Fédération Francophone de Cancérologie Digestive, EPICAD INSERM LNC-UMR 1231, University of Burgundy and Franche Comté, Dijon, France
| | - Côme Lepage
- Fédération Francophone de Cancérologie Digestive, EPICAD INSERM LNC-UMR 1231, University of Burgundy and Franche Comté, Dijon, France; Department of digestive oncology University hospital Dijon, University of Burgundy and Franche Comté, Dijon, France
| | - David Tougeron
- Gastroenterology Department and Medical Oncology Department, Poitiers University Hospital, Poitiers, France
| |
Collapse
|
24
|
Lisby AN, Flickinger JC, Bashir B, Weindorfer M, Shelukar S, Crutcher M, Snook AE, Waldman SA. GUCY2C as a biomarker to target precision therapies for patients with colorectal cancer. EXPERT REVIEW OF PRECISION MEDICINE AND DRUG DEVELOPMENT 2021; 6:117-129. [PMID: 34027103 DOI: 10.1080/23808993.2021.1876518] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Introduction Colorectal cancer (CRC) is one of the most-deadly malignancies worldwide. Current therapeutic regimens for CRC patients are relatively generic, based primarily on disease type and stage, with little variation. As the field of molecular oncology advances, so too must therapeutic management of CRC. Understanding molecular heterogeneity has led to a new-found promotion for precision therapy in CRC; underlining the diversity of molecularly targeted therapies based on individual tumor characteristics. Areas covered We review current approaches for the treatment of CRC and discuss the potential of precision therapy in advanced CRC. We highlight the utility of the intestinal protein guanylyl cyclase C (GUCY2C), as a multi-purpose biomarker and unique therapeutic target in CRC. Here, we summarize current GUCY2C-targeted approaches for treatment of CRC. Expert opinion The GUCY2C biomarker has multi-faceted utility in medicine. Developmental investment of GUCY2C as a diagnostic and therapeutic biomarker offers a variety of options taking the molecular characteristics of cancer into account. From GUCY2C-targeted therapies, namely cancer vaccines, CAR-T cells, and monoclonal antibodies, to GUCY2C agonists for chemoprevention in those who are at high risk for developing colorectal cancer, the utility of this protein provides many avenues for exploration with significance in the field of precision medicine.
Collapse
Affiliation(s)
- Amanda N Lisby
- Department of Pharmacology and Experimental Therapeutics, Thomas Jefferson University, Philadelphia, PA 19107, United States
| | - John C Flickinger
- Department of Pharmacology and Experimental Therapeutics, Thomas Jefferson University, Philadelphia, PA 19107, United States
| | - Babar Bashir
- Department of Medical Oncology, Thomas Jefferson University, Philadelphia, PA 19107, United States
| | - Megan Weindorfer
- Department of Pharmacology and Experimental Therapeutics, Thomas Jefferson University, Philadelphia, PA 19107, United States
| | - Sanjna Shelukar
- Department of Pharmacology and Experimental Therapeutics, Thomas Jefferson University, Philadelphia, PA 19107, United States
| | - Madison Crutcher
- Department of Surgery, Thomas Jefferson University, Philadelphia, PA 19107, United States
| | - Adam E Snook
- Department of Pharmacology and Experimental Therapeutics, Thomas Jefferson University, Philadelphia, PA 19107, United States
| | - Scott A Waldman
- Department of Pharmacology and Experimental Therapeutics, Thomas Jefferson University, Philadelphia, PA 19107, United States
| |
Collapse
|
25
|
Wang J, Li S, Li H, Zhou X, Wen H, Lai B. IRF4 overexpression promotes the transdifferentiation of tregs into macrophage-like cells to inhibit the development of colon cancer. Cancer Cell Int 2021; 21:58. [PMID: 33468159 PMCID: PMC7816309 DOI: 10.1186/s12935-021-01766-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 01/06/2021] [Indexed: 01/02/2023] Open
Abstract
Background Interferon regulatory factor 4 (IRF4) is a transcription factor from the IRF factor family that exerts regulatory functions in the immune system and oncogenesis. However, the biological role of IRF4 in colon cancer is still unclear. The aim of this study is to investigate whether IRF4 participates in the immune response in colon cancer. Methods We compared the expression of IRF4, the number of regulatory T cells (Tregs) and macrophages in the colon cancer tissues and paracancerous colon tissues from colon cancer patients. Colon cancer mouse model was established by inoculation with colon cancer cells (SW480) as a xenograft tumor, and we observed tumor growth of colon cancer. Furthermore, the mechanism of action of IRF4 in transdifferentiation of Tregs into macrophage-like cells and the effect of IRF4 on colon cancer cells were investigated in vitro. Results IRF4 was severely down-regulated in the colon cancer tissues. Colon cancer tissues exhibited an increase in the number of regulatory T cells (Tregs) and macrophages. Furthermore, IRF4 overexpression repressed proliferation, migration and invasion of colon cancer cells (SW480 and HT116 cells). Moreover, IRF4 up-regulation ameliorated tumor growth of colon cancer by promoting the transdifferentiation of Tregs into macrophage-like cells through inhibition of BCL6 expression. Exosomes derived from colon cancer cells repressed IRF4 expression in Tregs by transmitting miR-27a-3p, miR-30a-5p and miR-320c. Conclusions IRF4 overexpression promoted the transdifferentiation of Tregs into macrophage-like cells to inhibit the occurrence and development of colon cancer. Thus, IRF4 may be a potential target for colon cancer treatment.
Collapse
Affiliation(s)
- Jiwei Wang
- Department of Ultrasound, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Song Li
- Mudanjiang Medical College, Mudanjiang, Heilongjiang, China
| | - Honglang Li
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, No 1. Minde Road, Nanchang, Jiangxi, 330006, China
| | - Xiaoshuang Zhou
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, No 1. Minde Road, Nanchang, Jiangxi, 330006, China
| | - Huabin Wen
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, No 1. Minde Road, Nanchang, Jiangxi, 330006, China
| | - Bin Lai
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, No 1. Minde Road, Nanchang, Jiangxi, 330006, China.
| |
Collapse
|
26
|
Bakhrebah MA, Nasrullah M, Abdulaal WH, Hassan MA, Siddiqui H, Al Doghaither H, Omar UM, Helmi N, Fallatah MM, Al-Ghafari AB, Khan MI, Choudhry H. High Expression of Pd-1 in Circulating Cells of Patients With Advanced Colorectal Cancer Receiving Adjuvant Therapy. Technol Cancer Res Treat 2020; 19:1533033820969446. [PMID: 33153413 PMCID: PMC7658510 DOI: 10.1177/1533033820969446] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Among all cancer types, colorectal cancer is the third most common in men and the second most common in women globally. Generally, the risk of colorectal cancer increases with age, and colorectal cancer is modulated by various genetic alterations. Alterations in the immune response serve a significant role in the development of colorectal cancer. In primary cancer types, immune cells express a variety of inhibitory molecules that dampen the immune response against tumor cells. Additionally, few reports have demonstrated that classical chemotherapy promotes the immunosuppressive microenvironment in both tissues and immune cells. This study assessed the expression levels of genes using RT-qPCR associated with the immune system, including interferon-γ, programmed death-1, β2-microglobulin, human leukocyte antigen-A, CD3e, CD28 and intracellular adhesion molecule 1, in patients with colorectal cancer, as these genes are known to serve important roles in immune regulation during cancer incidence. Gene expression analysis was performed with the whole blood cells of patients with colorectal cancer and healthy volunteers. Compared with the normal controls, programmed death-1was highly expressed in patients with advanced-stage colorectal cancer. Furthermore, the expression of programmed death-1 was higher in patients receiving adjuvant therapy, which suggests the therapy dampened the immune response against tumor cells. The results of the present study indicate that classical adjuvant therapies, which are currently used for patients with colorectal cancer, should be modulated, and a combination of classical therapy with anti-programmed death-1 antibody should be conducted for improved management of patients with colorectal cancer.
Collapse
Affiliation(s)
- Muhammed A Bakhrebah
- Life Science and Environment Research Institute, 83527King Abdulaziz City for Science and Technology, Riyadh, Saudi Arabia
| | - Mohammad Nasrullah
- Cancer Metabolism and Epigenetic Unit, Department of Biochemistry, Faculty of Science, 2495King Abdulaziz University, Jeddah, Saudi Arabia.,Cancer and Mutagenesis Unit, King Fahd Medical Research Center, 37848King Abdulaziz University, Jeddah, Saudi Arabia.,Department of Oncology, Faculty of Medicine and Dentistry, 3158University of Alberta, Edmonton AB, Canada
| | - Wesam H Abdulaal
- Cancer Metabolism and Epigenetic Unit, Department of Biochemistry, Faculty of Science, 2495King Abdulaziz University, Jeddah, Saudi Arabia.,Cancer and Mutagenesis Unit, King Fahd Medical Research Center, 37848King Abdulaziz University, Jeddah, Saudi Arabia
| | - Mohammed A Hassan
- Cancer Metabolism and Epigenetic Unit, Department of Biochemistry, Faculty of Science, 2495King Abdulaziz University, Jeddah, Saudi Arabia.,Cancer and Mutagenesis Unit, King Fahd Medical Research Center, 37848King Abdulaziz University, Jeddah, Saudi Arabia.,Department of Basic Medical Sciences, College of Medicine and Health Sciences, Hadhramout University, Mukalla, Yemen
| | - Halima Siddiqui
- Cancer Metabolism and Epigenetic Unit, Department of Biochemistry, Faculty of Science, 2495King Abdulaziz University, Jeddah, Saudi Arabia.,Cancer and Mutagenesis Unit, King Fahd Medical Research Center, 37848King Abdulaziz University, Jeddah, Saudi Arabia
| | - Huda Al Doghaither
- Cancer Metabolism and Epigenetic Unit, Department of Biochemistry, Faculty of Science, 2495King Abdulaziz University, Jeddah, Saudi Arabia
| | - Ulfat M Omar
- Cancer Metabolism and Epigenetic Unit, Department of Biochemistry, Faculty of Science, 2495King Abdulaziz University, Jeddah, Saudi Arabia
| | - Nawal Helmi
- Cancer and Mutagenesis Unit, King Fahd Medical Research Center, 37848King Abdulaziz University, Jeddah, Saudi Arabia.,Department of Applied Biochemistry, Faculty of Sciences, University of Jeddah, Jeddah, Saudi Arabia
| | - Mohannad M Fallatah
- Life Science and Environment Research Institute, 83527King Abdulaziz City for Science and Technology, Riyadh, Saudi Arabia
| | - Ayat B Al-Ghafari
- Cancer Metabolism and Epigenetic Unit, Department of Biochemistry, Faculty of Science, 2495King Abdulaziz University, Jeddah, Saudi Arabia.,Cancer and Mutagenesis Unit, King Fahd Medical Research Center, 37848King Abdulaziz University, Jeddah, Saudi Arabia
| | - Mohammad Imran Khan
- Cancer Metabolism and Epigenetic Unit, Department of Biochemistry, Faculty of Science, 2495King Abdulaziz University, Jeddah, Saudi Arabia.,Cancer and Mutagenesis Unit, King Fahd Medical Research Center, 37848King Abdulaziz University, Jeddah, Saudi Arabia
| | - Hani Choudhry
- Cancer Metabolism and Epigenetic Unit, Department of Biochemistry, Faculty of Science, 2495King Abdulaziz University, Jeddah, Saudi Arabia.,Cancer and Mutagenesis Unit, King Fahd Medical Research Center, 37848King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
27
|
Haak F, Obrecht I, Tosti N, Weixler B, Mechera R, Däster S, von Strauss M, Delko T, Spagnoli GC, Terracciano L, Sconocchia G, von Flüe M, Kraljević M, Droeser RA. Tumor Infiltration by OX40+ Cells Enhances the Prognostic Significance of CD16+ Cell Infiltration in Colorectal Cancer. Cancer Control 2020; 27:1073274820903383. [PMID: 32107932 PMCID: PMC7053789 DOI: 10.1177/1073274820903383] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Objectives: Analysis of tumor immune infiltration has been suggested to outperform tumor,
node, metastasis staging in predicting clinical course of colorectal cancer
(CRC). Infiltration by cells expressing OX40, a member of the tumor necrosis
factor receptor family, or CD16, expressed by natural killer cells,
monocytes, and dendritic cells, has been associated with favorable prognosis
in patients with CRC. We hypothesized that assessment of CRC infiltration by
both OX40+ and CD16+ cells might result in enhanced prognostic
significance. Methods: Colorectal cancer infiltration by OX40 and CD16 expressing cells was
investigated in 441 primary CRCs using tissue microarrays and specific
antibodies, by immunohistochemistry. Patients’ survival was evaluated by
Kaplan-Meier and log-rank tests. Multivariate Cox regression analysis,
hazard ratios, and 95% confidence intervals were also used to evaluate
prognostic significance of OX40+ and CD16+ cell infiltration. Results: Colorectal cancer infiltration by OX40+ and CD16+ cells was subclassified
into 4 groups with high or low infiltration levels in all possible
combinations. High levels of infiltration by both OX40+ and CD16+ cells were
associated with lower pT stage, absence of peritumoral lymphocytic (PTL)
inflammation, and a positive prognostic impact. Patients bearing tumors with
high infiltration by CD16+ and OX40+ cells were also characterized by
significantly longer overall survival, as compared with the other groups.
These results were confirmed by analyzing an independent validation
cohort. Conclusions: Combined infiltration by OX40+ and CD16+ immune cells is an independent
favorable prognostic marker in CRC. The prognostic value of CD16+ immune
cell infiltration is significantly improved by the combined analysis with
OX40+ cell infiltration.
Collapse
Affiliation(s)
- Fabian Haak
- Department of Abdominal Surgery, Clarunis, University Center for Gastrointestinal and Liver Diseases, Basel, Switzerland
| | - Isabelle Obrecht
- Department of Abdominal Surgery, Clarunis, University Center for Gastrointestinal and Liver Diseases, Basel, Switzerland
| | - Nadia Tosti
- Institute of Pathology, University Hospital Basel, Basel, Switzerland
| | - Benjamin Weixler
- Department of Abdominal Surgery, Clarunis, University Center for Gastrointestinal and Liver Diseases, Basel, Switzerland.,Department of General, Visceral and Vascular Surgery, Charite Campus Benjamin Franklin, Berlin, Germany
| | - Robert Mechera
- Department of Abdominal Surgery, Clarunis, University Center for Gastrointestinal and Liver Diseases, Basel, Switzerland
| | - Silvio Däster
- Department of Abdominal Surgery, Clarunis, University Center for Gastrointestinal and Liver Diseases, Basel, Switzerland
| | - Marco von Strauss
- Department of Abdominal Surgery, Clarunis, University Center for Gastrointestinal and Liver Diseases, Basel, Switzerland
| | - Tarik Delko
- Department of Abdominal Surgery, Clarunis, University Center for Gastrointestinal and Liver Diseases, Basel, Switzerland
| | - Giulio C Spagnoli
- Department of Biomedicine, University Hospital Basel, Basel, Switzerland.,Institute of Translational Pharmacology, National Research Council, Rome, Italy
| | - Luigi Terracciano
- Institute of Pathology, University Hospital Basel, Basel, Switzerland
| | - Giuseppe Sconocchia
- Institute of Translational Pharmacology, National Research Council, Rome, Italy
| | - Markus von Flüe
- Department of Abdominal Surgery, Clarunis, University Center for Gastrointestinal and Liver Diseases, Basel, Switzerland
| | - Marko Kraljević
- Department of Abdominal Surgery, Clarunis, University Center for Gastrointestinal and Liver Diseases, Basel, Switzerland
| | - Raoul A Droeser
- Department of Abdominal Surgery, Clarunis, University Center for Gastrointestinal and Liver Diseases, Basel, Switzerland.,Department of Biomedicine, University Hospital Basel, Basel, Switzerland
| |
Collapse
|
28
|
Immune scores in colorectal cancer: Where are we? Eur J Cancer 2020; 140:105-118. [PMID: 33075623 DOI: 10.1016/j.ejca.2020.08.024] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 08/28/2020] [Indexed: 12/24/2022]
Abstract
There is growing evidence that the immune system may prevent the occurrence, growth and metastatic diffusion of colorectal cancer (CRC). The role played by the adaptive immune response at the tumour site is critical in the balance between tumour invasion and defence against cancer. Recent data have shown that the evaluation of this immune response may help to define the prognosis and possibly the treatment of localised CRC as well as metastatic CRC. Tumour infiltrates with T cells (CD3+), cytotoxic T cells (CD8+) and memory T cells (CD45RO+) are the immune parameters most consistently and strongly associated with good clinical outcome in CRC. Several scoring systems have been developed, including the Immunoscore®, based on the immunohistochemical determination with a digital image analysis system of the density of CD3+ and CD8+ lymphocytes in the centre and the invasive margin of the tumour. This review will focus on the different immunoscoring systems developed in CRC, their performance, their limitations and their potential for improving patients' care in the future.
Collapse
|
29
|
Nugteren S, Goos JACM, Delis-van Diemen PM, Simons-Oosterhuis Y, Lindenbergh-Kortleve DJ, van Haaften DH, Sanders J, Meijer GA, Fijneman RJA, Samsom JN. Expression of the immune modulator secretory leukocyte protease inhibitor (SLPI) in colorectal cancer liver metastases and matched primary tumors is associated with a poorer prognosis. Oncoimmunology 2020; 9:1832761. [PMID: 33101778 PMCID: PMC7556627 DOI: 10.1080/2162402x.2020.1832761] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Secretory leukocyte protease inhibitor (SLPI), a pleiotropic protein expressed by healthy intestinal epithelial cells, functions as an inhibitor of NF-κB and neutrophil proteases and exerts antimicrobial activity. We previously showed SLPI suppresses intestinal epithelial chemokine production in response to microbial contact. Increased SLPI expression was recently detected in various types of carcinoma. In addition, accumulating evidence indicates SLPI expression is favorable for tumor cells. In view of these findings and the abundance of SLPI in the colonic epithelium, we hypothesized SLPI promotes colorectal cancer (CRC) growth and metastasis. Here, we aimed to establish whether SLPI expression in CRC is related to clinical outcome. Using a cohort of 507 patients with CRC who underwent resection of liver metastases, we show that high SLPI protein expression in both liver metastases and primary CRC is associated with significantly shorter overall survival after resection of liver metastases. The prognostic value of SLPI in CRC patients with liver metastases implies a role for SLPI in the formation of metastasis of human CRC. Based on the immune regulatory functions of SLPI, we anticipate that expression of SLPI provides tumors with a mechanism to evade infiltration by immune cells.
Collapse
Affiliation(s)
- Sandrine Nugteren
- Laboratory of Pediatrics, Division Gastroenterology and Nutrition, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Jeroen A C M Goos
- Departments of Clinical Neuroscience, Radiopharmacy, and Oncology & Pathology, Karolinska Institute, Stockholm, Sweden
| | | | - Ytje Simons-Oosterhuis
- Laboratory of Pediatrics, Division Gastroenterology and Nutrition, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Dicky J Lindenbergh-Kortleve
- Laboratory of Pediatrics, Division Gastroenterology and Nutrition, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Daniëlle H van Haaften
- Laboratory of Pediatrics, Division Gastroenterology and Nutrition, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Joyce Sanders
- Department of Pathology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Gerrit A Meijer
- Department of Pathology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Remond J A Fijneman
- Department of Pathology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Janneke N Samsom
- Laboratory of Pediatrics, Division Gastroenterology and Nutrition, Erasmus University Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
30
|
Rappaport JA, Waldman SA. An update on guanylyl cyclase C in the diagnosis, chemoprevention, and treatment of colorectal cancer. Expert Rev Clin Pharmacol 2020; 13:1125-1137. [PMID: 32945718 DOI: 10.1080/17512433.2020.1826304] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Introduction: Colorectal cancer remains the second leading cause of cancer death in the United States, underscoring the need for novel therapies. Despite the successes of new targeted agents for other cancers, colorectal cancer suffers from a relative scarcity of actionable biomarkers. In this context, the intestinal receptor, guanylyl cyclase C (GUCY2C), has emerged as a promising target.Areas covered: GUCY2C regulates a tumor-suppressive signaling axis that is silenced through loss of its endogenous ligands at the earliest stages of tumorigenesis. A body of literature supports a cancer chemoprevention strategy involving reactivation of GUCY2C through FDA-approved cGMP-elevating agents such as linaclotide, plecanatide, and sildenafil. Its limited expression in extra-intestinal tissues, and retention on the surface of cancer cells, also positions GUCY2C as a target for immunotherapies to treat metastatic disease, including vaccines, chimeric antigen receptor T-cells, and antibody-drug conjugates. Likewise, GUCY2C mRNA identifies metastatic cells, enhancing colorectal cancer detection, and staging. Pre-clinical and clinical programs exploring these GUCY2C-targeting strategies will be reviewed.Expert opinion: Recent mechanistic insights characterizing GUCY2C ligand loss early in tumorigenesis, coupled with results from the first clinical trials testing GUCY2C-targeting strategies, continue to elevate GUCY2C as an ideal target for prevention, detection, and therapy.
Collapse
Affiliation(s)
- Jeffrey A Rappaport
- Department of Pharmacology and Experimental Therapeutics, Thomas Jefferson University , Philadelphia, PA, USA
| | - Scott A Waldman
- Department of Pharmacology and Experimental Therapeutics, Thomas Jefferson University , Philadelphia, PA, USA
| |
Collapse
|
31
|
Johdi NA, Sukor NF. Colorectal Cancer Immunotherapy: Options and Strategies. Front Immunol 2020; 11:1624. [PMID: 33042104 PMCID: PMC7530194 DOI: 10.3389/fimmu.2020.01624] [Citation(s) in RCA: 225] [Impact Index Per Article: 56.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Accepted: 06/17/2020] [Indexed: 12/24/2022] Open
Abstract
Colorectal cancer is the third most common cancer in the world with increasing incidence and mortality rates globally. Standard treatments for colorectal cancer have always been surgery, chemotherapy and radiotherapy which may be used in combination to treat patients. However, these treatments have many side effects due to their non-specificity and cytotoxicity toward any cells including normal cells that are growing and dividing. Furthermore, many patients succumb to relapse even after a series of treatments. Thus, it is crucial to have more alternative and effective treatments to treat CRC patients. Immunotherapy is one of the new alternatives in cancer treatment. The strategy is to utilize patients' own immune systems in combating the cancer cells. Cancer immunotherapy overcomes the issue of specificity which is the major problem in chemotherapy and radiotherapy. The normal cells with no cancer antigens are not affected. The outcomes of some cancer immunotherapy have been astonishing in some cases, but some which rely on the status of patients' own immune systems are not. Those patients who responded well to cancer immunotherapy have a better prognostic and better quality of life.
Collapse
Affiliation(s)
- Nor Adzimah Johdi
- UKM Medical Molecular Biology Institute (UMBI), National University of Malaysia, Bangi, Malaysia
| | | |
Collapse
|
32
|
Chai BY, Yip WK, Dusa N, Mohtarrudin N, Seow HF. Loss of Interleukin-17RA Expression is Associated with Tumour Progression in Colorectal Carcinoma. Pathol Oncol Res 2020; 26:2291-2298. [PMID: 32462420 DOI: 10.1007/s12253-020-00820-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Revised: 04/14/2020] [Accepted: 05/12/2020] [Indexed: 11/28/2022]
Abstract
Interleukin-17 (IL-17) is a pro-inflammatory cytokine found in various cancers. Current evidence indicates that IL-17 plays a vital role in tumour initiation and progression in colorectal carcinoma (CRC) via binding with its receptor, IL-17RA. However, the association between clinicopathological features and presence of IL-17 and IL-17RA protein in primary CRC tissues remains unclear. This study also investigates the difference between the presence of IL-17 and IL-17RA in the paired tumour tissues versus adjacent normal tissues. The presence of IL-17RA and IL-17 protein in primary CRC tissues was determined by immunohistochemistry. Associations between clinicopathological features and IL-17RA and IL-17 immunoreactivity, were analyzed by χ2 tests. We found that both IL-17RA (p = 0.001) and IL-17 (p = 0.025) in tumour cells of primary CRC tissues was significantly lower as compared to adjacent normal tissue. Positive immunoreactivity for IL-17RA and IL-17 were detected in 51.0% and 16.8% of tumour tissues, respectively. Furthermore, negative immunoreactivity of IL-17R was significantly associated with advanced stage according to TNM classifier (p = 0.027), high grade of tumour (p = 0.019), increased depth of tumour invasion (p = 0.023) and vascular invasion (p = 0.039). Positive IL-17 immunoreactivity was associated with advanced stage (p = 0.008) and lymph node metastasis (p = 0.008). Thus, this study suggests that the loss of IL-17RA expression occurs as tumour progresses and this may predict the aggressiveness of tumour whilst expression of IL-17 promotes tumour progression and lymph node metastasis. Thus, loss of IL-17RA could be a useful prognostic biomarker for tumour progression in CRC patients.
Collapse
Affiliation(s)
- Boon Yean Chai
- Immunology Laboratory, Department of Pathology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Selangor, 43400, Malaysia
| | - Wai Kien Yip
- Immunology Laboratory, Department of Pathology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Selangor, 43400, Malaysia
| | - Noraini Dusa
- Department of Pathology, Hospital Kuala Lumpur, Kuala Lumpur, Malaysia
| | - Norhafizah Mohtarrudin
- Department of Pathology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Selangor, 43400, Malaysia
| | - Heng Fong Seow
- Immunology Laboratory, Department of Pathology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Selangor, 43400, Malaysia. .,Immunology Unit, Department of Pathology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Selangor, 43400, Malaysia.
| |
Collapse
|
33
|
Yu AI, Zhao L, Eaton KA, Ho S, Chen J, Poe S, Becker J, Gonzalez A, McKinstry D, Hasso M, Mendoza-Castrejon J, Whitfield J, Koumpouras C, Schloss PD, Martens EC, Chen GY. Gut Microbiota Modulate CD8 T Cell Responses to Influence Colitis-Associated Tumorigenesis. Cell Rep 2020; 31:107471. [PMID: 32268087 PMCID: PMC7934571 DOI: 10.1016/j.celrep.2020.03.035] [Citation(s) in RCA: 106] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 02/13/2020] [Accepted: 03/12/2020] [Indexed: 02/07/2023] Open
Abstract
There is increasing evidence that gut microbiome perturbations, also known as dysbiosis, can influence colorectal cancer development. To understand the mechanisms by which the gut microbiome modulates cancer susceptibility, we examine two wild-type mouse colonies with distinct gut microbial communities that develop significantly different tumor numbers using a mouse model of inflammation-associated tumorigenesis. We demonstrate that adaptive immune cells contribute to the different tumor susceptibilities associated with the two microbial communities. Mice that develop more tumors have increased colon lamina propria CD8+ IFNγ+ T cells before tumorigenesis but reduced CD8+ IFNγ+ T cells in tumors and adjacent tissues compared with mice that develop fewer tumors. Notably, intratumoral T cells in mice that develop more tumors exhibit increased exhaustion. Thus, these studies suggest that microbial dysbiosis can contribute to colon tumor susceptibility by hyperstimulating CD8 T cells to promote chronic inflammation and early T cell exhaustion, which can reduce anti-tumor immunity.
Collapse
Affiliation(s)
- Amy I Yu
- Graduate Program in Immunology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Lili Zhao
- Department of Biostatistics, University of Michigan, University of Michigan, Ann Arbor, MI 48109, USA
| | - Kathryn A Eaton
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Sharon Ho
- College of Literature, Science, and the Arts, University of Michigan, Ann Arbor, MI 48109, USA
| | - Jiachen Chen
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Sara Poe
- Unit for Laboratory Animal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - James Becker
- Unit for Laboratory Animal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Allison Gonzalez
- College of Literature, Science, and the Arts, University of Michigan, Ann Arbor, MI 48109, USA
| | - Delaney McKinstry
- College of Literature, Science, and the Arts, University of Michigan, Ann Arbor, MI 48109, USA
| | - Muneer Hasso
- College of Literature, Science, and the Arts, University of Michigan, Ann Arbor, MI 48109, USA
| | | | - Joel Whitfield
- Cancer Center Immunology Core, University of Michigan, Ann Arbor, MI 48109, USA
| | - Charles Koumpouras
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Patrick D Schloss
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Eric C Martens
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Grace Y Chen
- Graduate Program in Immunology, University of Michigan, Ann Arbor, MI 48109, USA; Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
34
|
Payandeh Z, Khalili S, Somi MH, Mard-Soltani M, Baghbanzadeh A, Hajiasgharzadeh K, Samadi N, Baradaran B. PD-1/PD-L1-dependent immune response in colorectal cancer. J Cell Physiol 2020; 235:5461-5475. [PMID: 31960962 DOI: 10.1002/jcp.29494] [Citation(s) in RCA: 83] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Accepted: 01/09/2020] [Indexed: 12/19/2022]
Abstract
Colorectal cancer (CRC) is still considered as the third most frequent cancer in the world. Microsatellite instability (MSI), inflammation, and microRNAs have been demonstrated as the main contributing factors in CRC. Subtype 1 CRC is defined by NK cells infiltration, induction of Th1 lymphocyte and cytotoxic T cell responses as well as upregulation of immune checkpoint proteins including programmed cell death-1 (PD-1). Based on the diverse features of CRC, such as the stage and localization of the tumor, several treatment approaches are available. However, the efficiency of these treatments may be decreased due to the development of diverse resistance mechanisms. It has been proven that monoclonal antibodies (mAbs) can increase the effectiveness of CRC treatments. Nowadays, several mAbs including nivolumab and pembrolizumab have been approved for the treatment of CRC. Immune checkpoint receptors including PD-1 can be inhibited by these antibodies. Combination therapy gives an opportunity for advanced treatment for CRC patients. In this review, an update has been provided on the molecular mechanisms involved in MSI colorectal cancer immune microenvironment by focusing on PD-ligand 1 (PD-L1) and treatment of patients with advanced immunotherapy, which were examined in the different clinical trial phases. Considering induced expression of PD-L1 by conventional chemotherapeutics, we have summarized the role of PD-L1 in CRC, the chemotherapy effects on the PD-1/PD-L1 axis and novel combined approaches to enhance immunotherapy of CRC by focusing on PD-L1.
Collapse
Affiliation(s)
- Zahra Payandeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Saeed Khalili
- Department of Biology Sciences, Shahid Rajaee Teacher Training University, Tehran, Iran
| | - Mohammad Hossein Somi
- Liver and Gastrointestinal Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Maysam Mard-Soltani
- Department of Clinical Biochemistry, Faculty of Medical Sciences, Dezful University of Medical Sciences, Dezful, Iran
| | - Amir Baghbanzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Nasser Samadi
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.,Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Biochemistry and Clinical, Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
35
|
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: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar 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
|
36
|
Loktionov A. Eosinophils in the gastrointestinal tract and their role in the pathogenesis of major colorectal disorders. World J Gastroenterol 2019; 25:3503-3526. [PMID: 31367153 PMCID: PMC6658389 DOI: 10.3748/wjg.v25.i27.3503] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 05/22/2019] [Accepted: 06/01/2019] [Indexed: 02/06/2023] Open
Abstract
Eosinophils are currently regarded as versatile mobile cells controlling and regulating multiple biological pathways and responses in health and disease. These cells store in their specific granules numerous biologically active substances (cytotoxic cationic proteins, cytokines, growth factors, chemokines, enzymes) ready for rapid release. The human gut is the main destination of eosinophils that are produced and matured in the bone marrow and then transferred to target tissues through the circulation. In health the most important functions of gut-residing eosinophils comprise their participation in the maintenance of the protective mucosal barrier and interactions with other immune cells in providing immunity to microbiota of the gut lumen. Eosinophils are closely involved in the development of inflammatory bowel disease (IBD), when their cytotoxic granule proteins cause damage to host tissues. However, their roles in Crohn’s disease and ulcerative colitis appear to follow different immune response patterns. Eosinophils in IBD are especially important in altering the structure and protective functions of the mucosal barrier and modulating massive neutrophil influx to the lamina propria followed by transepithelial migration to colorectal mucus. IBD-associated inflammatory process involving eosinophils then appears to expand to the mucus overlaying the internal gut surface. The author hypothesises that immune responses within colorectal mucus as well as ETosis exerted by both neutrophils and eosinophils on the both sides of the colonic epithelial barrier act as additional pathogenetic factors in IBD. Literature analysis also shows an association between elevated eosinophil levels and better colorectal cancer (CRC) prognosis, but mechanisms behind this effect remain to be elucidated. In conclusion, the author emphasises the importance of investigating colorectal mucus in IBD and CRC patients as a previously unexplored milieu of disease-related inflammatory responses.
Collapse
|
37
|
Xia T, Sun H, Huang H, Bi H, Pu R, Zhang L, Zhang Y, Liu Y, Xu J, Onwuka JU, Liu Y, Cui B, Zhao Y. Androgen receptor gene methylation related to colorectal cancer risk. Endocr Connect 2019; 8:979-987. [PMID: 31176300 PMCID: PMC6612065 DOI: 10.1530/ec-19-0122] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Accepted: 06/06/2019] [Indexed: 12/14/2022]
Abstract
According to its incidence patterns, colorectal cancer (CRC) tends to occur more frequently in males than in females, and the evidence shows that CRC is a hormone-related tumor. These findings indicate that androgen receptor (AR) gene methylation might be important for the regulation of the CRC risk in the different sexes. We used a case-control study to investigate the association between AR methylation in peripheral blood (PBL) and CRC risk. A cohort study was conducted to analyze the effect of AR methylation levels in both PBL and tissue on the prognosis of CRC. AR methylation levels were detected using methylation-sensitive high-resolution melting (MS-HRM). The results indicate that the hypomethylation of AR was significantly associated with the risk of CRC (OR = 1.869, 95% CI: 1.629-2.141, P < 0.001), and the results remained similar after adjusting for the propensity score (PS) (OR = 1.344, 95% CI: 1.147-1.575, P < 0.001) and PS matching (OR = 1.138, 95% CI: 1.000-1.292 P = 0.049). The hypomethylation of AR was significantly associated with CRC in males (OR = 2.309, 95% CI: 1.200-4.245; P = 0.012) but not females (OR = 1.000, 95% CI: 0.567-1.765; P = 0.999). The methylation status of AR in PBL and tissue does not seem to be associated with prognosis in colorectal cancer (OR = 1.425, 95% CI: 0.895-2.269, P = 0.135; OR = 0.930, 95% CI: 0.674-1.285, P = 0.661). We conclude that AR hypomethylation in PBL is associated with a high risk of CRC and may serve as a biomarker. Further studies involving large sample sizes are needed to validate the results of this study.
Collapse
Affiliation(s)
- Tingting Xia
- Department of Epidemiology, Public Health College, Harbin Medical University, Harbin, Heilongjiang Province, People’s Republic of China
| | - Hongru Sun
- Department of Epidemiology, Public Health College, Harbin Medical University, Harbin, Heilongjiang Province, People’s Republic of China
| | - Hao Huang
- Department of Epidemiology, Public Health College, Harbin Medical University, Harbin, Heilongjiang Province, People’s Republic of China
| | - Haoran Bi
- Department of Epidemiology, Public Health College, Harbin Medical University, Harbin, Heilongjiang Province, People’s Republic of China
| | - Rui Pu
- Department of Epidemiology, Public Health College, Harbin Medical University, Harbin, Heilongjiang Province, People’s Republic of China
| | - Lei Zhang
- Department of Epidemiology, Public Health College, Harbin Medical University, Harbin, Heilongjiang Province, People’s Republic of China
| | - Yuanyuan Zhang
- Department of Epidemiology, Public Health College, Harbin Medical University, Harbin, Heilongjiang Province, People’s Republic of China
| | - Ying Liu
- Department of Epidemiology, Public Health College, Harbin Medical University, Harbin, Heilongjiang Province, People’s Republic of China
| | - Jing Xu
- Department of Epidemiology, Public Health College, Harbin Medical University, Harbin, Heilongjiang Province, People’s Republic of China
| | - Justina Ucheojor Onwuka
- Department of Epidemiology, Public Health College, Harbin Medical University, Harbin, Heilongjiang Province, People’s Republic of China
| | - Yupeng Liu
- Department of Epidemiology, Public Health College, Harbin Medical University, Harbin, Heilongjiang Province, People’s Republic of China
- Correspondence should be addressed to Y Liu or B Cui or Y Zhao: or or
| | - Binbin Cui
- Department of Colorectal Surgery, The Third Hospital of Harbin Medical University, Harbin, Heilongjiang Province, People’s Republic of China
- Correspondence should be addressed to Y Liu or B Cui or Y Zhao: or or
| | - Yashuang Zhao
- Department of Epidemiology, Public Health College, Harbin Medical University, Harbin, Heilongjiang Province, People’s Republic of China
- Correspondence should be addressed to Y Liu or B Cui or Y Zhao: or or
| |
Collapse
|
38
|
Spinner CA, Lamsoul I, Métais A, Febrissy C, Moog-Lutz C, Lutz PG. The E3 Ubiquitin Ligase Asb2α in T Helper 2 Cells Negatively Regulates Antitumor Immunity in Colorectal Cancer. Cancer Immunol Res 2019; 7:1332-1344. [PMID: 31175139 DOI: 10.1158/2326-6066.cir-18-0562] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Revised: 01/14/2019] [Accepted: 06/05/2019] [Indexed: 11/16/2022]
Abstract
The escape of cancer cells from host immunosurveillance involves a shift in immune responses, including an imbalance in Th1 and Th2 cells. A Th1-dominated immune response predicts positive outcomes in colorectal cancer. The E3 ubiquitin ligase, Asb2α, is expressed in Th2 cells, but its roles in T-cell maturation and cancer are unclear. We provide evidence that the Th2 master regulator, Gata3, induces Asb2 Loss of Asb2 did not affect Th differentiation ex vivo, but reduced IL4 production from Th2 cells. We found that high ASB2 expression was associated with poor outcome in colorectal cancer. Loss of Asb2 from hematopoietic cells promoted a Th1 response and attenuated colitis-associated tumorigenesis in mice. Diminished Th2 function correlated with increased IFNγ production and an enhanced type 1 antitumor immune response in Asb2-deficient mice. Our work suggests that Asb2α promotes a Th2 phenotype in vivo, which in turn is associated with tumor progression in a mouse model of colitis.
Collapse
Affiliation(s)
- Camille A Spinner
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Isabelle Lamsoul
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, UPS, Toulouse, France.
| | - Arnaud Métais
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Chanaëlle Febrissy
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Christel Moog-Lutz
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Pierre G Lutz
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, UPS, Toulouse, France.
| |
Collapse
|
39
|
Process of immunogenic cell death caused by disulfiram as the anti-colorectal cancer candidate. Biochem Biophys Res Commun 2019; 513:891-897. [DOI: 10.1016/j.bbrc.2019.03.192] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Accepted: 03/28/2019] [Indexed: 02/07/2023]
|
40
|
Marchiori C, Scarpa M, Kotsafti A, Morgan S, Fassan M, Guzzardo V, Porzionato A, Angriman I, Ruffolo C, Sut S, Dall'Acqua S, Bardini R, De Caro R, Castoro C, Scarpa M, Castagliuolo I. Epithelial CD80 promotes immune surveillance of colonic preneoplastic lesions and its expression is increased by oxidative stress through STAT3 in colon cancer cells. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:190. [PMID: 31072360 PMCID: PMC6509793 DOI: 10.1186/s13046-019-1205-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Accepted: 05/01/2019] [Indexed: 12/30/2022]
Abstract
BACKGROUND One of the most potent costimulatory molecules involved in the recognition and killing of tumor cells is CD80. However, its role and the molecular mechanisms regulating its expression in sporadic colorectal carcinogenesis remain elusive. Here, we provide evidence for CD80 overexpression in human colon epithelial cells derived from preneoplastic mucosa. METHODS Expression of CD80 on colonic epithelial cells isolated from normal human colonic mucosa, preneoplastic and neoplastic specimens was assessed by flow cytometry. WT and CD80KO mice received azoxymethane to induce colon preneoplastic lesions and sacrificed to perform histology, flow cytometry analysis and immunohistochemistry of colonic mucosa. Some WT mice were treated with a monoclonal anti-CD80 antibody following AOM administration. Primary colon epithelial cells and CT26 cell line were used to quantify the expression of CD80 in response to pro-oxidant stimuli. Specific pharmacological inhibitors and siRNA silencing were used to inhibit MAPK pathways and STAT3. RESULTS CD80 expression was significantly increased in colon epithelial cells of human preneoplastic lesions. In the AOM model, CD80 impairment by administration of neutralizing antibodies or use of CD80 knockout mice enhanced dysplasia development. In vitro, CD80 upregulation was induced by oxidative stress in colon cancer cells and primary colon epithelial cells. In addition, reactive oxygen species could induce CD80 expression via the JNK and p38 MAPK pathways, that activated STAT3 transcription factor in colon cancer epithelial cells. CONCLUSION This study provide evidence for a major role of CD80 in orchestrating immune surveillance of colon preneoplastic lesions and might help to develop novel approaches that exploit anti-tumor immunity to prevent and control colon cancer.
Collapse
Affiliation(s)
- Chiara Marchiori
- Department of Molecular Medicine DMM, University of Padua, Padua, Italy
| | - Melania Scarpa
- Laboratory of Advanced Translational Research, Veneto Institute of Oncology IOV - IRCCS, via Gattamelata 64, 35128, Padua, Italy.
| | - Andromachi Kotsafti
- Laboratory of Advanced Translational Research, Veneto Institute of Oncology IOV - IRCCS, via Gattamelata 64, 35128, Padua, Italy
| | - Susan Morgan
- Pathology Unit, Sheffield Teaching Hospitals, Sheffield, UK
| | - Matteo Fassan
- Surgical Pathology Unit from the Department of Medicine DIMED, University of Padua, Padua, Italy
| | - Vincenza Guzzardo
- Surgical Pathology Unit from the Department of Medicine DIMED, University of Padua, Padua, Italy
| | | | - Imerio Angriman
- General Surgery Unit from the Department of Surgery, Oncology and Gastroenterology DISCOG, University of Padua, Padua, Italy
| | - Cesare Ruffolo
- General Surgery Unit (IV), Ca' Foncello Hospital, Treviso, Italy
| | - Stefania Sut
- Department of Pharmaceutical and Pharmacological Sciences DSF, University of Padua, Padua, Italy
| | - Stefano Dall'Acqua
- Department of Pharmaceutical and Pharmacological Sciences DSF, University of Padua, Padua, Italy
| | - Romeo Bardini
- General Surgery Unit from the Department of Surgery, Oncology and Gastroenterology DISCOG, University of Padua, Padua, Italy
| | | | - Carlo Castoro
- Laboratory of Advanced Translational Research, Veneto Institute of Oncology IOV - IRCCS, via Gattamelata 64, 35128, Padua, Italy
| | - Marco Scarpa
- Laboratory of Advanced Translational Research, Veneto Institute of Oncology IOV - IRCCS, via Gattamelata 64, 35128, Padua, Italy
| | | |
Collapse
|
41
|
Burden of unique and low prevalence somatic mutations correlates with cancer survival. Sci Rep 2019; 9:4848. [PMID: 30890735 PMCID: PMC6425006 DOI: 10.1038/s41598-019-41015-5] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Accepted: 02/25/2019] [Indexed: 12/13/2022] Open
Abstract
Tumor mutational burden correlates with improved survival and immunotherapy response in some malignancies, and with tumor aggressiveness in others. To study the link between mutational burden and survival, we analyzed survival effects of tumor exonic missense mutation burden (TEMMB) across 6947 specimens spanning 31 cancers which have undergone whole exome sequencing as part of TCGA. We adjusted TEMMB for age, sex, stage, and recruitment center, and computed Cox-proportional models of TEMMB survival effects. We assigned a recurrence score (RS) to each cohort, defining RS as the burden of recurrent mutations exceeding 1% population prevalence. High TEMMB was associated with improved survival in cutaneous melanoma: hazard ratio (HR) = 0.71 [0.60–0.85], p = 0.0002, urothelial bladder carcinoma: HR = 0.74 [0.59–0.93], p = 0.01, and ovarian carcinoma: HR = 0.80 [0.70–0.93], p = 0.003. High TEMMB was associated with decreased survival in colorectal adenocarcinoma: HR = 1.32 [1.00–1.74], p < 0.05. We identified that TEMMB survival effects were governed by the balance of recurrent and non-recurrent mutations. In cancers with a low RS, high TEMMB was correlated with better survival outcomes (r = 0.49, p = 0.02). In conclusion, TEMMB effects on survival depend on recurrent mutation enrichment; tumor types that are highly enriched in passenger mutations show a survival benefit in the setting of high tumor mutational burden.
Collapse
|
42
|
Lo Nigro C, Macagno M, Sangiolo D, Bertolaccini L, Aglietta M, Merlano MC. NK-mediated antibody-dependent cell-mediated cytotoxicity in solid tumors: biological evidence and clinical perspectives. ANNALS OF TRANSLATIONAL MEDICINE 2019; 7:105. [PMID: 31019955 DOI: 10.21037/atm.2019.01.42] [Citation(s) in RCA: 135] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The process of antibody-dependent cell-mediated cytotoxicity (ADCC) makes use of the innate immune cells providing antitumor cytotoxicity activated by antibodies linked to target cells. Natural killer (NK) cells are a small set of lymphocytes, but are considered the most important cells among those able to induce ADCC. They provoke innate immune responses and harmonise spontaneous cytotoxicity towards tumor and virus-infected cells. They are able to swiftly produce biochemical signals and cytokines so as to stimulate subsequent adaptive immune responses. Immunotherapeutics that target NK cells, augmenting their immune response, can cause the antitumor dynamics of the antibodies to be improved. The recent developments in the field of NK cell immunotherapy and genotypic factors which might affect patient responses to antibody-dependent immunotherapies are the main subject of this review, with a particular focus on the manipulations and strategies used to augment ADCC. In the next years combined treatment with monoclonal antibodies (mAbs) and immunomodulatory drugs will be an important part in antitumor therapy. The main challenge remains the difficulty in distinguishing in the clinical setting, between the target effect that many mAbs exert against specific cell membrane receptors and the ADCC effect that they too also can induce. Drugs able to activate NK cells, that are major actors in mAb-mediated ADCC, will improve the ADCC effect against tumors.
Collapse
Affiliation(s)
- Cristiana Lo Nigro
- Department of Oncology, S. Croce & Carle Teaching Hospital, Cuneo, Italy
| | - Marco Macagno
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy
| | - Dario Sangiolo
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy.,Department of Oncology, University of Torino, Torino, Italy
| | - Luca Bertolaccini
- Department of Thoracic Surgery, AUSL Bologna Maggiore Teaching Hospital, Bologna, Italy
| | - Massimo Aglietta
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy.,Department of Oncology, University of Torino, Torino, Italy
| | | |
Collapse
|
43
|
Atreya I, Kindermann M, Wirtz S. Innate lymphoid cells in intestinal cancer development. Semin Immunol 2019; 41:101267. [PMID: 30772139 DOI: 10.1016/j.smim.2019.02.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2018] [Revised: 01/25/2019] [Accepted: 02/05/2019] [Indexed: 02/06/2023]
Abstract
Colorectal cancer (CRC) is a highly prominent cause of cancer-related deaths worldwide. Although the functions of immune cells in the colorectal tumor microenvironment are complex and heterogeneous, dysregulated changes in the composition and activation state of immune cells are believed to represent key events supporting the establishment of pro- or anti-tumorigenic immune states. Recently, innate lymphoid cells (ILCs) emerged as central innate immune mediators during both gastrointestinal homeostasis and inflammatory pathologies. Hence, ILCs might also represent promising targets in the context of cancer therapy and are increasingly recognized as innate immune cells with potent immunomodulatory properties. In this review, we summarize the pleiotropic roles of the different ILC subsets for intestinal homeostasis and discuss the recent evidence on their potential involvement in the development and growth of intestinal cancers.
Collapse
Affiliation(s)
- Imke Atreya
- Department of Medicine 1, Friedrich-Alexander-University, Erlangen, Germany
| | - Markus Kindermann
- Department of Medicine 1, Friedrich-Alexander-University, Erlangen, Germany
| | - Stefan Wirtz
- Department of Medicine 1, Friedrich-Alexander-University, Erlangen, Germany.
| |
Collapse
|
44
|
Rioux CR, Clapper ML, Cooper HS, Michaud J, St Amant N, Koohsari H, Workman L, Kaunga E, Hensley H, Pilorget A, Gerard C. Self-antigen MASH2 combined with the AS15 immunostimulant induces tumor protection in colorectal cancer mouse models. PLoS One 2019; 14:e0210261. [PMID: 30682058 PMCID: PMC6347180 DOI: 10.1371/journal.pone.0210261] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Accepted: 12/19/2018] [Indexed: 12/15/2022] Open
Abstract
Human achaete scute homolog 2 (HASH2) and its murine ortholog MASH2 are potential targets for colorectal cancer immunotherapy. We assessed immunogenicity and antitumor potential of recombinant MASH2 protein combined with AS15 immunostimulant (recMASH2+AS15) in CB6F1 and Apc+/Min-FCCC mice. CB6F1 mice received 4 injections of recMASH2+AS15 or AS15 alone before challenge with TC1-MASH2 tumor cells (Tumor Challenge). Apc+/Min-FCCC mice received 9 injections of recMASH2+AS15 or vehicle (phosphate buffer saline [PBS] or AS15 alone), before (two independent Prophylactic Studies) or after (Immunotherapy) colon adenomas were detectable by colonoscopy. CB6F1 mice immunized with recMASH2+AS15 had a significantly smaller mean tumor size and improved survival rate compared to controls (104 mm2 vs. 197 mm2 [p = 0.009] and 67% vs. 7% [p = 0.001], respectively). In Prophylactic Study 1, the mean number of colon adenomas was significantly lower in Apc+/Min-FCCC mice receiving recMASH2+AS15 compared to PBS (1.8 [95% confidence interval 1.0–3.3] vs. 5.2 [3.7–7.4], p = 0.003). Fewer microadenomas were observed in recMASH2+AS15 groups compared to PBS in both Prophylactic Studies (Study 1: mean 0.4 [0.2–1.0] vs. 1.5 [0.9–2.4], p = 0.009; Study 2: 0.4 [0.2–0.6] vs. 1.1 [0.8–1.5], p = 0.001). In the Immunotherapy Study, fewer colon adenomas tended to be observed in recMASH2+AS15-treated mice (4.1 [2.9–6.0]) compared to controls (AS15 4.7 [3.3–6.6]; PBS 4.9 [3.5–6.9]; no significant difference). recMASH2+AS15 induced MASH2-specific antibody and CD4+ responses in both mouse models. recMASH2+AS15 partially protected mice against MASH2-expressing tumors and reduced spontaneous colorectal adenomas in Apc+/Min-FCCC mice, indicating that MASH2/HASH2 antigens are targets for colorectal cancer immunotherapy.
Collapse
Affiliation(s)
| | - Margie L. Clapper
- Fox Chase Cancer Center, Philadelphia, PA, United States of America
- * E-mail:
| | - Harry S. Cooper
- Fox Chase Cancer Center, Philadelphia, PA, United States of America
| | | | | | | | - Laura Workman
- Fox Chase Cancer Center, Philadelphia, PA, United States of America
| | - Esther Kaunga
- Fox Chase Cancer Center, Philadelphia, PA, United States of America
| | - Harvey Hensley
- Fox Chase Cancer Center, Philadelphia, PA, United States of America
| | | | | |
Collapse
|
45
|
Lemdani K, Mignet N, Boudy V, Seguin J, Oujagir E, Bawa O, Peschaud F, Emile JF, Capron C, Malafosse R. Local immunomodulation combined to radiofrequency ablation results in a complete cure of local and distant colorectal carcinoma. Oncoimmunology 2019; 8:1550342. [PMID: 30723580 PMCID: PMC6350685 DOI: 10.1080/2162402x.2018.1550342] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Revised: 11/05/2018] [Accepted: 11/09/2018] [Indexed: 12/21/2022] Open
Affiliation(s)
- Katia Lemdani
- EA4340 BCOH, Versailles University, Paris-Saclay University, Boulogne, France
- Department of Surgery and Oncology, Centre Hospitalier Universitaire Ambroise Paré, Assistance Publique-Hôpitaux de Paris, Boulogne, France
- CNRS, UTCBS UMR 8258, Paris, France
- Paris Descartes University, Sorbonne-Paris-Cité, Paris, France
- INSERM, UTCBS U1022, Paris, France
| | - Nathalie Mignet
- CNRS, UTCBS UMR 8258, Paris, France
- Paris Descartes University, Sorbonne-Paris-Cité, Paris, France
- INSERM, UTCBS U1022, Paris, France
- Chimie ParisTech, PSL Research University, Unité de Technologies Chimiques et Biologiques pour la Santé (UTCBS), Paris, France
| | - Vincent Boudy
- CNRS, UTCBS UMR 8258, Paris, France
- Paris Descartes University, Sorbonne-Paris-Cité, Paris, France
- INSERM, UTCBS U1022, Paris, France
- Chimie ParisTech, PSL Research University, Unité de Technologies Chimiques et Biologiques pour la Santé (UTCBS), Paris, France
- Innovation in Pharmaceutical Formulation Department, Agence Générale des Equipements et des Produits de Santé (AGEPS), AP-HP, Paris, France
| | - Johanne Seguin
- CNRS, UTCBS UMR 8258, Paris, France
- Paris Descartes University, Sorbonne-Paris-Cité, Paris, France
- INSERM, UTCBS U1022, Paris, France
- Chimie ParisTech, PSL Research University, Unité de Technologies Chimiques et Biologiques pour la Santé (UTCBS), Paris, France
- Optical imaging platform LIOPA, Life imaging, PIV, Faculty of Pharmacy, Paris Descartes University, Paris, France
| | | | - Olivia Bawa
- Institut Gustave Roussy, Unité de pathologie expérimentale de l’IRCIV, Villejuif, France
| | - Frédérique Peschaud
- EA4340 BCOH, Versailles University, Paris-Saclay University, Boulogne, France
- Department of Surgery and Oncology, Centre Hospitalier Universitaire Ambroise Paré, Assistance Publique-Hôpitaux de Paris, Boulogne, France
| | - Jean-François Emile
- EA4340 BCOH, Versailles University, Paris-Saclay University, Boulogne, France
- Pathology Department, Ambroise Paré Hospital, AP-HP, Boulogne, France
| | - Claude Capron
- EA4340 BCOH, Versailles University, Paris-Saclay University, Boulogne, France
- Immunology and hematology Department, Ambroise Paré Hospital, Boulogne, France
| | - Robert Malafosse
- EA4340 BCOH, Versailles University, Paris-Saclay University, Boulogne, France
- Department of Surgery and Oncology, Centre Hospitalier Universitaire Ambroise Paré, Assistance Publique-Hôpitaux de Paris, Boulogne, France
| |
Collapse
|
46
|
de Jesus VHF, Felismino TC, de Barros e Silva MJ, de Souza e Silva V, Riechelmann RP. Current approaches to immunotherapy in noncolorectal gastrointestinal malignancies. Clinics (Sao Paulo) 2018; 73:e510s. [PMID: 30365605 PMCID: PMC6173942 DOI: 10.6061/clinics/2018/e510s] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Accepted: 07/02/2018] [Indexed: 11/24/2022] Open
Abstract
Noncolorectal gastrointestinal (GI) malignancies are among the most frequently diagnosed cancers. Despite the undeniable progress in systemic treatments in recent decades, further improvements using cytotoxic chemotherapy seem unlikely. In this setting, recent discoveries regarding the mechanism underlying immune evasion have prompted the study of molecules capable of inducing strong antitumor responses. Thus, according to early data, immunotherapy is a very promising tool for the treatment of patients with GI malignancies. Noncolorectal GI cancers are a major public health problem worldwide. Traditional treatment options, such as chemotherapy, surgery, radiation therapy, monoclonal antibodies and antiangiogenic agents, have been the backbone of treatment for various stages of GI cancers, but overall mortality remains a major problem. Thus, there is a substantial unmet need for new drugs and therapies to further improve the outcomes of treatment for noncolorectal GI malignancies. "Next-generation" immunotherapy is emerging as an effective and promising treatment option in several types of cancers. Therefore, encouraged by this recent success, many clinical trials evaluating the efficacy of immune checkpoint inhibitors and other strategies in treating noncolorectal GI malignancies are ongoing. This review will summarize the current clinical progress of modern immunotherapy in the field of noncolorectal GI tumors.
Collapse
Affiliation(s)
| | | | | | | | - Rachel P Riechelmann
- Departamento de Oncologia Médica, A.C. Camargo Cancer Center, Sao Paulo, SP, BR
- Departamento de Radiologia e Oncologia, Instituto do Cancer do Estado de Sao Paulo (ICESP), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, BR
| |
Collapse
|
47
|
Zhang Q, Berger FG, Love B, Banister CE, Murphy EA, Hofseth LJ. Maternal stress and early-onset colorectal cancer. Med Hypotheses 2018; 121:152-159. [PMID: 30396471 DOI: 10.1016/j.mehy.2018.09.035] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Revised: 09/10/2018] [Accepted: 09/20/2018] [Indexed: 02/07/2023]
Abstract
Early-onset colorectal cancer (EOCRC) is defined as colorectal cancer (CRC) diagnosed before the age of 50. Alarmingly, there has been a significant increase in EOCRC diagnoses' worldwide over the past several decades. Emerging data suggest EOCRCs have distinguishing clinical, pathological, biological and molecular features; and thus, are a fundamentally different subtype of CRCs. Unfortunately, there is no simple explanation for the causes of EOCRC. Scientifically rigorous studies are needed to determine what may be driving the challenging epidemiology of EOCRC. We contend here that a reasonable hypothesis is that prenatal risk factors such as maternal stress and associated sleeping disorders influence offspring epigenetic make-up, and shape immune system and gut health contributing to an increased risk for EOCRC.
Collapse
Affiliation(s)
- Qi Zhang
- Department of Drug Discovery and Biomedical Science, College of Pharmacy, University of South Carolina, Columbia, SC, USA
| | - Franklin G Berger
- Department of Biology, College of Arts and Sciences, University of South Carolina, Columbia, SC, USA
| | - Bryan Love
- Department of Clinical Pharmacy & Outcomes Sciences, College of Pharmacy, University of South Carolina, Columbia, SC, USA
| | - Carolyn E Banister
- Department of Drug Discovery and Biomedical Science, College of Pharmacy, University of South Carolina, Columbia, SC, USA
| | - Elizabeth A Murphy
- Department of Pathology, Microbiology and Immunology, University of South Carolina, Columbia, SC, USA
| | - Lorne J Hofseth
- Department of Drug Discovery and Biomedical Science, College of Pharmacy, University of South Carolina, Columbia, SC, USA.
| |
Collapse
|
48
|
A diagnostic autoantibody signature for primary cutaneous melanoma. Oncotarget 2018; 9:30539-30551. [PMID: 30093967 PMCID: PMC6078131 DOI: 10.18632/oncotarget.25669] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Accepted: 06/04/2018] [Indexed: 01/16/2023] Open
Abstract
Melanoma is an aggressive form of skin cancer that is curable by surgical excision in the majority of cases, if detected at an early stage. To improve early stage melanoma detection, the development of a highly sensitive diagnostic test is of utmost importance. Here we aimed to identify antibodies to a panel of tumour associated antigens that can differentiate primary melanoma patients and healthy individuals. A total of 245 sera from primary melanoma patients and healthy volunteers were screened against a high-throughput microarray platform containing 1627 functional proteins. Following rigorous statistical analysis, we identified a combination of 10 autoantibody biomarkers that, as a panel, displays a sensitivity of 79%, specificity of 84% and an AUC of 0.828 for primary melanoma detection. This melanoma autoantibody signature may prove valuable for the development of a diagnostic blood test for routine population screening that, when used in conjunction with current melanoma diagnostic techniques, could improve the early diagnosis of this malignancy and ultimately decrease the mortality rate of patients.
Collapse
|
49
|
Liu J, Wang L, Wang T, Wang J. Expression of IL-23R and IL-17 and the pathology and prognosis of urinary bladder carcinoma. Oncol Lett 2018; 16:4325-4330. [PMID: 30214568 PMCID: PMC6126236 DOI: 10.3892/ol.2018.9145] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Accepted: 07/02/2018] [Indexed: 12/23/2022] Open
Abstract
Expression of interleukin-23 receptor (IL-23R) and IL-17 in urinary bladder carcinoma (UBC) was investigated to explore the correlations with prognosis. IL-23/IL-17 axis significantly inhibited the development of inflammatory bowel disease. Thirty patients with UBC were enrolled in Zhengzhou Central Hospital Affiliated to Zhengzhou University from September 2013 to September 2014. Tumor tissue and adjacent healthy tissue were collected, and the levels of IL-23R and IL-17 mRNA were detected by RT-PCR. Thirty healthy people were also selected to serve as normal control group. Serum levels of IL-23R and IL-17 in serum of UBC patients and normal controls were detected by ELISA, and the correlations with clinical features of UBC were analyzed. Pearson's correlation analysis was used to analyze the correlation between IL-23R and IL-17 protein expression. Follow-up study was performed by phone or during patient's visit to out-patient department. Overall survival (OS) and disease-free survival (DFS) curves were plotted by Kaplan-Meier method to analyze the correlation between expression of IL-23R and IL-17 and survival time. ROC curve was used to detect the diagnostic values of IL-23R and IL-17 protein for UBC. Levels of IL-23R and IL-17 mRNA in UBC tissue were 3.26 and 2.65 times higher than those in adjacent tissue (P<0.05), and serum levels of IL-23R and IL-17 protein in UBC patients were significantly higher than those in normal control group. Protein expression levels of IL-23R and IL-17 were correlated with clinical stage and lymph node metastasis in UBC patients (P<0.05), and Cox hazard model showed that L-23R and IL-17 expression may be independent factors for UBC (P<0.05), and high expression levels of IL-23R and IL-17 significantly shortened the OS and DFS (P<0.05). Serum levels of IL-23R and IL-17 can be used to effectively diagnose clinical stage and lymph node metastasis of UBC patients, and the combined diagnosis has a higher sensitivity and specificity than the diagnosis using a single factor. These findings indicated that expression levels of IL-23R and IL-17 were increased in tumor tissue and serum of UBC patients, and the increased expression levels of IL-23R and IL-17 were correlated with poor prognosis. Detection of IL-23R and IL-17 levels has certain clinical significance in the diagnosis and prognosis of UBC.
Collapse
Affiliation(s)
- Jian Liu
- Department of Urology, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, Henan 450000, P.R. China
| | - Lei Wang
- Department of Urology, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, Henan 450000, P.R. China
| | - Tongqing Wang
- Department of Urology, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, Henan 450000, P.R. China
| | - Jizheng Wang
- Department of Urology, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, Henan 450000, P.R. China
| |
Collapse
|
50
|
Proquin H, Jetten MJ, Jonkhout MCM, Garduño-Balderas LG, Briedé JJ, de Kok TM, van Loveren H, Chirino YI. Transcriptomics analysis reveals new insights in E171-induced molecular alterations in a mouse model of colon cancer. Sci Rep 2018; 8:9738. [PMID: 29950665 PMCID: PMC6021444 DOI: 10.1038/s41598-018-28063-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2017] [Accepted: 06/15/2018] [Indexed: 12/19/2022] Open
Abstract
Titanium dioxide as a food additive (E171) has been demonstrated to facilitate growth of chemically induced colorectal tumours in vivo and induce transcriptomic changes suggestive of an immune system impairment and cancer development. The present study aimed to investigate the molecular mechanisms behind the tumour stimulatory effects of E171 in combination with azoxymethane (AOM)/dextran sodium sulphate (DSS) and compare these results to a recent study performed under the same conditions with E171 only. BALB/c mice underwent exposure to 5 mg/kgbw/day of E171 by gavage for 2, 7, 14, and 21 days. Whole genome mRNA microarray analyses on the distal colon were performed. The results show that E171 induced a downregulation of genes involved in the innate and adaptive immune system, suggesting impairment of this system. In addition, over time, signalling genes involved in colorectal cancer and other types of cancers were modulated. In relation to cancer development, effects potentially associated with oxidative stress were observed through modulation of genes related to antioxidant production. E171 affected genes involved in biotransformation of xenobiotics which can form reactive intermediates resulting in toxicological effects. These transcriptomics data reflect the early biological responses induced by E171 which precede tumour formation in an AOM/DSS mouse model.
Collapse
Affiliation(s)
- Héloïse Proquin
- Department of Toxicogenomics, GROW institute of Oncology and Developmental Biology, Maastricht University, Maastricht, The Netherlands.
| | - Marlon J Jetten
- Department of Toxicogenomics, GROW institute of Oncology and Developmental Biology, Maastricht University, Maastricht, The Netherlands
| | - Marloes C M Jonkhout
- Department of Toxicogenomics, GROW institute of Oncology and Developmental Biology, Maastricht University, Maastricht, The Netherlands
| | | | - Jacob J Briedé
- Department of Toxicogenomics, GROW institute of Oncology and Developmental Biology, Maastricht University, Maastricht, The Netherlands
| | - Theo M de Kok
- Department of Toxicogenomics, GROW institute of Oncology and Developmental Biology, Maastricht University, Maastricht, The Netherlands
| | - Henk van Loveren
- Department of Toxicogenomics, GROW institute of Oncology and Developmental Biology, Maastricht University, Maastricht, The Netherlands
| | - Yolanda I Chirino
- Laboratorio de Carcinogénesis y Toxicología, Unidad de Biomedicina, FES-Iztacala, UNAM, Estado de México, Mexico.,IUF-Leibniz Research Institute for Environmental Medicine, Auf'm Hennekamp 50, 40225, DE Düsseldorf, Germany
| |
Collapse
|