51
|
Hanus M, Parada-Venegas D, Landskron G, Wielandt AM, Hurtado C, Alvarez K, Hermoso MA, López-Köstner F, De la Fuente M. Immune System, Microbiota, and Microbial Metabolites: The Unresolved Triad in Colorectal Cancer Microenvironment. Front Immunol 2021; 12:612826. [PMID: 33841394 PMCID: PMC8033001 DOI: 10.3389/fimmu.2021.612826] [Citation(s) in RCA: 81] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 03/02/2021] [Indexed: 12/16/2022] Open
Abstract
Colorectal cancer (CRC) is one of the most common cancers worldwide. As with other cancers, CRC is a multifactorial disease due to the combined effect of genetic and environmental factors. Most cases are sporadic, but a small proportion is hereditary, estimated at around 5-10%. In both, the tumor interacts with heterogeneous cell populations, such as endothelial, stromal, and immune cells, secreting different signals (cytokines, chemokines or growth factors) to generate a favorable tumor microenvironment for cancer cell invasion and metastasis. There is ample evidence that inflammatory processes have a role in carcinogenesis and tumor progression in CCR. Different profiles of cell activation of the tumor microenvironment can promote pro or anti-tumor pathways; hence they are studied as a key target for the control of cancer progression. Additionally, the intestinal mucosa is in close contact with a microorganism community, including bacteria, bacteriophages, viruses, archaea, and fungi composing the gut microbiota. Aberrant composition of this microbiota, together with alteration in the diet-derived microbial metabolites content (such as butyrate and polyamines) and environmental compounds has been related to CRC. Some bacteria, such as pks+ Escherichia coli or Fusobacterium nucleatum, are involved in colorectal carcinogenesis through different pathomechanisms including the induction of genetic mutations in epithelial cells and modulation of tumor microenvironment. Epithelial and immune cells from intestinal mucosa have Pattern-recognition receptors and G-protein coupled receptors (receptor of butyrate), suggesting that their activation can be regulated by intestinal microbiota and metabolites. In this review, we discuss how dynamics in the gut microbiota, their metabolites, and tumor microenvironment interplays in sporadic and hereditary CRC, modulating tumor progression.
Collapse
Affiliation(s)
- Michelle Hanus
- Laboratory of Innate Immunity, Program of Immunology, Faculty of Medicine, Institute of Biomedical Sciences, Universidad de Chile, Santiago, Chile
| | - Daniela Parada-Venegas
- Laboratory of Innate Immunity, Program of Immunology, Faculty of Medicine, Institute of Biomedical Sciences, Universidad de Chile, Santiago, Chile
| | - Glauben Landskron
- Laboratory of Innate Immunity, Program of Immunology, Faculty of Medicine, Institute of Biomedical Sciences, Universidad de Chile, Santiago, Chile
| | | | - Claudia Hurtado
- Research Core, Academic Department, Clínica Las Condes, Santiago, Chile
| | - Karin Alvarez
- Cancer Center, Clínica Universidad de los Andes, Santiago, Chile
| | - Marcela A. Hermoso
- Laboratory of Innate Immunity, Program of Immunology, Faculty of Medicine, Institute of Biomedical Sciences, Universidad de Chile, Santiago, Chile
| | | | | |
Collapse
|
52
|
Ahmad R, Singh JK, Wunnava A, Al-Obeed O, Abdulla M, Srivastava SK. Emerging trends in colorectal cancer: Dysregulated signaling pathways (Review). Int J Mol Med 2021; 47:14. [PMID: 33655327 PMCID: PMC7834960 DOI: 10.3892/ijmm.2021.4847] [Citation(s) in RCA: 93] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 12/14/2020] [Indexed: 02/06/2023] Open
Abstract
Colorectal cancer (CRC) is the third most frequently detected type of cancer, and the second most common cause of cancer‑related mortality globally. The American Cancer Society predicted that approximately 147,950 individuals would be diagnosed with CRC, out of which 53,200 individuals would succumb to the disease in the USA alone in 2020. CRC‑related mortality ranks third among both males and females in the USA. CRC arises from 3 major pathways: i) The adenoma‑carcinoma sequence; ii) serrated pathway; and iii) the inflammatory pathway. The majority of cases of CRC are sporadic and result from risk factors, such as a sedentary lifestyle, obesity, processed diets, alcohol consumption and smoking. CRC is also a common preventable cancer. With widespread CRC screening, the incidence and mortality from CRC have decreased in developed countries. However, over the past few decades, CRC cases and mortality have been on the rise in young adults (age, <50 years). In addition, CRC cases are increasing in developing countries with a low gross domestic product (GDP) due to lifestyle changes. CRC is an etiologically heterogeneous disease classified by tumor location and alterations in global gene expression. Accumulating genetic and epigenetic perturbations and aberrations over time in tumor suppressor genes, oncogenes and DNA mismatch repair genes could be a precursor to the onset of colorectal cancer. CRC can be divided as sporadic, familial, and inherited depending on the origin of the mutation. Germline mutations in APC and MLH1 have been proven to play an etiological role, resulting in the predisposition of individuals to CRC. Genetic alterations cause the dysregulation of signaling pathways leading to drug resistance, the inhibition of apoptosis and the induction of proliferation, invasion and migration, resulting in CRC development and metastasis. Timely detection and effective precision therapies based on the present knowledge of CRC is essential for successful treatment and patient survival. The present review presents the CRC incidence, risk factors, dysregulated signaling pathways and targeted therapies.
Collapse
Affiliation(s)
- Rehan Ahmad
- Colorectal Research Chair, Department of Surgery, King Saud University College of Medicine, Riyadh 11472, Saudi Arabia
| | - Jaikee Kumar Singh
- Department of Biosciences, Manipal University Jaipur, Jaipur, Rajasthan 303007, India
| | - Amoolya Wunnava
- Department of Biosciences, Manipal University Jaipur, Jaipur, Rajasthan 303007, India
| | - Omar Al-Obeed
- Colorectal Research Chair, Department of Surgery, King Saud University College of Medicine, Riyadh 11472, Saudi Arabia
| | - Maha Abdulla
- Colorectal Research Chair, Department of Surgery, King Saud University College of Medicine, Riyadh 11472, Saudi Arabia
| | | |
Collapse
|
53
|
Heo G, Lee Y, Im E. Interplay between the Gut Microbiota and Inflammatory Mediators in the Development of Colorectal Cancer. Cancers (Basel) 2021; 13:734. [PMID: 33578830 PMCID: PMC7916585 DOI: 10.3390/cancers13040734] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 02/05/2021] [Accepted: 02/06/2021] [Indexed: 02/08/2023] Open
Abstract
Inflammatory mediators modulate inflammatory pathways during the development of colorectal cancer. Inflammatory mediators secreted by both immune and tumor cells can influence carcinogenesis, progression, and tumor metastasis. The gut microbiota, which colonize the entire intestinal tract, especially the colon, are closely linked to colorectal cancer through an association with inflammatory mediators such as tumor necrosis factor, nuclear factor kappa B, interleukins, and interferons. This association may be a potential therapeutic target, since therapeutic interventions targeting the gut microbiota have been actively investigated in both the laboratory and in clinics and include fecal microbiota transplantation and probiotics.
Collapse
Affiliation(s)
| | | | - Eunok Im
- College of Pharmacy, Pusan National University, Busan 46241, Korea; (G.H.); (Y.L.)
| |
Collapse
|
54
|
Jin KT, Chen B, Liu YY, Lan HUR, Yan JP. Monoclonal antibodies and chimeric antigen receptor (CAR) T cells in the treatment of colorectal cancer. Cancer Cell Int 2021; 21:83. [PMID: 33522929 PMCID: PMC7851946 DOI: 10.1186/s12935-021-01763-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 01/06/2021] [Indexed: 02/07/2023] Open
Abstract
Colorectal cancer (CRC) is the third most common cancer and the second leading cause of cancer deaths worldwide. Besides common therapeutic approaches, such as surgery, chemotherapy, and radiotherapy, novel therapeutic approaches, including immunotherapy, have been an advent in CRC treatment. The immunotherapy approaches try to elicit patients` immune responses against tumor cells to eradicate the tumor. Monoclonal antibodies (mAbs) and chimeric antigen receptor (CAR) T cells are two branches of cancer immunotherapy. MAbs demonstrate the great ability to completely recognize cancer cell-surface receptors and blockade proliferative or inhibitory pathways. On the other hand, T cell activation by genetically engineered CAR receptor via the TCR/CD3 and costimulatory domains can induce potent immune responses against specific tumor-associated antigens (TAAs). Both of these approaches have beneficial anti-tumor effects on CRC. Herein, we review the different mAbs against various pathways and their applications in clinical trials, the different types of CAR-T cells, various specific CAR-T cells against TAAs, and their clinical use in CRC treatment.
Collapse
Affiliation(s)
- Ke-Tao Jin
- Department of Colorectal Surgery, Affiliated Jinhua Hosptial, Zhejiang University School of Medicine, Zhejiang Province, Jinhua, 312000, P.R. China
| | - Bo Chen
- Department of Neurology, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, 310014, China
| | - Yu-Yao Liu
- Department of Colorectal Surgery, Affiliated Jinhua Hosptial, Zhejiang University School of Medicine, Zhejiang Province, Jinhua, 312000, P.R. China
| | - H Uan-Rong Lan
- Department of Breast and Thyroid Surgery, Affiliated Jinhua Hosptial, Zhejiang University School of Medicine, Zhejiang Province, Jinhua, 312000, P.R. China
| | - Jie-Ping Yan
- Department of Pharmacy, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, No. 158 Shangtang Road, Hangzhou, 310014, China.
| |
Collapse
|
55
|
Huang H, Zhang D, Fu J, Zhao L, Li D, Sun H, Liu X, Xu J, Tian T, Zhang L, Liu Y, Zhang Y, Zhao Y. Tsukushi is a novel prognostic biomarker and correlates with tumor-infiltrating B cells in non-small cell lung cancer. Aging (Albany NY) 2021; 13:4428-4451. [PMID: 33428594 PMCID: PMC7906171 DOI: 10.18632/aging.202403] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 11/03/2020] [Indexed: 01/21/2023]
Abstract
A recent study has reported that tsukushi (TSKU) may be related to the development of lung cancer. However, few studies focused on if TSKU associated with the prognosis and immune infiltration cells in non-small cell lung cancer (NSCLC). The effect of TSKU expression on prognosis with NSCLC was analyzed in the PrognoScan database and validated in The Cancer Genome Atlas. The composition of tumor infiltrating cells was quantified by methylation and expression data. We combined levels of tumor infiltrating cells with TSKU to evaluate the survival of patients. The analysis of a cohort (GSE31210, N=204) of lung cancer patients demonstrated that high TSKU expression was strongly associated with poor overall survival (P =1.90E-05). The combination of high TSKU expression and low infiltration B cells identified a subtype of patients with poor survival in NSCLC. Besides, the proportion of B cells in NSCLC patients with TSKU hypermethylation were higher than those patients with TSKU hypomethylation (P <0.001). Overall, high TSKU expression combined with low infiltration of B cells may associate with a poor prognosis of NSCLC patients. TSKU might be a potential prognostic biomarker involved in tumor immune infiltration in NSCLC.
Collapse
Affiliation(s)
- Hao Huang
- Department of Epidemiology, Public Health College, Harbin Medical University, Harbin, Heilongjiang Province, People's Republic of China
| | - Ding Zhang
- Department of Epidemiology, Public Health College, Harbin Medical University, Harbin, Heilongjiang Province, People's Republic of China
| | - Jinming Fu
- Department of Epidemiology, Public Health College, Harbin Medical University, Harbin, Heilongjiang Province, People's Republic of China
| | - Liyuan Zhao
- Department of Epidemiology, Public Health College, Harbin Medical University, Harbin, Heilongjiang Province, People's Republic of China
| | - Dapeng Li
- 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
| | - Xinyan 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
| | - Tian Tian
- 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
| | - Ying Liu
- 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
| | - Yashuang Zhao
- Department of Epidemiology, Public Health College, Harbin Medical University, Harbin, Heilongjiang Province, People's Republic of China
| |
Collapse
|
56
|
Gamez-Belmonte R, Erkert L, Wirtz S, Becker C. The Regulation of Intestinal Inflammation and Cancer Development by Type 2 Immune Responses. Int J Mol Sci 2020; 21:ijms21249772. [PMID: 33371444 PMCID: PMC7767427 DOI: 10.3390/ijms21249772] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 12/17/2020] [Accepted: 12/18/2020] [Indexed: 12/11/2022] Open
Abstract
The gut is among the most complex organs of the human body. It has to exert several functions including food and water absorption while setting up an efficient barrier to the outside world. Dysfunction of the gut can be life-threatening. Diseases of the gastrointestinal tract such as inflammatory bowel disease, infections, or colorectal cancer, therefore, pose substantial challenges to clinical care. The intestinal epithelium plays an important role in intestinal disease development. It not only establishes an important barrier against the gut lumen but also constantly signals information about the gut lumen and its composition to immune cells in the bowel wall. Such signaling across the epithelial barrier also occurs in the other direction. Intestinal epithelial cells respond to cytokines and other mediators of immune cells in the lamina propria and shape the microbial community within the gut by producing various antimicrobial peptides. Thus, the epithelium can be considered as an interpreter between the microbiota and the mucosal immune system, safeguarding and moderating communication to the benefit of the host. Type 2 immune responses play important roles in immune-epithelial communication. They contribute to gut tissue homeostasis and protect the host against infections with helminths. However, they are also involved in pathogenic pathways in inflammatory bowel disease and colorectal cancer. The current review provides an overview of current concepts regarding type 2 immune responses in intestinal physiology and pathophysiology.
Collapse
|
57
|
Zhang M, Lu X, Wei C, Li X. Association between αβ and γδ T-cell subsets and clinicopathological characteristics in patients with breast cancer. Oncol Lett 2020; 20:325. [PMID: 33123241 PMCID: PMC7584013 DOI: 10.3892/ol.2020.12188] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Accepted: 09/14/2020] [Indexed: 12/30/2022] Open
Abstract
The aim of the present study was to discuss the effect of surgery on the T-lymphocyte subsets of patients with breast cancer (BC) and investigate the association between peripheral blood αβ and γδ T-cell counts and the clinicopathological characteristics of BC. The CD3+, CD4+, CD8+ and γδ T-cell subsets in the peripheral blood of healthy volunteers and Patients with BC before and after surgery were determined using flow cytometry. The association between αβ and γδ T-cell counts in the peripheral blood and clinicopathological characteristics was analyzed by comparing the differences in the αβ and γδ T-cell counts in the peripheral blood of Patients with BC before and after surgery with those of healthy volunteers and combining with clinicopathological data. The CD3+, CD4+ and γδ T-cell counts in the peripheral blood of Patients with BC were lower compared with those in healthy volunteers (P=0.0077, 0.0116 and 0.0003, respectively), whereas the number of CD8+ cells was higher (P=0.0241). The CD3+, CD4+ and γδ T-cell counts and the CD4+/CD8+ ratio after surgery were significantly higher compared with those before surgery (P=0.0109, 0.0031, 0.0165 and 0.018, respectively). There was no significant difference between the number of CD8+ cells before and after surgery (P=0.0053), but the number of CD8+ cells was higher in healthy volunteers compared with that in Patients with BC (P<0.05). Moreover, the CD3+ cell number was higher in patients with TNM stage II/III compared with those with TNM stage I disease (P=0.187 and 0.022, respectively), and the peripheral blood CD4+/CD8+ ratio and number of γδ T cells were lower in stage III compared with stage I Patients with BC (P=0.0065 and 0.0176, respectively). Histological grading demonstrated that the CD4+/CD8+ ratio and number of γδ T cells in patients with stage III BC were lower compared with those with stage I BC (P=0.02 and 0.0128, respectively). The γδ T-cell count in patients with luminal A and B subtypes was significantly higher compared with that in patients with basal-like subtype (P=0.004 and 0.0104, respectively). The CD3+, CD4+ and γδ T-cell counts were significantly lower in patients with lymph node (LN) metastasis compared with those without LN metastasis, and the CD8+ cell number was lower in patients without LN metastasis compared with that in patients with >10 LN metastases (P=0.0086, 0.0000 and 0.00468, respectively). The CD8+ cell count in patients without LN metastasis was lower compared with that in patients with 4-9 and >10 LN metastases (P=0.0435 and 0.0283, respectively). Surgery affects the T-lymphocyte subpopulations in patients with BC, and αβ and γδ T-cell counts may increase following mastectomy. Therefore, measurement of peripheral blood lymphocyte subsets is crucial for understanding the immune function status of Patients with BC with differences in TNM stage, histological grade, cell subtypes and LN metastases, and may provide a basis for the application of T-cell subsets in the comprehensive treatment of BC.
Collapse
Affiliation(s)
- Meng Zhang
- First Clinical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250355, P.R. China
| | - Xueling Lu
- Department of Nuclear Medicine, Tai'an City Central Hospital, Tai'an, Shandong 271000, P.R. China
| | - Changran Wei
- First Clinical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250355, P.R. China
| | - Xiangqi Li
- Department of Breast Surgery, The Second Affiliated Hospital of Shandong First Medical University, Tai'an, Shandong 271000, P.R. China
| |
Collapse
|
58
|
Oyenuga M, Vierkant RA, Lynch CF, Pengo T, Tillmans LS, Cerhan JR, Church TR, Lazovich D, Anderson KE, Limburg PJ, Prizment AE. Associations between tissue-based CD3+ T-lymphocyte count and colorectal cancer survival in a prospective cohort of older women. Mol Carcinog 2020; 60:15-24. [PMID: 33200476 DOI: 10.1002/mc.23267] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 10/18/2020] [Accepted: 10/28/2020] [Indexed: 12/11/2022]
Abstract
Tumor-infiltrating lymphocytes in colorectal cancer (CRC) predict better survival. However, associations between T-lymphocyte count in histologically normal tissues from patients with CRC and survival remain uncertain. We examined associations of CD3+ T-cells in colorectal tumor and histologically normal tissues with CRC-specific and all-cause mortality in the prospective Iowa Women's Health Study. Tissue microarrays were constructed using paraffin-embedded colorectal tissue samples from 464 women with tumor tissues and 314 women with histologically normal tissues (55-69 years at baseline) diagnosed with incident CRC from 1986 to 2002 and followed through 2014 (median follow-up 20.5 years). Three tumor and two histologically normal tissue cores for each patient were immunostained using CD3+ antibody and quantified, and the counts were averaged across the cores in each tissue. Cox proportional hazards regression estimated hazard ratios (HR) and 95% confidence interval (CI) for CRC-specific and all-cause mortality. After adjustment for age at diagnosis, body mass index, smoking status, tumor grade, and stage, HRs (95% CI) for the highest versus lowest tertile of tumor CD3+ score were 0.59 (0.38-0.89) for CRC-specific mortality and 0.82 (0.63-1.05) for all-cause mortality; for histologically normal CD3+ score, the corresponding HRs (95% CI) were 0.47 (0.19-1.17) and 0.50 (0.27-0.90), respectively. The CD3+ score combining the tumor and histologically normal scores was inversely associated with CRC-specific and all-cause mortality. Although the association between tumor CD3+ score and all-cause mortality was not significant, both higher CD3+ T-lymphocyte counts in tumor and histologically normal scores tended to be associated with lower CRC-specific and all-cause mortality.
Collapse
Affiliation(s)
- Mosunmoluwa Oyenuga
- Department of Internal Medicine, SSM St Mary's Hospital, St. Louis, Missouri, USA.,Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, Minnesota, USA
| | - Robert A Vierkant
- Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, Minnesota, USA
| | - Charles F Lynch
- Department of Epidemiology, University of Iowa, Iowa City, Iowa, USA
| | - Thomas Pengo
- University of Minnesota Informatics Institute, University of Minnesota, Minneapolis, Minnesota, USA
| | - Lori S Tillmans
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, USA
| | - James R Cerhan
- Division of Epidemiology, Mayo Clinic, Rochester, Minnesota, USA
| | - Timothy R Church
- Division of Environmental Health Sciences, School of Public Health, University of Minnesota, Minneapolis, Minnesota, USA
| | - DeAnn Lazovich
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, Minnesota, USA.,Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA
| | - Kristin E Anderson
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, Minnesota, USA.,Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA
| | - Paul J Limburg
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Anna E Prizment
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, Minnesota, USA.,Division of Hematology, Oncology, and Transplantation, University of Minnesota Medical School, Minneapolis, Minnesota, USA.,Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|
59
|
Huang Q, Cao Y, Wang S, Zhu R. Creation of a Novel Inflammation-Based Score for Operable Colorectal Cancer Patients. J Inflamm Res 2020; 13:659-671. [PMID: 33116746 PMCID: PMC7547789 DOI: 10.2147/jir.s271541] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 09/09/2020] [Indexed: 12/22/2022] Open
Abstract
Aim Systemic inflammation has been implicated in the progression of patients with colorectal cancer (CRC). We evaluated the prognostic ability of a comprehensive score based on several inflammatory indexes in operable CRC patients. Patients and Methods Between July 2013 and September 2017, this study retrospectively identified 1279 CRC patients receiving radical surgery in Wuhan Union Hospital and randomly assigned them into training (N=921) and validation (N=358) sets. A novel score, the CRC-specific inflammatory index (CSII), was developed from a series of inflammatory indexes significantly associated with survival in patients with CRC. This novel score was then divided into three categories and compared to the well-known systematic inflammatory index (SII) and TNM stage. Finally, a survival nomogram was generated by combining the CSII and other informative clinical features. Results The CSII-OS was calculated as 1.110×lg ALRI + 1.082×CAR + 0.792×PI, while CSII-DFS was 1.709×lg ALRI + 1.033×CAR based on multivariable Cox regression analysis. Patients with high CSII experienced a worse OS (HR=23.72, 95% CI, 11.30-49.78, P <0.001) and worse DFS (HR=15.62, 95% CI, 6.95-35.08, P <0.001) compared to those in CRC patients with low CSII. Moreover, ROC analyses showed that the CSII possessed excellent performance (AUC=0.859) in predicting OS and DFS. The AUC of the OS nomogram based on CSII, TNM stage, and chemotherapy was 0.897, while that of the DFS nomogram based on CSII, T stage, and TNM stage was 0.873. High-quality calibration curves in both OS and DFS nomograms were observed. Verification in the validation dataset showed results consistent with those in the training dataset. Conclusion The CSII is a CRC-specific prognostic score based on the combination of available inflammatory indexes. High CSII is a strong predictor of worse survival outcomes. The CSII also exhibits better predictive performance compared to SII or TNM stage in operable CRC patients.
Collapse
Affiliation(s)
- Qian Huang
- Department of Pediatric, Zhongnan Hospital of Wuhan University, Wuhan, People's Republic of China
| | - Yinghao Cao
- Department of Colorectal Surgery and Gastroenterology, Wuhan Union Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Shouyi Wang
- Department of Pediatric, Zhongnan Hospital of Wuhan University, Wuhan, People's Republic of China
| | - Rui Zhu
- Department of Pediatric, Zhongnan Hospital of Wuhan University, Wuhan, People's Republic of China
| |
Collapse
|
60
|
Perillo F, Amoroso C, Strati F, Giuffrè MR, Díaz-Basabe A, Lattanzi G, Facciotti F. Gut Microbiota Manipulation as a Tool for Colorectal Cancer Management: Recent Advances in Its Use for Therapeutic Purposes. Int J Mol Sci 2020; 21:E5389. [PMID: 32751239 PMCID: PMC7432108 DOI: 10.3390/ijms21155389] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 07/27/2020] [Accepted: 07/28/2020] [Indexed: 02/06/2023] Open
Abstract
Colorectal cancer (CRC) is a multifaceted disease influenced by both environmental and genetic factors. A large body of literature has demonstrated the role of gut microbes in promoting inflammatory responses, creating a suitable microenvironment for the development of skewed interactions between the host and the gut microbiota and cancer initiation. Even if surgery is the primary therapeutic strategy, patients with advanced disease or cancer recurrence after surgery remain difficult to cure. Therefore, the gut microbiota has been proposed as a novel therapeutic target in light of recent promising data in which it seems to modulate the response to cancer immunotherapy. The use of microbe-targeted therapies, including antibiotics, prebiotics, live biotherapeutics, and fecal microbiota transplantation, is therefore considered to support current therapies in CRC management. In this review, we will discuss the importance of host-microbe interactions in CRC and how promoting homeostatic immune responses through microbe-targeted therapies may be useful in preventing/treating CRC development.
Collapse
Affiliation(s)
- Federica Perillo
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, 20139 Milan, Italy; (F.P.); (C.A.); (M.R.G.); (A.D.-B.); (G.L.)
| | - Chiara Amoroso
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, 20139 Milan, Italy; (F.P.); (C.A.); (M.R.G.); (A.D.-B.); (G.L.)
| | - Francesco Strati
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, 20139 Milan, Italy; (F.P.); (C.A.); (M.R.G.); (A.D.-B.); (G.L.)
| | - Maria Rita Giuffrè
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, 20139 Milan, Italy; (F.P.); (C.A.); (M.R.G.); (A.D.-B.); (G.L.)
| | - Angélica Díaz-Basabe
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, 20139 Milan, Italy; (F.P.); (C.A.); (M.R.G.); (A.D.-B.); (G.L.)
- Department of Oncology and Hemato-Oncology, Università degli Studi di Milano, 20135 Milan, Italy
| | - Georgia Lattanzi
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, 20139 Milan, Italy; (F.P.); (C.A.); (M.R.G.); (A.D.-B.); (G.L.)
- Department of Oncology and Hemato-Oncology, Università degli Studi di Milano, 20135 Milan, Italy
| | - Federica Facciotti
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, 20139 Milan, Italy; (F.P.); (C.A.); (M.R.G.); (A.D.-B.); (G.L.)
| |
Collapse
|
61
|
Fucà G, Guarini V, Antoniotti C, Morano F, Moretto R, Corallo S, Marmorino F, Lonardi S, Rimassa L, Sartore-Bianchi A, Borelli B, Tampellini M, Bustreo S, Claravezza M, Boccaccino A, Murialdo R, Zaniboni A, Tomasello G, Loupakis F, Adamo V, Tonini G, Cortesi E, de Braud F, Cremolini C, Pietrantonio F. The Pan-Immune-Inflammation Value is a new prognostic biomarker in metastatic colorectal cancer: results from a pooled-analysis of the Valentino and TRIBE first-line trials. Br J Cancer 2020; 123:403-409. [PMID: 32424148 PMCID: PMC7403416 DOI: 10.1038/s41416-020-0894-7] [Citation(s) in RCA: 170] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2020] [Revised: 04/03/2020] [Accepted: 04/22/2020] [Indexed: 11/25/2022] Open
Abstract
Background Immune-inflammatory biomarkers (IIBs) showed a prognostic relevance in patients with metastatic CRC (mCRC). We aimed at evaluating the prognostic power of a new comprehensive biomarker, the Pan-Immune-Inflammation Value (PIV), in patients with mCRC receiving first-line therapy. Methods In the present pooled-analysis, we included patients enrolled in the Valentino and TRIBE trials. PIV was calculated as: (neutrophil count × platelet count × monocyte count)/lymphocyte count. A cut-off was determined using the maximally selected rank statistics method. Generalised boosted regression (GBR), the Kaplan–Meier method and Cox hazards regression models were used for survival analyses. Results A total of 438 patients were included. Overall, 208 patients (47%) had a low-baseline PIV and 230 (53%) had a high-baseline PIV. Patients with high PIV experienced a worse PFS (HR, 1.66; 95% CI, 1.36–2.03, P < 0.001) and worse OS (HR, 2.01; 95% CI, 1.57–2.57; P < 0.001) compared to patients with low PIV. PIV outperformed the other IIBs in the GBR model and in the multivariable models. Conclusion PIV is a strong predictor of survival outcomes with better performance than other well-known IIBs in patients with mCRC treated with first-line therapy. PIV should be prospectively validated to better stratify mCRC patients undergoing first-line therapy.
Collapse
Affiliation(s)
- Giovanni Fucà
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | - Vincenzo Guarini
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | - Carlotta Antoniotti
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy.,Unit of Medical Oncology 2, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy
| | - Federica Morano
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | - Roberto Moretto
- Unit of Medical Oncology 2, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy
| | - Salvatore Corallo
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | - Federica Marmorino
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy.,Unit of Medical Oncology 2, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy
| | - Sara Lonardi
- Unit of Medical Oncology 1, Department of Clinical and Experimental Oncology, Istituto Oncologico Veneto, IRCCS, Padua, Italy
| | - Lorenza Rimassa
- Medical Oncology and Hematology Unit, Humanitas Cancer Center, Humanitas Clinical and Research Center-IRCCS, Rozzano, Italy.,Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy
| | - Andrea Sartore-Bianchi
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan, Italy.,Oncology and Hemato-oncology Department, University of Milan, Milan, Italy
| | - Beatrice Borelli
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy.,Unit of Medical Oncology 2, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy
| | - Marco Tampellini
- Department of Oncology, AOU San Luigi di Orbassano, University of Torino, Orbassano, Italy
| | - Sara Bustreo
- Colorectal Cancer Unit, Medical Oncology Division 1, AOU Città della Salute e della Scienza, Torino, Italy
| | - Matteo Claravezza
- Medical Oncology Unit, Ente Ospedaliero Ospedali Galliera, Genoa, Italy
| | - Alessandra Boccaccino
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy.,Unit of Medical Oncology 2, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy
| | - Roberto Murialdo
- Department of Internal Medicine, University of Genoa and IRCCS AOU San Martino-IST, Genoa, Italy
| | | | - Gianluca Tomasello
- Medical Oncology Unit, Azienda Socio-Sanitaria Territoriale (ASST) Ospedale di Cremona, Cremona, Italy
| | - Fotios Loupakis
- Unit of Medical Oncology 1, Department of Clinical and Experimental Oncology, Istituto Oncologico Veneto, IRCCS, Padua, Italy
| | - Vincenzo Adamo
- Medical Oncology Unit, A.O. Papardo, Messina, Italy.,Department of Human Pathology, University of Messina, Messina, Italy
| | - Giuseppe Tonini
- Oncology Department, Policlinico Campus Bio-Medico di Roma, Rome, Italy
| | | | - Filippo de Braud
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy.,Oncology and Hemato-oncology Department, University of Milan, Milan, Italy
| | - Chiara Cremolini
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy.,Unit of Medical Oncology 2, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy
| | - Filippo Pietrantonio
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy. .,Oncology and Hemato-oncology Department, University of Milan, Milan, Italy.
| |
Collapse
|
62
|
Xu X, Lv J, Guo F, Li J, Jia Y, Jiang D, Wang N, Zhang C, Kong L, Liu Y, Zhang Y, Lv J, Li Z. Gut Microbiome Influences the Efficacy of PD-1 Antibody Immunotherapy on MSS-Type Colorectal Cancer via Metabolic Pathway. Front Microbiol 2020; 11:814. [PMID: 32425919 PMCID: PMC7212380 DOI: 10.3389/fmicb.2020.00814] [Citation(s) in RCA: 103] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Accepted: 04/06/2020] [Indexed: 12/12/2022] Open
Abstract
Colorectal cancer (CRC) appears to be rather refractory to checkpoint blockers except the patient with deficient in mismatch repair (dMMR). Therefore, new advances in the treatment of most mismatch repair proficiency (pMMR) (also known as microsatellite stability, MSS) type of CRC patients are considered to be an important clinical issue associated with programmed death 1 (PD-1) inhibitors. In the present study, we evaluated the effects of gut microbiome of MSS-type CRC tumor-bearing mice treated with different antibiotics on PD-1 antibody immunotherapy response. Our results confirmed that the gut microbiome played a key role in the treatment of CT26 tumor-bearing mice with PD-1 antibody. After PD-1 antibody treatment, the injection of antibiotics counteracted the efficacy of PD-1 antibody in inhibiting tumor growth when compared with the Control group (mice were treated with sterile drinking water). Bacteroides_sp._CAG:927 and Bacteroidales_S24-7 were enriched in Control group. Bacteroides_sp._CAG:927, Prevotella_sp._CAG: 1031 and Bacteroides were enriched in Coli group [mice were treated with colistin (2 mg/ml)], Prevotella_sp._CAG:485 and Akkermansia_muciniphila were enriched in Vanc group [mice were treated with vancomycin alone (0.25 mg/ml)]. The metabolites were enriched in the glycerophospholipid metabolic pathway consistent with the metagenomic prediction pathway in Vanc group, Prevotella_sp._CAG:485 and Akkermansia may maintain the normal efficacy of PD-1 antibody by affecting the metabolism of glycerophospholipid. Changes in gut microbiome leaded to changes in glycerophospholipid metabolism level, which may affect the expression of immune-related cytokines IFN-γ and IL-2 in the tumor microenvironment, resulting in a different therapeutic effect of PD-1 antibody. Our findings show that changes in the gut microbiome affect the glycerophospholipid metabolic pathway, thereby regulating the therapeutic potential of PD-1 antibody in the immunotherapy of MSS-type CRC tumor-bearing mice.
Collapse
Affiliation(s)
- Xinjian Xu
- Second Department of Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Ji Lv
- Second Department of Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
- Department of Surgery, First Hospital of Qinhuangdao, Qinhuangdao, China
| | - Fang Guo
- Department of Pharmacology, Hebei Medical University, Shijiazhuang, China
| | - Jing Li
- Department of Traditional Chinese Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
- College of Combine Traditional Chinese and Western Medicine, Hebei Medical University, Shijiazhuang, China
| | - Yitao Jia
- Third Department of Oncology, Hebei General Hospital, Shijiazhuang, China
| | - Da Jiang
- Department of Oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Na Wang
- Department of Pharmacology, Hebei Medical University, Shijiazhuang, China
| | - Chao Zhang
- Department of Clinical Laboratory, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Lingyu Kong
- College of Combine Traditional Chinese and Western Medicine, Hebei Medical University, Shijiazhuang, China
| | - Yabin Liu
- Second Department of Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Yanni Zhang
- Second Department of Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Jian Lv
- Second Department of Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Zhongxin Li
- Second Department of Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
63
|
Al Obaydi MF, Hamed WM, Al Kury LT, Talib WH. Terfezia boudieri: A Desert Truffle With Anticancer and Immunomodulatory Activities. Front Nutr 2020; 7:38. [PMID: 32322585 PMCID: PMC7156637 DOI: 10.3389/fnut.2020.00038] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Accepted: 03/17/2020] [Indexed: 12/24/2022] Open
Abstract
Desert truffles have high nutritional value and grow wild in the Mediterranean basin and Western Asia. Although, many studies were performed to evaluate truffles nutritious values and phytochemical composition, studies are limited to evaluate their anticancer and/ or immunomodulatory effects. Our study was conducted to evaluate the anticancer and immunomodulatory effects of Terfezia boudieri (desert truffle). Different solvent extracts were prepared from the truffle and MTT assay was used to measure their anticancer activity against cancer cell lines (T47D, MCF-7, MDA-MB231, HCT-116, and Hela). Total phenolic content in each extract was determined by using Folin-Ciocalteu reagent and qualitative phytochemical screening was performed using standard methods. The degree of apoptosis induction (using caspase 3 assay) and vascular endothelial growth factor expression were detected using standard kits. Also, ELISA was used to measure levels of IFN-γ, IL-2, IL-4, and IL-10 secreted by splenocytes after treatment with the extracts. The effect of the extracts on splenocytes proliferation was measured using MTT assay. Macrophage function was evaluated using nitro blue tetrazolium assay and pinocytosis function was evaluated using neutral red method. Terpenoids, phytosterols, and carbohydrates were present in all the solvent extracts, while tannins, alkaloids and flavonoids were detected only in aqueous/methanol and aqueous extracts. The highest total phenolic content was observed in aqueous and aqueous methanol extracts. The growth of cancer cell lines was inhibited by T. boudieri extracts in a dose dependent manner. N-hexane extract was the most potent against most cell lines. Aqueous/methanol extract showed high apoptosis induction and angiogenesis suppression effects. An increase in TH1 cytokines (IFN-γ, IL-2) level and a decrease in TH2 cytokine (IL-4) level were evident after lymphocytes stimulation by aqueous/methanol, n-hexane and ethyl acetate extracts of T. boudieri. Ethyl acetate extract of T. boudieri were the most potent extracts to stimulate lymphocytes proliferation while all other extracts showed moderate stimulation. Aqueous/methanol extract was the most active extract to stimulate phagocytosis. Ethyl acetate extract was the most active extract to stimulate pinocytosis. The use of T. boudieri provides variable health benefits. N-hexane, ethyl acetate, and aqueous/methanol extracts exhibited anticancer activities and are potent stimulators of innate and acquired immunity. Further testing is needed to identify the biologically active compounds and detect them quantitatively using GC-MS analysis.
Collapse
Affiliation(s)
- Maha Farid Al Obaydi
- Department of Clinical Pharmacy and Therapeutic, Applied Science Private University, Amman, Jordan
| | - Wafaa M Hamed
- Pharmacy Department, AlNoor University College, Mosul, Iraq
| | - Lina T Al Kury
- Department of Health Sciences, College of Natural and Health Sciences, Zayed University, Abu Dhabi, United Arab Emirates
| | - Wamidh H Talib
- Department of Clinical Pharmacy and Therapeutic, Applied Science Private University, Amman, Jordan
| |
Collapse
|
64
|
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: 111] [Impact Index Per Article: 22.2] [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
|
65
|
Damilakis E, Mavroudis D, Sfakianaki M, Souglakos J. Immunotherapy in Metastatic Colorectal Cancer: Could the Latest Developments Hold the Key to Improving Patient Survival? Cancers (Basel) 2020; 12:E889. [PMID: 32268531 PMCID: PMC7225960 DOI: 10.3390/cancers12040889] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 04/01/2020] [Accepted: 04/03/2020] [Indexed: 12/19/2022] Open
Abstract
Immunotherapy has considerably increased the number of anticancer agents in many tumor types including metastatic colorectal cancer (mCRC). Anti-PD-1 (programmed death 1) and cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4) immune checkpoint inhibitors (ICI) have been shown to benefit the mCRC patients with mismatch repair deficiency (dMMR) or high microsatellite instability (MSI-H). However, ICI is not effective in mismatch repair proficient (pMMR) colorectal tumors, which constitute a large population of patients. Several clinical trials evaluating the efficacy of immunotherapy combined with chemotherapy, radiation therapy, or other agents are currently ongoing to extend the benefit of immunotherapy to pMMR mCRC cases. In dMMR patients, MSI testing through immunohistochemistry and/or polymerase chain reaction can be used to identify patients that will benefit from immunotherapy. Next-generation sequencing has the ability to detect MSI-H using a low amount of nucleic acids and its application in clinical practice is currently being explored. Preliminary data suggest that radiomics is capable of discriminating MSI from microsatellite stable mCRC and may play a role as an imaging biomarker in the future. Tumor mutational burden, neoantigen burden, tumor-infiltrating lymphocytes, immunoscore, and gastrointestinal microbiome are promising biomarkers that require further investigation and validation.
Collapse
Affiliation(s)
- Emmanouil Damilakis
- Department of Medical Oncology, School of Medicine, University of Crete, 71003 Heraklion, Greece; (D.M.); (J.S.)
| | - Dimitrios Mavroudis
- Department of Medical Oncology, School of Medicine, University of Crete, 71003 Heraklion, Greece; (D.M.); (J.S.)
- Laboratory of Translational Oncology, School of Medicine, University of Crete, 71003 Heraklion, Greece;
| | - Maria Sfakianaki
- Laboratory of Translational Oncology, School of Medicine, University of Crete, 71003 Heraklion, Greece;
| | - John Souglakos
- Department of Medical Oncology, School of Medicine, University of Crete, 71003 Heraklion, Greece; (D.M.); (J.S.)
- Laboratory of Translational Oncology, School of Medicine, University of Crete, 71003 Heraklion, Greece;
| |
Collapse
|
66
|
Markman JL, Porritt RA, Wakita D, Lane ME, Martinon D, Noval Rivas M, Luu M, Posadas EM, Crother TR, Arditi M. Loss of testosterone impairs anti-tumor neutrophil function. Nat Commun 2020; 11:1613. [PMID: 32235862 PMCID: PMC7109066 DOI: 10.1038/s41467-020-15397-4] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Accepted: 03/10/2020] [Indexed: 12/15/2022] Open
Abstract
In men, the incidence of melanoma rises rapidly after age 50, and nearly two thirds of melanoma deaths are male. The immune system is known to play a key role in controlling the growth and spread of malignancies, but whether age- and sex-dependent changes in immune cell function account for this effect remains unknown. Here, we show that in castrated male mice, neutrophil maturation and function are impaired, leading to elevated metastatic burden in two models of melanoma. Replacement of testosterone effectively normalized the tumor burden in castrated male mice. Further, the aberrant neutrophil phenotype was also observed in prostate cancer patients receiving androgen deprivation therapy, highlighting the evolutionary conservation and clinical relevance of the phenotype. Taken together, these results provide a better understanding of the role of androgen signaling in neutrophil function and the impact of this biology on immune control of malignancies. It is known that there are sex differences in the incidence and prognosis of certain cancers, including melanoma. In this study, the authors utilize a melanoma model to reveal that castrated mice have a higher metastatic burden associated with androgen dependent impaired neutrophil function.
Collapse
Affiliation(s)
- Janet L Markman
- Department of Pediatrics, Division of Infectious Diseases and Immunology, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Rebecca A Porritt
- Department of Pediatrics, Division of Infectious Diseases and Immunology, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Daiko Wakita
- Department of Pediatrics, Division of Infectious Diseases and Immunology, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Malcolm E Lane
- Department of Pediatrics, Division of Infectious Diseases and Immunology, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Daisy Martinon
- Department of Pediatrics, Division of Infectious Diseases and Immunology, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Magali Noval Rivas
- Department of Pediatrics, Division of Infectious Diseases and Immunology, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA.,Department of Biomedical Sciences, Infectious and Immunologic Disease Research Center, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA.,Department of Biomedical Science, Research Division of Immunology, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA.,David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA
| | - Michael Luu
- Biostatistics and Bioinformatics Core, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Edwin M Posadas
- Urologic Oncology Program/Uro-Oncology Research Laboratories, Samuel Oschin Comprehensive Center Institute, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA.,Division of Hematology/Oncology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Timothy R Crother
- Department of Pediatrics, Division of Infectious Diseases and Immunology, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA.,Department of Biomedical Sciences, Infectious and Immunologic Disease Research Center, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA.,Department of Biomedical Science, Research Division of Immunology, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA.,David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA
| | - Moshe Arditi
- Department of Pediatrics, Division of Infectious Diseases and Immunology, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA. .,Department of Biomedical Sciences, Infectious and Immunologic Disease Research Center, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA. .,Department of Biomedical Science, Research Division of Immunology, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA. .,David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA.
| |
Collapse
|
67
|
Trac NT, Chung EJ. Peptide-based targeting of immunosuppressive cells in cancer. Bioact Mater 2020; 5:92-101. [PMID: 31956738 PMCID: PMC6962647 DOI: 10.1016/j.bioactmat.2020.01.006] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 01/06/2020] [Accepted: 01/09/2020] [Indexed: 02/07/2023] Open
Abstract
Cancer progression is marked by the infiltration of immunosuppressive cells, such as tumor-associated macrophages (TAMs), regulatory T lymphocytes (Tregs), and myeloid-derived suppressor cells (MDSCs). These cells play a key role in abrogating the cytotoxic T lymphocyte-mediated (CTL) immune response, allowing tumor growth to proceed unabated. Furthermore, targeting these immunosuppressive cells through the use of peptides and peptide-based nanomedicine has shown promising results. Here we review the origins and functions of immunosuppressive cells in cancer progression, peptide-based systems used in their targeting, and explore future avenues of research regarding cancer immunotherapy. The success of these studies demonstrates the importance of the tumor immune microenvironment in the propagation of cancer and the potential of peptide-based nanomaterials as immunomodulatory agents.
Collapse
Affiliation(s)
- Noah T. Trac
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, USA
| | - Eun Ji Chung
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, USA
- Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, CA, USA
- Division of Nephrology and Hypertension, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Department of Vascular Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, University of Southern California, Los Angeles, CA, USA
- Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
68
|
Zhang Y, Diao N, Lee CK, Chu HW, Bai L, Li L. Neutrophils Deficient in Innate Suppressor IRAK-M Enhances Anti-tumor Immune Responses. Mol Ther 2020; 28:89-99. [PMID: 31607540 PMCID: PMC6953792 DOI: 10.1016/j.ymthe.2019.09.019] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2019] [Revised: 09/06/2019] [Accepted: 09/12/2019] [Indexed: 02/07/2023] Open
Abstract
Tumor-associated immune-suppressive neutrophils are prevalent in various cancers, including colorectal cancer. However, mechanisms of immune-suppressive neutrophils are not well understood. We report that a key innate suppressor, IRAK-M (interleukin-1 receptor-associated kinase M), is critically involved in the establishment of immune-suppressive neutrophils. In contrast to the wild-type (WT) neutrophils exhibiting immune-suppressive signatures of CD11bhighPD-L1highCD80low, IRAK-M-deficient neutrophils are rewired with reduced levels of inhibitory molecules PD-L1 and CD11b, as well as enhanced expression of stimulatory molecules CD80 and CD40. The reprogramming of IRAK-M-deficient neutrophils is mediated by reduced activation of STAT1/3 and enhanced activation of STAT5. As a consequence, IRAK-M-deficient neutrophils demonstrate enhanced capability to promote, instead of suppress, the proliferation and activation of effector T cells both in vitro and in vivo. Functionally, we observed that the transfusion of IRAK-M-/- neutrophils can potently render an enhanced anti-tumor immune response in the murine inflammation-induced colorectal cancer model. Collectively, our study defines IRAK-M as an innate suppressor for neutrophil function and reveals IRAK-M as a promising target for rewiring neutrophils in anti-cancer immunotherapy.
Collapse
Affiliation(s)
- Yao Zhang
- Department of Biological Sciences, Virginia Tech, Blacksburg, VA 24061, USA.
| | - Na Diao
- Department of Biological Sciences, Virginia Tech, Blacksburg, VA 24061, USA; Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, PR China
| | - Christina K Lee
- Department of Biological Sciences, Virginia Tech, Blacksburg, VA 24061, USA
| | - Hong Wei Chu
- Department of Medicine, National Jewish Health, Denver, CO 80206, USA
| | - Lan Bai
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, PR China.
| | - Liwu Li
- Department of Biological Sciences, Virginia Tech, Blacksburg, VA 24061, USA.
| |
Collapse
|
69
|
Sureban SM, Berahovich R, Zhou H, Xu S, Wu L, Ding K, May R, Qu D, Bannerman-Menson E, Golubovskaya V, Houchen CW. DCLK1 Monoclonal Antibody-Based CAR-T Cells as a Novel Treatment Strategy against Human Colorectal Cancers. Cancers (Basel) 2019; 12:cancers12010054. [PMID: 31878090 PMCID: PMC7016951 DOI: 10.3390/cancers12010054] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 12/04/2019] [Accepted: 12/16/2019] [Indexed: 02/06/2023] Open
Abstract
CAR-T (chimeric antigen receptor T cells) immunotherapy is effective in many hematological cancers; however, efficacy in solid tumors is disappointing. Doublecortin-like kinase 1 (DCLK1) labels tumor stem cells (TSCs) in genetic mouse models of colorectal cancer (CRC). Here, we describe a novel CAR-T targeting DCLK1 (CBT-511; with our proprietary DCLK1 single-chain antibody variable fragment) as a treatment strategy to eradicate CRC TSCs. The cell surface expression of DCLK1 and cytotoxicity of CBT-511 were assessed in CRC cells (HT29, HCT116, and LoVo). LoVo-derived tumor xenografts in NOD Scid gamma (NSGTM)mice were treated with CBT-511 or mock CAR-T cells. Adherent CRC cells express surface DCLK1 (two-dimensional, 2D). A 4.5-fold increase in surface DCLK1 was observed when HT29 cells were grown as spheroids (three-dimensional, 3D). CBT-511 induced cytotoxicity (2D; p < 0.0001), and increased Interferon gamma (IFN-γ) release in CRC cells (2D) compared to mock CAR-T (p < 0.0001). Moreover, an even greater increase in IFN-γ release was observed when cells were grown in 3D. CBT-511 reduced tumor growth by approximately 50 percent compared to mock CAR-T. These data suggest that CRC cells with increased clonogenic capacity express increased surface DCLK1. A DCLK1-targeted CAR-T can induce cytotoxicity in vitro and inhibit xenograft growth in vivo.
Collapse
Affiliation(s)
- Sripathi M. Sureban
- COARE Holdings Inc., Oklahoma, OK 73104, USA; (R.M.); (D.Q.); (E.B.-M.)
- Department of Internal Medicine, Digestive Diseases and Nutrition Section, The University of Oklahoma Health Science Center, Oklahoma, OK 73014, USA;
- Correspondence: (S.M.S.); (C.W.H.); Tel.: +1-405-271-5428 (S.M.S. & C.W.H.)
| | - Robert Berahovich
- ProMab Biotechnologies Inc., Richmond, CA 94806, USA; (R.B.); (H.Z.); (S.X.); (L.W.); (V.G.)
| | - Hua Zhou
- ProMab Biotechnologies Inc., Richmond, CA 94806, USA; (R.B.); (H.Z.); (S.X.); (L.W.); (V.G.)
| | - Shirley Xu
- ProMab Biotechnologies Inc., Richmond, CA 94806, USA; (R.B.); (H.Z.); (S.X.); (L.W.); (V.G.)
| | - Lijun Wu
- ProMab Biotechnologies Inc., Richmond, CA 94806, USA; (R.B.); (H.Z.); (S.X.); (L.W.); (V.G.)
| | - Kai Ding
- Department of Internal Medicine, Digestive Diseases and Nutrition Section, The University of Oklahoma Health Science Center, Oklahoma, OK 73014, USA;
| | - Randal May
- COARE Holdings Inc., Oklahoma, OK 73104, USA; (R.M.); (D.Q.); (E.B.-M.)
- Department of Internal Medicine, Digestive Diseases and Nutrition Section, The University of Oklahoma Health Science Center, Oklahoma, OK 73014, USA;
| | - Dongfeng Qu
- COARE Holdings Inc., Oklahoma, OK 73104, USA; (R.M.); (D.Q.); (E.B.-M.)
- Department of Internal Medicine, Digestive Diseases and Nutrition Section, The University of Oklahoma Health Science Center, Oklahoma, OK 73014, USA;
| | | | - Vita Golubovskaya
- ProMab Biotechnologies Inc., Richmond, CA 94806, USA; (R.B.); (H.Z.); (S.X.); (L.W.); (V.G.)
| | - Courtney W. Houchen
- COARE Holdings Inc., Oklahoma, OK 73104, USA; (R.M.); (D.Q.); (E.B.-M.)
- Department of Internal Medicine, Digestive Diseases and Nutrition Section, The University of Oklahoma Health Science Center, Oklahoma, OK 73014, USA;
- Veterans Affairs Medical Center, Oklahoma, OK 73104, USA
- Correspondence: (S.M.S.); (C.W.H.); Tel.: +1-405-271-5428 (S.M.S. & C.W.H.)
| |
Collapse
|
70
|
Baritaki S, de Bree E, Chatzaki E, Pothoulakis C. Chronic Stress, Inflammation, and Colon Cancer: A CRH System-Driven Molecular Crosstalk. J Clin Med 2019; 8:E1669. [PMID: 31614860 PMCID: PMC6833069 DOI: 10.3390/jcm8101669] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 10/08/2019] [Accepted: 10/11/2019] [Indexed: 12/12/2022] Open
Abstract
Chronic stress is thought to be involved in the occurrence and progression of multiple diseases, via mechanisms that still remain largely unknown. Interestingly, key regulators of the stress response, such as members of the corticotropin-releasing-hormone (CRH) family of neuropeptides and receptors, are now known to be implicated in the regulation of chronic inflammation, one of the predisposing factors for oncogenesis and disease progression. However, an interrelationship between stress, inflammation, and malignancy, at least at the molecular level, still remains unclear. Here, we attempt to summarize the current knowledge that supports the inseparable link between chronic stress, inflammation, and colorectal cancer (CRC), by modulation of a cascade of molecular signaling pathways, which are under the regulation of CRH-family members expressed in the brain and periphery. The understanding of the molecular basis of the link among these processes may provide a step forward towards personalized medicine in terms of CRC diagnosis, prognosis and therapeutic targeting.
Collapse
Affiliation(s)
- Stavroula Baritaki
- Division of Surgery, School of Medicine, University of Crete, Heraklion, 71500 Crete, Greece.
| | - Eelco de Bree
- Division of Surgery, School of Medicine, University of Crete, Heraklion, 71500 Crete, Greece.
| | - Ekaterini Chatzaki
- Laboratory of Pharmacology, Medical School, Democritus University of Thrace, 68100 Alexandroupolis, Greece.
| | - Charalabos Pothoulakis
- IBD Center, Division of Digestive Diseases, David Geffen School of Medicine at UCLA, Los Angeles, CA 10833, USA.
| |
Collapse
|
71
|
Kim SM, Kim EM, Ji KY, Lee HY, Yee SM, Woo SM, Yi JW, Yun CH, Choi H, Kang HS. TREM2 Acts as a Tumor Suppressor in Colorectal Carcinoma through Wnt1/ β-catenin and Erk Signaling. Cancers (Basel) 2019; 11:cancers11091315. [PMID: 31489935 PMCID: PMC6770495 DOI: 10.3390/cancers11091315] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 08/30/2019] [Accepted: 09/05/2019] [Indexed: 01/22/2023] Open
Abstract
TREM2 (triggering receptor expressed on myeloid cells) is involved in the development of malignancies. However, the function of TREM2 in colorectal cancer has not been clearly elucidated. Here, we investigated TREM2 function for the first time in colorectal epithelial cancer cells and demonstrated that TREM2 is a novel tumor suppressor in colorectal carcinoma. Blockade of TREM2 significantly promoted the proliferation of HT29 colorectal carcinoma cells by regulating cell cycle-related factors, such as p53 phosphorylation and p21 and cyclin D1 protein levels. HT29 cell migration was also increased by TREM2 inhibition via MMP9 (matrix metalloproteinase 9) expression upregulation. Furthermore, we found that the tumor suppressor effects of TREM2 were associated with Wnt/β-catenin and extracellular signal-regulated kinase (ERK) signaling. Importantly, the effect of TREM2 in the suppression of tumor development was demonstrated by in vivo and in vitro assays, as well as in human colon cancer patient tissue arrays. Overall, our results identify TREM2 as a potential prognostic biomarker and therapeutic target for colorectal cancer.
Collapse
Affiliation(s)
- Su-Man Kim
- School of Biological Sciences and Technology, Chonnam National University, Gwangju 500-757, Korea.
| | - Eun-Mi Kim
- Korea Institute of Toxicology, Daejeon, 34114, Korea.
| | - Kon-Young Ji
- Herbal Medicine Research Division, Korea Institute of Oriental Medicine, Daejeon 34054, Korea.
| | - Hwa-Youn Lee
- Medical Device Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu 701-310, Korea.
| | - Su-Min Yee
- School of Biological Sciences and Technology, Chonnam National University, Gwangju 500-757, Korea.
| | - Su-Min Woo
- School of Biological Sciences and Technology, Chonnam National University, Gwangju 500-757, Korea.
| | - Ja-Woon Yi
- School of Biological Sciences and Technology, Chonnam National University, Gwangju 500-757, Korea.
| | - Chul-Ho Yun
- School of Biological Sciences and Technology, Chonnam National University, Gwangju 500-757, Korea.
| | - Harim Choi
- Department of Nursing, Nambu University, Gwangju 506-706, Korea.
| | - Hyung-Sik Kang
- School of Biological Sciences and Technology, Chonnam National University, Gwangju 500-757, Korea.
| |
Collapse
|
72
|
Aldarouish M, Su X, Qiao J, Gao C, Chen Y, Dai A, Zhang T, Shu Y, Wang C. Immunomodulatory effects of chemotherapy on blood lymphocytes and survival of patients with advanced non-small cell lung cancer. Int J Immunopathol Pharmacol 2019; 33:2058738419839592. [PMID: 30968711 PMCID: PMC6458672 DOI: 10.1177/2058738419839592] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
A better understanding of the immune profile of non-small cell lung cancer (NSCLC) and the immunomodulatory impact of chemotherapy is essential to develop current therapeutic approaches. Herein, we collected peripheral blood from 20 healthy donors and 50 patients with advanced NSCLC, before and after chemotherapy, followed by phenotypic analysis of lymphocyte subsets and assessment of the correlation between their post-chemotherapy levels and progression-free survival (PFS). Results showed that, before chemotherapy, the levels of CD8+ lymphocytes, PD-1+CD4+, Th2, and Th17 cells were elevated in patients’ peripheral blood, in contrast to natural killer (NK) cells and Th1 cells. Besides, there was no remarkable difference in the frequency of PD-1+CD8+ cells between patients and healthy controls. After chemotherapy, the levels of CD8+ lymphocytes, NK, Th2, Th17, and Treg were declined, in contrast to the level of Th1 cells which was markedly increased. Importantly, chemotherapy had no impact on the frequencies of PD-1+CD8+ and PD-1+CD4+ cells. PFS was significantly better in patients with low percentage of PD-1+CD4+ T cells than those with high percentage. Patients with high content of Th1 cells showed longer PFS than those with low content. The low percentages of Th17 and Treg cells were correlated with longer PFS, even though the difference did not reach statistical significance. In conclusion, the imbalance of lymphocyte subsets is a hallmark of NSCLC. Furthermore, the high level of PD-1+CD4+ cells plays a crucial role in the progression of NSCLC and could be used as a prognostic marker; and the high level of Th1 could predict better clinical outcomes of chemotherapy.
Collapse
Affiliation(s)
- Mohanad Aldarouish
- 1 Department of Oncology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, People's Republic of China
| | - Xiangyu Su
- 1 Department of Oncology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, People's Republic of China
| | - Jianbing Qiao
- 2 Department of Respiratory, Nanjing Chest Hospital, Nanjing, People's Republic of China
| | - Chanchan Gao
- 1 Department of Oncology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, People's Republic of China
| | - Yan Chen
- 1 Department of Oncology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, People's Republic of China
| | - Anwei Dai
- 3 Department of Oncology, Kunshan Traditional Chinese Medicine Hospital, Kunshan, People's Republic of China
| | - Tianyu Zhang
- 4 Department of Microbiology and Immunology, School of Medicine, Southeast University, Nanjing, People's Republic of China
| | - Yongqian Shu
- 5 The First Affiliated Hospital of Nanjing Medical University (Jiangsu Provincial People's Hospital), Nanjing, People's Republic of China
| | - Cailian Wang
- 1 Department of Oncology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, People's Republic of China
| |
Collapse
|
73
|
Guo G, Wang Y, Zhou Y, Quan Q, Zhang Y, Wang H, Zhang B, Xia L. Immune cell concentrations among the primary tumor microenvironment in colorectal cancer patients predicted by clinicopathologic characteristics and blood indexes. J Immunother Cancer 2019; 7:179. [PMID: 31300050 PMCID: PMC6625045 DOI: 10.1186/s40425-019-0656-3] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Accepted: 06/27/2019] [Indexed: 12/14/2022] Open
Abstract
Background Immune cells play a key role in cancer progression and treatment. It is unclear whether the clinicopathologic characteristics and blood indexes of colorectal cancer (CRC) patients could predict immune cell concentrations in the tumor microenvironment. Methods CRC patients with detailed data and tumor tissue who visited Sun Yat-sen University Cancer Center between April 1, 2004, and September 1, 2017, were enrolled. The densities of CD3+ and CD8+ T cells examined by immunohistochemistry in both the core of the tumor (CT) and the invasive margin (IM) were summed as the Immunoscore. The relationships between the Immunoscore and clinicopathologic characteristics and blood indexes, including tumor biomarkers (carcinoembryonic antigen (CEA) and carbohydrate antigen 19–9 (CA 19–9)), inflammatory markers (lactate dehydrogenase (LDH), C-reactive protein (CRP), albumin (ALB), neutrophils, lymphocytes, monocytes, platelets, NLR (neutrophil-to-lymphocyte ratio), PLR (platelet-to-lymphocyte ratio) and LMR (lymphocyte-to-monocyte ratio)) and lipid metabolism markers (cholesterol (CHO), triglyceride (TG), high-density lipoprotein (HDL), low-density lipoprotein (LDL), apolipoprotein A1 (ApoA1), and apolipoprotein B (ApoB)), were analyzed using SPSS. Results Older patients had lower CD3+ and CD8+ T cell expression in the IM and a lower Immunoscore than did younger patients. CD8+ T cell expression in the IM and the Immunoscore were lower in right-side tumors than in left-sided tumors. High CD8+ T cell expression in the CT was found in the T4 stage group. The higher the CEA level in the blood, the fewer CD8+ T cells were in the CT. Either fewer monocytes or a higher LMR in the blood, the larger number of CD3+ T cells in the CT. The more ApoA1 was in the blood, the more CD3+ T cells were in both the CT and the IM. Conclusion Age, T stage, tumor location, CEA, monocytes, LMR and ApoA1 could reflect immune cells infiltrating the tumor microenvironment of CRC.
Collapse
Affiliation(s)
- Guifang Guo
- VIP Department, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, People's Republic of China. .,State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, People's Republic of China. .,Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, People's Republic of China.
| | - Yixing Wang
- VIP Department, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, People's Republic of China.,State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, People's Republic of China.,Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, People's Republic of China.,Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, People's Republic of China
| | - Yixin Zhou
- VIP Department, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, People's Republic of China.,State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, People's Republic of China.,Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, People's Republic of China.,Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, People's Republic of China
| | - Qi Quan
- VIP Department, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, People's Republic of China.,State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, People's Republic of China.,Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, People's Republic of China.,Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, People's Republic of China
| | - Yijun Zhang
- VIP Department, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, People's Republic of China.,State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, People's Republic of China.,Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, People's Republic of China.,Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, People's Republic of China
| | - Haohua Wang
- VIP Department, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, People's Republic of China.,State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, People's Republic of China.,Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, People's Republic of China.,Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, People's Republic of China
| | - Bei Zhang
- VIP Department, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, People's Republic of China. .,State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, People's Republic of China. .,Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, People's Republic of China. .,Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, People's Republic of China.
| | - Liangping Xia
- VIP Department, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, People's Republic of China. .,State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, People's Republic of China. .,Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, People's Republic of China. .,Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, People's Republic of China.
| |
Collapse
|
74
|
Basile D, Gerratana L, Buonadonna A, Garattini SK, Perin T, Grassilli E, Miolo G, Cerrito MG, Belluco C, Bertola G, De Paoli A, Cannizzaro R, Lavitrano M, Puglisi F, Canzonieri V. Role of Bruton's Tyrosine Kinase in Stage III Colorectal Cancer. Cancers (Basel) 2019; 11:E880. [PMID: 31238520 PMCID: PMC6627163 DOI: 10.3390/cancers11060880] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Revised: 06/16/2019] [Accepted: 06/19/2019] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Bruton's tyrosine kinase (BTK) is involved in the immune response and its deficiency impairs B cell maturation. We evaluated the expression of a novel BTK isoform, p65BTK, in colorectal cancer (CRC), to identify its impact on survival. MATERIALS AND METHODS This retrospective study evaluated 87 consecutive stage III CRC patients treated at the National Cancer Institute of Aviano (1999-2017). Multiple specimens were collected and analyzed for staining intensity and percentage of tumor cells positive for p65BTK. Prognostic impact was tested by univariate Cox regression analysis. RESULTS After a median follow-up of 82.59 months, median disease-free survival (DFS) and overall survival (OS) were 11.67 months and 31.33 months, respectively. Interestingly, 10% of patients did not express p65BTK. For the immunohistochemistry IHC intensity 1, the best cutoff point was 1% of p65BTK positivity; for IHC intensity 2, it was 50%; and for IHC intensity 3, it was 80%. Through univariate analysis, patients with highly expressed p65BTK (IHC intensity 3 and ≥80%) were shown to have the worst prognosis in terms of DFS (HR: 6.23; p = 0.005; 95% C.I. 1.75-22.79) and OS (HR: 2.54; p = 0.025; 95% C.I. 1.12-5.76). CONCLUSIONS p65BTK is frequently expressed in CRC and, if highly expressed, is an unfavourable prognostic factor. However, further confirmation is needed and its potential targeting needs to be studied.
Collapse
Affiliation(s)
- Debora Basile
- Department of Medicine (DAME), University of Udine, 33100 Udine (UD), Italy.
- Department of Medical Oncology, IRCCS CRO National Cancer Institute, 33081 Aviano (PN), Italy.
| | - Lorenzo Gerratana
- Department of Medicine (DAME), University of Udine, 33100 Udine (UD), Italy.
- Department of Medical Oncology, IRCCS CRO National Cancer Institute, 33081 Aviano (PN), Italy.
| | - Angela Buonadonna
- Department of Medical Oncology, IRCCS CRO National Cancer Institute, 33081 Aviano (PN), Italy.
| | - Silvio Ken Garattini
- Department of Medicine (DAME), University of Udine, 33100 Udine (UD), Italy.
- Department of Medical Oncology, IRCCS CRO National Cancer Institute, 33081 Aviano (PN), Italy.
| | - Tiziana Perin
- Pathology Unit, IRCCS CRO National Cancer Institute, 33081 Aviano (PN), Italy.
- CRO Biobank IRCCS CRO National Cancer Institute, 33081 Aviano (PN), Italy.
| | - Emanuela Grassilli
- Department of Medicine and Surgery, University of Milano-Bicocca, 33081 Milan, Italy.
| | - Gianmaria Miolo
- Department of Medical Oncology, IRCCS CRO National Cancer Institute, 33081 Aviano (PN), Italy.
| | - Maria Grazia Cerrito
- Department of Medicine and Surgery, University of Milano-Bicocca, 33081 Milan, Italy.
| | - Claudio Belluco
- Surgical Oncology, IRCCS CRO National Cancer Institute, 33081 Aviano (PN), Italy.
| | - Giulio Bertola
- Surgical Oncology, IRCCS CRO National Cancer Institute, 33081 Aviano (PN), Italy.
| | - Antonino De Paoli
- Radiation Oncology, IRCCS CRO National Cancer Institute, 33081 Aviano (PN), Italy.
| | - Renato Cannizzaro
- Division of Oncological Gastroenterology, IRCCS CRO National Cancer Institute, 33081 Aviano (PN), Italy.
| | - Marialuisa Lavitrano
- Department of Medicine and Surgery, University of Milano-Bicocca, 33081 Milan, Italy.
| | - Fabio Puglisi
- Department of Medicine (DAME), University of Udine, 33100 Udine (UD), Italy.
- Department of Medical Oncology, IRCCS CRO National Cancer Institute, 33081 Aviano (PN), Italy.
| | - Vincenzo Canzonieri
- Pathology Unit, IRCCS CRO National Cancer Institute, 33081 Aviano (PN), Italy.
- CRO Biobank IRCCS CRO National Cancer Institute, 33081 Aviano (PN), Italy.
- Department of Medical, Surgical and Health Science, University of Trieste School of Medicine, 34100 Trieste (TS), Italy.
| |
Collapse
|
75
|
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: 6.7] [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
|
76
|
Strasser K, Birnleitner H, Beer A, Pils D, Gerner MC, Schmetterer KG, Bachleitner-Hofmann T, Stift A, Bergmann M, Oehler R. Immunological differences between colorectal cancer and normal mucosa uncover a prognostically relevant immune cell profile. Oncoimmunology 2018; 8:e1537693. [PMID: 30713795 PMCID: PMC6343804 DOI: 10.1080/2162402x.2018.1537693] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2018] [Revised: 10/05/2018] [Accepted: 10/10/2018] [Indexed: 12/22/2022] Open
Abstract
T cells in colorectal cancer (CRC) are associated with improved survival. However, checkpoint immunotherapies antagonizing the suppression of these cells are ineffective in the great majority of patients. To better understand the immune cell regulation in CRC, we compared tumor-associated T lymphocytes and macrophages to the immune cell infiltrate of normal mucosa. Human colorectal tumor specimen and tumor-distant normal mucosa tissues of the same patients were collected. Phenotypes and functionality of tissue-derived T cells and macrophages were characterized using immunohistochemistry, RNA in situ hybridization, and multiparameter flow cytometry. CRC contained significantly higher numbers of potentially immunosuppressive CD39 and Helios-expressing regulatory T cells in comparison to normal mucosa. Surprisingly, we found a concomitant increase of pro-inflammatory IFNγ -producing T cells. PD-L1+ stromal cells were decreased in the tumor tissue. Macrophages in the tumor compared to tumor-distant normal tissue appear to have an altered phenotype, identified by HLA-DR, CD14, CX3CR1, and CD64, and tolerogenic CD206+ macrophages are quantitatively reduced. The prognostic effect of these observed differences between distant mucosa and tumor tissue on the overall survival was examined using gene expression data of 298 CRC patients. The combined gene expression of increased FOXP3, IFNγ, CD14, and decreased CD206 correlated with a poor prognosis in CRC patients. These data reveal that the CRC microenvironment promotes the coexistence of seemingly antagonistic suppressive and pro-inflammatory immune responses and might provide an explanation why a blockade of the PD1/PD-L1 axis is ineffective in CRC. This should be taken into account when designing novel treatment strategies.
Collapse
Affiliation(s)
- Katharina Strasser
- Department of Surgery and Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria.,CBmed GmbH - Center for Biomarker Research in Medicine, Graz, Austria
| | - Hanna Birnleitner
- Department of Surgery and Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Andrea Beer
- Department of Pathology, Medical University of Vienna, Vienna, Austria
| | - Dietmar Pils
- Department of Surgery and Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria.,Center for Medical Statistics, Informatics and Intelligent Systems, Section for Clinical Biometrics, Medical University of Vienna, Vienna, Austria
| | - Marlene C Gerner
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Klaus G Schmetterer
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | | | - Anton Stift
- Department of Surgery and Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Michael Bergmann
- Department of Surgery and Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Rudolf Oehler
- Department of Surgery and Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
77
|
Clifford R, Govindarajah N, Parsons JL, Gollins S, West NP, Vimalachandran D. Systematic review of treatment intensification using novel agents for chemoradiotherapy in rectal cancer. Br J Surg 2018; 105:1553-1572. [PMID: 30311641 PMCID: PMC6282533 DOI: 10.1002/bjs.10993] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2018] [Revised: 08/07/2018] [Accepted: 08/07/2018] [Indexed: 12/23/2022]
Abstract
BACKGROUND With the well established shift to neoadjuvant treatment for locally advanced rectal cancer, there is increasing focus on the use of radiosensitizers to improve the efficacy and tolerability of radiotherapy. There currently exist few randomized data exploring novel radiosensitizers to improve response and it is unclear what the clinical endpoints of such trials should be. METHODS A qualitative systematic review was performed according to the PRISMA guidelines using preset search criteria across the PubMed, Cochrane and Scopus databases from 1990 to 2017. Additional results were generated from the reference lists of included papers. RESULTS A total of 123 papers were identified, of which 37 were included; a further 60 articles were obtained from additional referencing to give a total of 97 articles. Neoadjuvant radiosensitization for locally advanced rectal cancer using fluoropyrimidine-based chemotherapy remains the standard of treatment. The oral derivative capecitabine has practical advantages over 5-fluorouracil, with equal efficacy, but the addition of a second chemotherapeutic agent has yet to show a consistent significant efficacy benefit in randomized clinical assessment. Preclinical and early-phase trials are progressing with promising novel agents, such as small molecular inhibitors and nanoparticles. CONCLUSION Despite extensive research and promising preclinical studies, a definite further agent in addition to fluoropyrimidines that consistently improves response rate has yet to be found.
Collapse
Affiliation(s)
- R. Clifford
- Institute of Cancer Medicine, University of LiverpoolLiverpoolUK
| | - N. Govindarajah
- Institute of Cancer Medicine, University of LiverpoolLiverpoolUK
| | - J. L. Parsons
- Institute of Cancer Medicine, University of LiverpoolLiverpoolUK
| | - S. Gollins
- North Wales Cancer Treatment Centre, Glan Clwyd HospitalBodelwyddanUK
| | - N. P. West
- Leeds Institute of Cancer and Pathology, University of LeedsLeedsUK
| | - D. Vimalachandran
- Institute of Cancer Medicine, University of LiverpoolLiverpoolUK
- Department of Colorectal SurgeryCountess of Chester NHS Foundation TrustChesterUK
| |
Collapse
|
78
|
Cantero-Cid R, Casas-Martin J, Hernández-Jiménez E, Cubillos-Zapata C, Varela-Serrano A, Avendaño-Ortiz J, Casarrubios M, Montalbán-Hernández K, Villacañas-Gil I, Guerra-Pastrián L, Peinado B, Marcano C, Aguirre LA, López-Collazo E. PD-L1/PD-1 crosstalk in colorectal cancer: are we targeting the right cells? BMC Cancer 2018; 18:945. [PMID: 30285662 PMCID: PMC6171318 DOI: 10.1186/s12885-018-4853-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Accepted: 09/24/2018] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND The analysis of tumour-infiltrating immune cells within patients' tumour samples in colorectal cancer (CRC) has become an independent predictor of patient survival. The tumour microenvironment and the immune checkpoints, such as PD-L1/PD-1, are relevant to the prognoses and also appear to be relevant for further CRC therapies. METHODS We analysed the presence and features of the infiltrated monocyte/macrophage and lymphocyte populations in both tumour and peritumour samples from patients with CRC (n = 15). RESULTS We detected a large number of CD14+ monocytes/macrophages with an alternative phenotype (CD64+CD163+) and CD4+ lymphocytes that infiltrated the tumour, but not the peritumour area. The monocytes/macrophages expressed PD-L1, whereas the lymphocytes were PD-1+; however, we did not find high PD-L1 levels in the tumour cells. Coculture of circulating naïve human monocytes/macrophages and lymphocytes with tumour cells from patients with proficient mismatch repair CRC induced both an alternative phenotype with higher expression of PD-L1 in CD14+ cells and the T-cell exhaustion phenomenon. The addition of an α-PD-1 antibody restored lymphocyte proliferation. CONCLUSION These results emphasise the interesting nature of immune checkpoint shifting therapies, which have potential clinical applications in the context of colorectal cancer.
Collapse
Affiliation(s)
- Ramón Cantero-Cid
- The Innate Immune Response Group, IdiPAZ, La Paz University Hospital, Madrid, Spain
- Tumour Immunology Laboratory, IdiPAZ, Madrid, Spain
- Surgery Department, La Paz University Hospital, Madrid, Spain
| | - José Casas-Martin
- The Innate Immune Response Group, IdiPAZ, La Paz University Hospital, Madrid, Spain
- Tumour Immunology Laboratory, IdiPAZ, Madrid, Spain
| | - Enrique Hernández-Jiménez
- The Innate Immune Response Group, IdiPAZ, La Paz University Hospital, Madrid, Spain
- Tumour Immunology Laboratory, IdiPAZ, Madrid, Spain
- Centre for Biomedical Research Network, CIBEres, Madrid, Spain
| | - Carolina Cubillos-Zapata
- The Innate Immune Response Group, IdiPAZ, La Paz University Hospital, Madrid, Spain
- Tumour Immunology Laboratory, IdiPAZ, Madrid, Spain
- Centre for Biomedical Research Network, CIBEres, Madrid, Spain
| | - Aníbal Varela-Serrano
- The Innate Immune Response Group, IdiPAZ, La Paz University Hospital, Madrid, Spain
- Tumour Immunology Laboratory, IdiPAZ, Madrid, Spain
| | - José Avendaño-Ortiz
- The Innate Immune Response Group, IdiPAZ, La Paz University Hospital, Madrid, Spain
- Tumour Immunology Laboratory, IdiPAZ, Madrid, Spain
| | - Marta Casarrubios
- The Innate Immune Response Group, IdiPAZ, La Paz University Hospital, Madrid, Spain
- Tumour Immunology Laboratory, IdiPAZ, Madrid, Spain
| | - Karla Montalbán-Hernández
- The Innate Immune Response Group, IdiPAZ, La Paz University Hospital, Madrid, Spain
- Tumour Immunology Laboratory, IdiPAZ, Madrid, Spain
| | - Ignacio Villacañas-Gil
- The Innate Immune Response Group, IdiPAZ, La Paz University Hospital, Madrid, Spain
- Tumour Immunology Laboratory, IdiPAZ, Madrid, Spain
| | | | - Begoña Peinado
- Surgery Department, La Paz University Hospital, Madrid, Spain
| | | | - Luis A Aguirre
- The Innate Immune Response Group, IdiPAZ, La Paz University Hospital, Madrid, Spain
- Tumour Immunology Laboratory, IdiPAZ, Madrid, Spain
| | - Eduardo López-Collazo
- The Innate Immune Response Group, IdiPAZ, La Paz University Hospital, Madrid, Spain
- Tumour Immunology Laboratory, IdiPAZ, Madrid, Spain
- Centre for Biomedical Research Network, CIBEres, Madrid, Spain
| |
Collapse
|
79
|
Yu W, Guo Y. Prognostic significance of programmed death ligand-1 immunohistochemical expression in esophageal cancer: A meta-analysis of the literature. Medicine (Baltimore) 2018; 97:e11614. [PMID: 30045299 PMCID: PMC6078683 DOI: 10.1097/md.0000000000011614] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND It is thought that expression of programmed death ligand-1 (PD-L1) in esophageal cancer (EC) might compromise patient survival. However, the association between PD-L1 expression and survival of patients with EC remains controversial. METHODS A meta-analysis combining eligible published studies was performed to evaluate the effect of PD-L1 expression in tumor cells detected by immunohistochemistry (IHC) on overall survival (OS) and disease-free survival (DFS) in patients with EC, using pooled hazard ratio (HR) with its 95% confidence interval (CI). RESULTS The pooled HR for 19 eligible studies (18 publications, n = 3306) suggested that PD-L1 overexpression had an unfavorable impact on OS (HR = 1.42, 95% CI: 1.09-1.86). No significant effect of PD-L1 overexpression on DFS was observed, and the combined HR was 1.08 (95% CI: 0.76-1.53) for 12 eligible studies (11 publications, n = 2260). CONCLUSION PD-L1 expression in tumor cells detected by IHC was associated with worse OS in EC. However, the prognostic value of PD-L1 expression in tumor cells on OS in EC still needs further large prospective trials to be clarified.
Collapse
Affiliation(s)
- Weiwei Yu
- Department of Radiation Oncology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital
| | - Yanmei Guo
- Department of Gastroenterology, The Tenth People's Hospital of Tongji University, Shanghai, China
| |
Collapse
|
80
|
Abstract
Tissues contain multiple different cell types and can be considered to be heterocellular systems. Signaling between different cells allows tissues to achieve phenotypes that no cell type can achieve in isolation. Such emergent tissue-level phenotypes can be said to 'supervene upon' heterocellular signaling. It is proposed here that cancer is also an emergent phenotype that supervenes upon heterocellular signaling. Using colorectal cancer (CRC) as an example, I review how heterotypic cells differentially communicate to support emergent malignancy. Studying tumors as integrated heterocellular systems - rather than as solitary expansions of mutated cells - may reveal novel ways to treat cancer.
Collapse
|
81
|
Gang W, Wang JJ, Guan R, Yan S, Shi F, Zhang JY, Li ZM, Gao J, Fu XL. Strategy to targeting the immune resistance and novel therapy in colorectal cancer. Cancer Med 2018; 7:1578-1603. [PMID: 29658188 PMCID: PMC5943429 DOI: 10.1002/cam4.1386] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Revised: 01/16/2018] [Accepted: 01/16/2018] [Indexed: 12/11/2022] Open
Abstract
Assessing the CRC subtypes that can predict the outcome of colorectal cancer (CRC) in patients with immunogenicity seems to be a promising strategy to develop new drugs that target the antitumoral immune response. In particular, the disinhibition of the antitumoral T‐cell response by immune checkpoint blockade has shown remarkable therapeutic promise for patients with mismatch repair (MMR) deficient CRC. In this review, the authors provide the update of the molecular features and immunogenicity of CRC, discuss the role of possible predictive biomarkers, illustrate the modern immunotherapeutic approaches, and introduce the most relevant ongoing preclinical study and clinical trials such as the use of the combination therapy with immunotherapy. Furthermore, this work is further to understand the complex interactions between the immune surveillance and develop resistance in tumor cells. As expected, if the promise of these developments is fulfilled, it could develop the effective therapeutic strategies and novel combinations to overcome immune resistance and enhance effector responses, which guide clinicians toward a more “personalized” treatment for advanced CRC patients.
Collapse
Affiliation(s)
- Wang Gang
- Department of Pharmaceutics, Shanghai Eighth People's Hospital, Jiangsu University, 200235, Shanghai, China
| | - Jun-Jie Wang
- Department of Pharmaceutics, Shanghai Eighth People's Hospital, Jiangsu University, 200235, Shanghai, China
| | - Rui Guan
- Hubei University of Medicine, NO. 30 People South Road, Shiyan City, Hubei Province, 442000, China
| | - Sun Yan
- Hubei University of Medicine, NO. 30 People South Road, Shiyan City, Hubei Province, 442000, China
| | - Feng Shi
- Department of Medicine, Jiangsu University, Zhenjiang City, Jiangsu Province, 212001, China
| | - Jia-Yan Zhang
- Department of Pharmaceutics, Shanghai Eighth People's Hospital, Jiangsu University, 200235, Shanghai, China
| | - Zi-Meng Li
- Department of Pharmaceutics, Shanghai Eighth People's Hospital, Jiangsu University, 200235, Shanghai, China
| | - Jing Gao
- Department of Medicine, Jiangsu University, Zhenjiang City, Jiangsu Province, 212001, China
| | - Xing-Li Fu
- Department of Medicine, Jiangsu University, Zhenjiang City, Jiangsu Province, 212001, China
| |
Collapse
|
82
|
De Mattia E, Cecchin E, Montico M, Labriet A, Guillemette C, Dreussi E, Roncato R, Bignucolo A, Buonadonna A, D'Andrea M, Coppola L, Lonardi S, Lévesque E, Jonker D, Couture F, Toffoli G. Association of STAT-3 rs1053004 and VDR rs11574077 With FOLFIRI-Related Gastrointestinal Toxicity in Metastatic Colorectal Cancer Patients. Front Pharmacol 2018; 9:367. [PMID: 29706892 PMCID: PMC5908896 DOI: 10.3389/fphar.2018.00367] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Accepted: 03/28/2018] [Indexed: 12/26/2022] Open
Abstract
Pharmacogenomics has largely been applied to the personalization of irinotecan-based treatment, focusing mainly on the study of genetic variants in adsorption, distribution, metabolism, and excretion (ADME) genes. The transcriptional control of ADME gene expression is mediated by a set of nuclear factors responding to cancer-related inflammation, which could have pharmacological implications. The aim of the present study was to uncover novel genetic predictors of neutropenia and gastrointestinal toxicity risk among 246 haplotype-tagging polymorphisms in 22 genes encoding inflammation-related cytokines and transcriptional regulators of ADME genes. The study comprised overall more than 400 metastatic colorectal cancer (mCRC) patients treated with first-line FOLFIRI, grouped in a discovery and a replication cohorts. A concordant protective effect of STAT-3 rs1053004 polymorphism against the risk of grade 3–4 gastrointestinal toxicity was observed in both the cohorts of patients (OR = 0.51, p = 0.045, q = 0.521 and OR = 0.39, p = 0.043, respectively). VDR rs11574077 polymorphism was demonstrated to affect both irinotecan biliary index (BI) and glucuronidation ratio (GR) by a pharmacokinetic analysis. This effect was consistent with an increased risk of grade 3–4 gastrointestinal toxicity in the discovery cohort (OR = 4.46, p = 0.010, q = 0.305). The association was not significant in the replication cohort (OR = 1.44, p = 0.601). These findings suggest an effect of STAT-3 and VDR polymorphisms on FOLFIRI-related gastrointestinal toxicity. If prospectively validated as predictive markers, they could be used to improve the clinical management of mCRC.
Collapse
Affiliation(s)
- Elena De Mattia
- Clinical and Experimental Pharmacology, Centro di Riferimento Oncologico, National Cancer Institute, Aviano, Italy
| | - Erika Cecchin
- Clinical and Experimental Pharmacology, Centro di Riferimento Oncologico, National Cancer Institute, Aviano, Italy
| | - Marcella Montico
- Clinical and Experimental Pharmacology, Centro di Riferimento Oncologico, National Cancer Institute, Aviano, Italy
| | - Adrien Labriet
- Pharmacogenomics Laboratory, Centre Hospitalier Universitaire de Québec, Research Center and Faculty of Pharmacy, Laval University, Québec, QC, Canada
| | - Chantal Guillemette
- Pharmacogenomics Laboratory, Centre Hospitalier Universitaire de Québec, Research Center and Faculty of Pharmacy, Laval University, Québec, QC, Canada
| | - Eva Dreussi
- Clinical and Experimental Pharmacology, Centro di Riferimento Oncologico, National Cancer Institute, Aviano, Italy
| | - Rossana Roncato
- Clinical and Experimental Pharmacology, Centro di Riferimento Oncologico, National Cancer Institute, Aviano, Italy
| | - Alessia Bignucolo
- Clinical and Experimental Pharmacology, Centro di Riferimento Oncologico, National Cancer Institute, Aviano, Italy
| | - Angela Buonadonna
- Medical Oncology Unit, Centro di Riferimento Oncologico, National Cancer Institute, Aviano, Italy
| | - Mario D'Andrea
- Medical Oncology Unit, San Filippo Neri Hospital, Rome, Italy
| | - Luigi Coppola
- Pathology Unit, San Filippo Neri Hospital, Rome, Italy
| | - Sara Lonardi
- Medical Oncology Unit 1, Istituto Oncologico Veneto, Istituto Di Ricovero e Cura a Carattere Scientifico, Padua, Italy
| | - Eric Lévesque
- Centre Hospitalier Universitaire de Québec Research Center, Faculty of Medicine, Laval University, Québec, QC, Canada
| | - Derek Jonker
- Division of Medical Oncology, Department of Medicine, Ottawa Hospital, University of Ottawa, Ottawa, ON, Canada
| | - Félix Couture
- Centre Hospitalier Universitaire de Québec Research Center, Faculty of Medicine, Laval University, Québec, QC, Canada
| | - Giuseppe Toffoli
- Clinical and Experimental Pharmacology, Centro di Riferimento Oncologico, National Cancer Institute, Aviano, Italy
| |
Collapse
|
83
|
Ling ZA, Zhang LJ, Ye ZH, Dang YW, Chen G, Li RL, Zeng JJ. Immunohistochemical distribution of FOXP3+ regulatory T cells in colorectal cancer patients. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2018; 11:1841-1854. [PMID: 31938291 PMCID: PMC6958190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Accepted: 02/22/2018] [Indexed: 06/10/2023]
Abstract
Background: Recent studies have focused on less invasive methods for diagnosing and predicting survival outcomes for colorectal cancer (CRC) patients. Objective: We studied the role of the transcription factor forkhead box P3 (FOXP3) in the tumorigenesis of CRC and investigated its prognostic value in survival outcome estimates. Methods: FOXP3+ regulatory T (Treg) cell levels in CRC and adjacent tissues were measured by immunohistochemistry (IHC) and compared statistically. A literature search was conducted on FOXP3+ Treg cell density and survival rates, including overall survival, disease-free survival, and cancer-specific survival. Meta-analyses were then performed to determine the prognostic value of FOXP3+ Treg cell levels in CRC patients. Results: FOXP3+ Treg cells were increased in CRC tissues over adjacent controls according to the IHC results (t = 14.321; P < 0.001) and cell densities in cases with metastases were higher than those without metastasis (P < 0.05). Cases with serosal infiltration showed higher FOXP3+ Treg cell densities compared to cases without infiltration (P < 0.05) and cell densities in cases differentiated to a lower degree than in cases showing a medium to high degree of differentiation (P < 0.05). Moreover, meta-analysis found a high FOXP3+ Treg cells density in CRC tissues (standardized mean differences = 0.30 [95% CI: 0.03-0.57]), which was correlated with better overall patient survival outcome (hazard ratio = 0.749 [95% CI: 0.648-0.867]). Conclusions: Increased FOXP3+ Treg cells may be an effective marker for tumorigenesis and clinical prognostic evaluation for CRC patients.
Collapse
Affiliation(s)
- Zhi-An Ling
- Department of Emergency, Second Affiliated Hospital of Guangxi Medical UniversityNanning, Guangxi, China
| | - Li-Jie Zhang
- Department of Pathology, First Affiliated Hospital of Guangxi Medical UniversityNanning, Guangxi Zhuang Autonomous Region, China
| | - Zhi-Hua Ye
- Department of Pathology, First Affiliated Hospital of Guangxi Medical UniversityNanning, Guangxi Zhuang Autonomous Region, China
| | - Yi-Wu Dang
- Department of Pathology, First Affiliated Hospital of Guangxi Medical UniversityNanning, Guangxi Zhuang Autonomous Region, China
| | - Gang Chen
- Department of Pathology, First Affiliated Hospital of Guangxi Medical UniversityNanning, Guangxi Zhuang Autonomous Region, China
| | - Ruo-Lin Li
- Department of Scientific Research, First Affiliated Hospital of Guangxi Medical UniversityNanning, Guangxi Zhuang Autonomous Region, China
| | - Jing-Jing Zeng
- Department of Pathology, First Affiliated Hospital of Guangxi Medical UniversityNanning, Guangxi Zhuang Autonomous Region, China
| |
Collapse
|
84
|
Yu Y, Ma X, Gong R, Zhu J, Wei L, Yao J. Recent advances in CD8 + regulatory T cell research. Oncol Lett 2018; 15:8187-8194. [PMID: 29805553 DOI: 10.3892/ol.2018.8378] [Citation(s) in RCA: 114] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2017] [Accepted: 02/01/2018] [Indexed: 11/05/2022] Open
Abstract
Various subgroups of CD8+ T lymphocytes do not only demonstrate cytotoxic effects, but also serve important regulatory roles in the body's immune response. In particular, CD8+ regulatory T cells (CD8+ Tregs), which possess important immunosuppressive functions, are able to effectively block the overreacting immune response and maintain the body's immune homeostasis. In recent years, studies have identified a small set of special CD8+ Tregs that can recognize major histocompatibility complex class Ib molecules, more specifically Qa-1 in mice and HLA-E in humans, and target the self-reactive CD4+ T ce lls. These findings have generated broad implications in the scientific community and attracted general interest to CD8+ Tregs. The present study reviews the recent research progress on CD8+ Tregs, including their origin, functional classification, molecular markers and underlying mechanisms of action.
Collapse
Affiliation(s)
- Yating Yu
- Department of Medical School, Guangxi University of Science and Technology, Liuzhou, Guangxi 545005, P.R. China
| | - Xinbo Ma
- Department of Medical School, Guangxi University of Science and Technology, Liuzhou, Guangxi 545005, P.R. China
| | - Rufei Gong
- Department of Medical School, Guangxi University of Science and Technology, Liuzhou, Guangxi 545005, P.R. China
| | - Jianmeng Zhu
- Department of Chunan First People's Hospital, Hangzhou, Zhejiang 310000, P.R. China
| | - Lihua Wei
- Department of Medical School, Guangxi University of Science and Technology, Liuzhou, Guangxi 545005, P.R. China
| | - Jinguang Yao
- Department of Medical School, Guangxi University of Science and Technology, Liuzhou, Guangxi 545005, P.R. China
| |
Collapse
|
85
|
Zhu Z, Dong W. Overexpression of HHLA2, a member of the B7 family, is associated with worse survival in human colorectal carcinoma. Onco Targets Ther 2018; 11:1563-1570. [PMID: 29593422 PMCID: PMC5865557 DOI: 10.2147/ott.s160493] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Background Colorectal carcinoma (CRC) is one of the most common malignancies, and immunotherapy has opened a new field of cancer treatment in recent years. Generally, CRC does not benefit from immunotherapy. HHLA2, a member of the B7 family, is a novel immune checkpoint molecule, and the prognostic value of HHLA2 in CRC patients and the association between HHLA2 expression and clinicopathological characteristics remains unknown. Materials and methods This study included 63 patients diagnosed with CRC, and their resected specimens were obtained and constructed as a tissue microarray. Expression of HHLA2 and CD8 was detected by the double immunohistochemistry method. Based on follow-up data, correlations of HHLA2 expression and clinicopathological features, including overall survival, in CRC patients were evaluated. Results High HHLA2 expression was detected in CRC tumor tissues, compared to the adjacent noncancerous tissues. HHLA2 expression level was significantly related to the depth of invasion (P=0.044) and CD8+ T-cell infiltration status (P=0.016), and predicted high mortality rate (P=0.035). HHLA2 acted as an independent predictive factor in the overall survival of CRC patients (P=0.039, hazard ratio=2.162, 95% CI 1.041–3.084). Conclusion HHLA2 expression is upregulated in CRC patients, and HHLA2 is an independent prognostic factor of overall survival of CRC patients. High HHLA2 expression is closely correlated with CD8 T-cell infiltration status and can predict poor prognosis in CRC patients.
Collapse
Affiliation(s)
- Ziwen Zhu
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China.,Hubei Key Laboratory of Digestive System Disease, Wuhan, People's Republic of China.,Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China
| | - Weiguo Dong
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China.,Hubei Key Laboratory of Digestive System Disease, Wuhan, People's Republic of China.,Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China
| |
Collapse
|
86
|
Abstract
Over the past few years, the role of immunotherapy in colorectal cancer (CRC) has expanded, specifically in subsets of CRC with microsatellite instability (MSI) for which newer agents, such as programmed death-1 (PD-1) inhibitors, are efficacious. While other immunotherapeutic agents are more immature in development, they have the potential to enhance the efficacy of PD-1 inhibitors and play a role in the treatment algorithm for all subsets of CRC patients. In this review, we will discuss immunotherapeutic agents in development in patients with CRC. We will review the later phase studies that elucidate the role of immunotherapy in CRC and provide hope for changing the treatment paradigm for CRC in the future.
Collapse
Affiliation(s)
- Sukeshi Patel Arora
- University of Texas Health Science Center San Antonio, 7979 Wurzbach Rd, MC8026, San Antonio, TX 78229, USA
| | - Devalingam Mahalingam
- University of Texas Health Science Center San Antonio, 7979 Wurzbach Rd, MC8026, San Antonio, TX 78229, USA
| |
Collapse
|
87
|
Li H, Huang K, Gao L, Wang L, Niu Y, Liu H, Wang Z, Wang L, Wang G, Wang J. TES inhibits colorectal cancer progression through activation of p38. Oncotarget 2018; 7:45819-45836. [PMID: 27323777 PMCID: PMC5216763 DOI: 10.18632/oncotarget.9961] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Accepted: 05/29/2016] [Indexed: 02/06/2023] Open
Abstract
The human TESTIN (TES) gene has been identified as a candidate tumor suppressor based on its location at a common fragile site – a region where loss of heterozygosity has been detected in numerous types of tumors. To investigate its role in colorectal cancer (CRC), we examined TES protein levels in CRC tissue samples and cell lines. We observed that TES was markedly reduced in both CRC tissue and cell lines. Additionally, overexpression of TES significantly inhibited cell proliferation, migration, and invasion, while increasing cell apoptosis in colon cancer cells. By contrast, shRNA-mediated TES knockdown elicited the opposite effects. TES inhibited the progression of CRC by up-regulating pro-apoptotic proteins, down-regulating anti-apoptotic proteins, and simultaneously activating p38 mitogen-activated protein kinase (MAPK) signaling pathways. Collectively, these data indicate that TES functions as a necessary suppressor of CRC progression by activating p38-MAPK signaling pathways. This suggests that TES may have a potential application in CRC diagnosis and targeted gene therapy.
Collapse
Affiliation(s)
- Huili Li
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Kun Huang
- Institution of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Lu Gao
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Lixia Wang
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yanfeng Niu
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Hongli Liu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Zheng Wang
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.,Research Center for Tissue Engineering and Regenerative Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Lin Wang
- Research Center for Tissue Engineering and Regenerative Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Guobin Wang
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jiliang Wang
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
88
|
Rumba R, Cipkina S, Cukure F, Vanags A. Systemic and local inflammation in colorectal cancer. Acta Med Litu 2018; 25:185-196. [PMID: 31308824 PMCID: PMC6591690 DOI: 10.6001/actamedica.v25i4.3929] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2018] [Accepted: 02/26/2019] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Colorectal cancer (CRC) is the third most common cancer in the world. The cornerstone of CRC treatment is surgical resection. However, patients in the same TNM stage show different recurrence rates and survival. Of patients with a local disease without lymph node or a distant metastasis, 20-25% still develop recurrence. There is evidence that inflammatory reaction is one of the key elements in tumour development. MATERIALS AND METHODS We reviewed literature on colorectal cancer and its relationships with the immune system, with special focus on local and systemic inflammatory reaction. The Pubmed and ClinicalKey databases were searched using the key words colorectal cancer, local inflammation, systemic inflammation, markers of inflammation. The relevant literature was reviewed and included in the article. RESULTS The immune system has two-sided relationships with cancer, so it not only performs anti-tumour activities, but can also promote tumour growth and spread. Research has shown that signs of local inflammation are associated with a better prognosis in CRC. Systemic inflammation has been associated with more aggressive behaviour and a worse prognosis for patients with several cancers, including CRC. CONCLUSIONS Recent findings in tumour biology have improved our understanding of colorectal cancer and of the natural course of this disease. Several markers of local and systemic inflammatory reaction have been identified. The next step is to find the most accurate and applicable marker, so that this promising tool can be used in clinical practice and aid in decision making.
Collapse
Affiliation(s)
- Roberts Rumba
- Department of Surgery, Riga Stradins University, Riga, Latvia
| | - Sandra Cipkina
- Faculty of Medicine, Riga Stradins University, Riga, Latvia
| | - Fanija Cukure
- Faculty of Medicine, Riga Stradins University, Riga, Latvia
| | - Andrejs Vanags
- Department of Surgery, Riga Stradins University, Riga, Latvia
| |
Collapse
|
89
|
Chantima W, Thepthai C, Cheunsuchon P, Dharakul T. EpCAM expression in squamous cell carcinoma of the uterine cervix detected by monoclonal antibody to the membrane-proximal part of EpCAM. BMC Cancer 2017; 17:811. [PMID: 29202724 PMCID: PMC5715649 DOI: 10.1186/s12885-017-3798-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2016] [Accepted: 11/17/2017] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Epithelial cell adhesion molecule (EpCAM) is a promising biomarker for squamous cell carcinoma (SCC) of the uterine cervix, because it is over-expressed in various cancers of epithelial origin. However, EpCAM expression reported in previous immunohistochemistry (IHC) studies was inconsistent. We hypothesize that the membrane-distal part of EpCAM may be lost during tissue preparation, leaving only the membrane-proximal part of EpCAM available for antibody binding and IHC staining. METHODS Two new anti-EpCAM MAbs to the membrane-proximal part (WC-2) and the membrane-distal part (WC-1) of EpCAM were generated and characterized. WC-2 was selected for its ability to detect EpCAM in cervical tissues by IHC. One hundred thirty-five archival paraffin-embedded tissues previously diagnosed as cervical SCC (n=44), high-grade (HSIL) (n=43), or low-grade (LSIL) (n=48) squamous intraepithelial lesions were examined. IHC score was collected, recorded, and analyzed for distribution, intensity, and percentage of cancer cells stained for EpCAM. RESULTS EpCAM expression was consistently detected on cervical tissues by WC-2, but not by WC-1. EpCAM was expressed with high IHC score in the majority of cervical SCC (37/44), but not in normal epithelial area adjacent to SCC. EpCAM was also highly expressed on precancerous lesion of the cervix, particularly in HSIL. More importantly, EpCAM expression could be used to distinguish between HSIL and LSIL, according to staining distribution. HSIL tissues displayed EpCAM expression in two-thirds to full thickness of the epithelium, while in LSIL the staining was limited to the lower one-third of the thickness. The IHC score of EpCAM expression was strongly correlated with cervical cancer and grades of precancerous lesions (r=0.875, p<0.001). CONCLUSION Only the anti-EpCAM MAb to the membrane-proximal part is able to detect EpCAM on paraffin-embedded cervical cancer tissues. A strong positive correlation between EpCAM expression level and the grades of SILs provides the possibility that EpCAM can be used to predict prognosis and severity in these patients.
Collapse
Affiliation(s)
- Warangkana Chantima
- Graduate Program in Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Charin Thepthai
- Department of Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Pornsuk Cheunsuchon
- Department of Pathology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand.
| | - Tararaj Dharakul
- Department of Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand.
| |
Collapse
|
90
|
Barliana MI, Diantini A, Subarnas A, Abdulah R, Izumi T. Inhibition of Phosphorylated c-Jun NH(2)-terminal Kinase by 2',4'-dihydroxy-6-methoxy-3,5-dimethylchalcone Isolated from Eugenia aquea Burm f. Leaves in Jurkat T-cells. Pharmacogn Mag 2017; 13:S573-S577. [PMID: 29142417 PMCID: PMC5669100 DOI: 10.4103/pm.pm_16_17] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Revised: 03/01/2017] [Indexed: 01/15/2023] Open
Abstract
Background: Indonesian medicinal plants have been used for their anticancer activity for decades. However, the therapeutic effects of medicinal plants have not been fully examined scientifically. As cancer is a major health problem worldwide, searching for a new anticancer compound has attracted considerable attention. Our previous study found that 2’,4’-dihydroxy-6-methoxy-3,5-dimethylchalcone, an active compound isolated from leaves of Indonesian medicinal plants Eugenia aquea Burm f. (Myrtaceae), had anticancer activity in MCF-7 human breast cancer cells through induction of apoptosis. Objective: To investigate the molecular mechanism of 2’,4’-dihydroxy-6-methoxy-3,5-dimethylchalcone antiproliferative activity. Materials and Methods: Leaves of E. aquea were extracted by ethanol, fractionated by ethyl acetate, n-hexane, or water, and isolated for its active compound. Jurkat T-cells were treated with 2’,4’-dihydroxy-6-methoxy-3,5-dimethylchalcone for 12 and 24 h, and a cell viability assay and real-time-reverse transcriptase polymerase chain reaction for interleukin-2 (IL-2) mRNA measurement were performed. The effects of active compound to mitogen-activated protein kinases were also examined to investigate the mechanism of its antiproliferative activity. Results: 2’,4’-dihydroxy-6-methoxy-3,5-dimethylchalcone inhibited Jurkat T-cell proliferation with a half maximal inhibitory concentration of 59.5 mM. Although IL-2 mRNA expression was slightly increased after treatment, it inhibited c-Jun N-terminal kinase expression but not p38 and extracellular signal-regulated kinase expression. Conclusions: Our study indicated that the molecular mechanism mediating the antiproliferative activity of 2’,4’-dihydroxy-6-methoxy-3,5-dimethylchalcone may be attributed to the stimulation of an immunological microenvironment in the cells. SUMMARY 2’,4’-dihydroxy-6-methoxy-3,5-dimethylchalcone was isolated from Eugenia aquea. The antiproliferative activity of 2’,4’-dihydroxy-6- methoxy-3,5-dimethylchalcone significantly showed in Jurkat T-cells with a half maximal inhibitory concentration of 59.5 mM through inhibition of c-Jun N-terminal kinase phosphorylation. Interleukin-2 mRNA expression was also slightly increased after treatment with the compound, and this result may be indicated to the stimulation of the immunological microenvironment in T-cells.
Abbreviations used:E. aquea: Eugenia aquea, IL-2: Interleukin-2, MAPK: Mitogen-activated protein kinase, ERKs: Extracellular signal-regulated kinases, JNKs: c-Jun N-terminal kinases, p38: p38 MAPK, PI3K: Phosphatidylinositol-3 kinase, IC50: Half maximal inhibitory concentration.
Collapse
Affiliation(s)
- Melisa I Barliana
- Department of Biological Pharmacy, Faculty of Pharmacy, Faculty of Pharmacy, Universitas Padjadjaran, Jatinangor 45363, Indonesia.,Center for Drug Discovery and Product Development, Faculty of Pharmacy, Universitas Padjadjaran, Jatinangor 45363, Indonesia
| | - Ajeng Diantini
- Department of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, Universitas Padjadjaran, Jatinangor 45363, Indonesia
| | - Anas Subarnas
- Department of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, Universitas Padjadjaran, Jatinangor 45363, Indonesia
| | - Rizky Abdulah
- Center for Drug Discovery and Product Development, Faculty of Pharmacy, Universitas Padjadjaran, Jatinangor 45363, Indonesia.,Department of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, Universitas Padjadjaran, Jatinangor 45363, Indonesia
| | - Takashi Izumi
- Department of Biochemistry, Graduate School of Medicine, Gunma University, Gunma Prefecture, Japan
| |
Collapse
|
91
|
Pan P, Kang S, Wang Y, Liu K, Oshima K, Huang YW, Zhang J, Yearsley M, Yu J, Wang LS. Black Raspberries Enhance Natural Killer Cell Infiltration into the Colon and Suppress the Progression of Colorectal Cancer. Front Immunol 2017; 8:997. [PMID: 28861089 PMCID: PMC5561013 DOI: 10.3389/fimmu.2017.00997] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Accepted: 08/04/2017] [Indexed: 12/24/2022] Open
Abstract
Natural killer (NK) cells are an essential component of innate immunity against cancer development. Many studies have been conducted to evaluate immune-modulating effects using dietary compounds. Our laboratory has been investigating the chemopreventive potential of black raspberries (BRBs) and previously demonstrated their beneficial modulation of genetic and epigenetic biomarkers in patients with colorectal cancer (CRC). The current study investigated their potential on modulating NK cells. To avoid the excessive inflammation caused by the dextran sulfate sodium (DSS) treatment that leads to colitis, we treated the mice with overnight DSS so that it would slightly irritate the colon but still promote colon carcinogenesis with 100% incidence in both the ApcMin/+ mice and azoxymethane (AOM)-treated mice. A significant decrease of tissue-infiltrating NK cells along the progression of microadenoma-to-adenoma and adenoma-to-adenocarcinoma was observed in the ApcMin/+ /DSS and AOM/DSS mice, respectively. Depletion of NK cells significantly promoted the development of CRC, suggesting a critical role of NK cells in combating CRC progression. BRBs significantly suppressed the CRC progression and increased the number of tissue-infiltrating NK cells in both mouse models. Moreover, we further determined BRBs' effects on NK cells in the human biopsy specimens collected from our previously completed clinical trial, in which CRC patients consumed BRBs for an average of 4 weeks during a presurgical window. We observed an increased number and an enhanced cytotoxicity of NK cells by BRB intervention. The current study provides evidence that BRBs have the potential to enhance the tumor immunesurveillance of NK cells that can be beneficial in the setting of CRC prevention and treatment.
Collapse
Affiliation(s)
- Pan Pan
- Division of Hematology and Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Siwen Kang
- Division of Hematology, Department of Internal Medicine, College of Medicine, The Ohio State University, Columbus, OH, United States
| | - Youwei Wang
- Division of Hematology, Department of Internal Medicine, College of Medicine, The Ohio State University, Columbus, OH, United States
| | - Ka Liu
- Division of Hematology, Department of Internal Medicine, College of Medicine, The Ohio State University, Columbus, OH, United States
| | - Kiyoko Oshima
- Department of Pathology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Yi-Wen Huang
- Department of Obstetrics and Gynecology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Jianying Zhang
- Center for Biostatistics, The Ohio State University, Columbus, OH, United States
| | - Martha Yearsley
- Department of Pathology, The Ohio State University, Columbus, OH, United States
| | - Jianhua Yu
- Division of Hematology, Department of Internal Medicine, College of Medicine, The Ohio State University, Columbus, OH, United States.,Comprehensive Cancer Center, The James Cancer Hospital, The Ohio State University, Columbus, OH, United States
| | - Li-Shu Wang
- Division of Hematology and Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, United States
| |
Collapse
|
92
|
Garajová I, Ferracin M, Porcellini E, Palloni A, Abbati F, Biasco G, Brandi G. Non-Coding RNAs as Predictive Biomarkers to Current Treatment in Metastatic Colorectal Cancer. Int J Mol Sci 2017; 18:ijms18071547. [PMID: 28714940 PMCID: PMC5536035 DOI: 10.3390/ijms18071547] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2017] [Revised: 07/13/2017] [Accepted: 07/14/2017] [Indexed: 12/22/2022] Open
Abstract
The onset and selection of resistant clones during cancer treatment with chemotherapy or targeted therapy is a major issue in the clinical management of metastatic colorectal cancer patients. It is possible that a more personalized treatment selection, using reliable response-to-therapy predictive biomarkers, could lead to an improvement in the success rate of the proposed therapies. Although the process of biomarker selection and validation could be a long one, requiring solid statistics, large cohorts and multicentric validations, non-coding RNAs (ncRNAs) and in particular microRNAs, proved to be extremely promising in this field. Here we summarize some of the main studies correlating specific ncRNAs with sensitivity/resistance to chemotherapy, anti-VEGF therapy, anti-EGFR therapy and immunotherapy in colorectal cancer (CRC).
Collapse
Affiliation(s)
- Ingrid Garajová
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), Sant'Orsola-Malpighi Hospital, University of Bologna, 40138 Bologna, Italy.
- Interdepartmental Centre of Cancer Research "Giorgio Prodi", University of Bologna, 40138 Bologna, Italy.
| | - Manuela Ferracin
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), Sant'Orsola-Malpighi Hospital, University of Bologna, 40138 Bologna, Italy.
| | - Elisa Porcellini
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), Sant'Orsola-Malpighi Hospital, University of Bologna, 40138 Bologna, Italy.
| | - Andrea Palloni
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), Sant'Orsola-Malpighi Hospital, University of Bologna, 40138 Bologna, Italy.
| | - Francesca Abbati
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), Sant'Orsola-Malpighi Hospital, University of Bologna, 40138 Bologna, Italy.
| | - Guido Biasco
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), Sant'Orsola-Malpighi Hospital, University of Bologna, 40138 Bologna, Italy.
- Interdepartmental Centre of Cancer Research "Giorgio Prodi", University of Bologna, 40138 Bologna, Italy.
| | - Giovanni Brandi
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), Sant'Orsola-Malpighi Hospital, University of Bologna, 40138 Bologna, Italy.
| |
Collapse
|
93
|
Procaccio L, Schirripa M, Fassan M, Vecchione L, Bergamo F, Prete AA, Intini R, Manai C, Dadduzio V, Boscolo A, Zagonel V, Lonardi S. Immunotherapy in Gastrointestinal Cancers. BIOMED RESEARCH INTERNATIONAL 2017; 2017:4346576. [PMID: 28758114 PMCID: PMC5512095 DOI: 10.1155/2017/4346576] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Accepted: 05/18/2017] [Indexed: 12/29/2022]
Abstract
Gastrointestinal cancers represent a major public health problem worldwide. Immunotherapeutic strategies are currently under investigation in this setting and preliminary results of ongoing trials adopting checkpoint inhibitors are striking. Indeed, although a poor immunogenicity for GI has been reported, a strong biological rationale supports the development of immunotherapy in this field. The clinical and translational research on immunotherapy for the treatment of GI cancers started firstly with the identification of immune-related mechanisms possibly relevant to GI tumours and secondly with the development of immunotherapy-based agents in clinical trials. In the present review a general overview is firstly provided followed by a focus on major findings on gastric, colorectal, and hepatocellular carcinomas. Finally, pathological and molecular perspectives are provided since many efforts are ongoing in order to identify possible predictive biomarkers and to improve patients' selection. Many issues are still unsolved in this field; however, we strongly believe that immunotherapy might positively affect the natural history of a subgroup of GI cancer patients improving outcome and the overall quality of life.
Collapse
Affiliation(s)
- Letizia Procaccio
- Division of Medical Oncology 1, Istituto Oncologico Veneto, IRCCS, Padova, Italy
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy
| | - Marta Schirripa
- Division of Medical Oncology 1, Istituto Oncologico Veneto, IRCCS, Padova, Italy
| | - Matteo Fassan
- Department of Medicine, Surgical Pathology & Cytopathology Unit, University of Padova, Padova, Italy
| | - Loredana Vecchione
- Division of Molecular Carcinogenesis, Cancer Genomics Center Netherlands, The Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Francesca Bergamo
- Division of Medical Oncology 1, Istituto Oncologico Veneto, IRCCS, Padova, Italy
| | - Alessandra Anna Prete
- Division of Medical Oncology 1, Istituto Oncologico Veneto, IRCCS, Padova, Italy
- Department of Radiological, Oncological and Pathological Sciences, Policlinico Umberto I University Hospital, Rome, Italy
| | - Rossana Intini
- Division of Medical Oncology 1, Istituto Oncologico Veneto, IRCCS, Padova, Italy
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy
| | - Chiara Manai
- Division of Medical Oncology 1, Istituto Oncologico Veneto, IRCCS, Padova, Italy
- Department of Radiological, Oncological and Pathological Sciences, Policlinico Umberto I University Hospital, Rome, Italy
| | - Vincenzo Dadduzio
- Division of Medical Oncology 1, Istituto Oncologico Veneto, IRCCS, Padova, Italy
- Department of Medical Oncology, Fondazione Policlinico Universitario Agostino Gemelli, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Alice Boscolo
- Division of Medical Oncology 1, Istituto Oncologico Veneto, IRCCS, Padova, Italy
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy
| | - Vittorina Zagonel
- Division of Medical Oncology 1, Istituto Oncologico Veneto, IRCCS, Padova, Italy
| | - Sara Lonardi
- Division of Medical Oncology 1, Istituto Oncologico Veneto, IRCCS, Padova, Italy
| |
Collapse
|
94
|
Mion F, Vetrano S, Tonon S, Valeri V, Piontini A, Burocchi A, Petti L, Frossi B, Gulino A, Tripodo C, Colombo MP, Pucillo CE. Reciprocal influence of B cells and tumor macro and microenvironments in the ApcMin/+ model of colorectal cancer. Oncoimmunology 2017; 6:e1336593. [PMID: 28919998 DOI: 10.1080/2162402x.2017.1336593] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Revised: 05/25/2017] [Accepted: 05/26/2017] [Indexed: 12/22/2022] Open
Abstract
One of the most fascinating aspects of the immune system is its dynamism, meant as the ability to change and readapt according to the organism needs. Following an insult, we assist to the spontaneous organization of different immune cells which cooperate, locally and at distance, to build up an appropriate response. Throughout tumor progression, adaptations within the systemic tumor environment, or macroenvironment, result in the promotion of tumor growth, tumor invasion and metastasis to distal organs, but also to dramatic changes in the activity and composition of the immune system. In this work, we show the changes of the B-cell arm of the immune system following tumor progression in the ApcMin/+ model of colorectal cancer. Tumor macroenvironment leads to an increased proportion of total and IL-10-competent B cells in draining LNs while activates a differentiation route that leads to the expansion of IgA+ lymphocytes in the spleen and peritoneum. Importantly, serum IgA levels were significantly higher in ApcMin/+ than Wt mice. The peculiar involvement of IgA response in the adenomatous transformation had correlates in the gut-mucosal compartment where IgA-positive elements increased from normal mucosa to areas of low grade dysplasia while decreasing upon overt carcinomatous transformation. Altogether, our findings provide a snapshot of the tumor education of B lymphocytes in the ApcMin/+ model of colorectal cancer. Understanding how tumor macroenvironment affects the differentiation, function and distribution of B lymphocytes is pivotal to the generation of specific therapies, targeted to switching B cells to an anti-, rather than pro-, tumoral phenotype.
Collapse
Affiliation(s)
- Francesca Mion
- Department of Medicine, University of Udine, Udine, Italy
| | - Stefania Vetrano
- Inflammatory Bowel Disease Center, Humanitas Research Hospital, Rozzano, Italy.,Department of Biomedical Sciences, Humanitas University, Milano, Italy
| | - Silvia Tonon
- Department of Medicine, University of Udine, Udine, Italy
| | - Viviana Valeri
- Department of Medicine, University of Udine, Udine, Italy
| | - Andrea Piontini
- Department of Biomedical Sciences, Humanitas University, Milano, Italy
| | - Alessia Burocchi
- Department of Experimental Oncology and Molecular Medicine, Molecular Immunology Unit, Fondazione IRCCS Istituto Nazionale Tumori, Milano, Italy
| | - Luciana Petti
- Department of Biotechnology and Translational Medicine, University of Milan, Milano, Italy
| | - Barbara Frossi
- Department of Medicine, University of Udine, Udine, Italy
| | - Alessandro Gulino
- Department of Health Science, Tumor Immunology Unit, Human Pathology Section, Palermo University School of Medicine, Palermo, Italy
| | - Claudio Tripodo
- Department of Health Science, Tumor Immunology Unit, Human Pathology Section, Palermo University School of Medicine, Palermo, Italy
| | - Mario P Colombo
- Department of Experimental Oncology and Molecular Medicine, Molecular Immunology Unit, Fondazione IRCCS Istituto Nazionale Tumori, Milano, Italy
| | | |
Collapse
|
95
|
Augestad KM, Merok MA, Ignatovic D. Tailored Treatment of Colorectal Cancer: Surgical, Molecular, and Genetic Considerations. Clin Med Insights Oncol 2017; 11:1179554917690766. [PMID: 28469509 PMCID: PMC5395262 DOI: 10.1177/1179554917690766] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2016] [Accepted: 01/06/2017] [Indexed: 12/12/2022] Open
Abstract
Colorectal cancer (CRC) is a complex cancer disease, and approximately 40% of the surgically cured patients will experience cancer recurrence within 5 years. During recent years, research has shown that CRC treatment should be tailored to the individual patient due to the wide variety of risk factors, genetic factors, and surgical complexity. In this review, we provide an overview of the considerations that are needed to provide an individualized, patient-tailored treatment. We emphasize the need to assess the predictors of CRC, and we summarize the latest research on CRC genetics and immunotherapy. Finally, we provide a summary of the significant variations in the colon and rectal anatomy that is important to consider in an individualized surgical approach. For the individual patient with CRC, a tailored treatment approach is needed in the preoperative, operative, and postoperative phase.
Collapse
Affiliation(s)
- Knut Magne Augestad
- Department of Gastrointestinal Surgery, Akershus University Hospital, Oslo, Norway
| | - Marianne A Merok
- Department of Gastrointestinal Surgery, Akershus University Hospital, Oslo, Norway
| | | |
Collapse
|
96
|
Lynch D, Murphy A. The emerging role of immunotherapy in colorectal cancer. ANNALS OF TRANSLATIONAL MEDICINE 2016; 4:305. [PMID: 27668225 DOI: 10.21037/atm.2016.08.29] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Modulation of the interaction between the immune system and the tumor microenvironment has long been a target of cancer research, including colorectal cancer (CRC). Approaches explored to date include vaccines (autologous, peptide, dendritic cell, viral and bacterial), cytokine therapy, toll-like receptors (TLRs), autologous cell therapy and checkpoint inhibition. Until recently these approaches have been shown to have only modest efficacy in reducing tumor burden. However, significant breakthroughs have been made, with the use of checkpoint inhibitors targeting programmed cell death protein-1 (PD-1), programmed cell death ligand-1 (PD-L1), and cytotoxic T lymphocyte antigen-4 (CTLA-4). Immunotherapy now represents a possible avenue of curative treatment for those with chemo-otherwise refractory tumors. Success with this approach to immunotherapy has largely been confined to tumors with high mutational burdens such as melanoma, renal cell carcinoma (RCC) and non-small cell lung cancer. This observation led to the exploration and successful use of checkpoint inhibitors in those with mismatch repair colorectal cancer which have a relatively high mutational burden. Ongoing trials are focused on further exploring the use of checkpoint inhibitors in addition to investigating the various combinations of immunotherapeutic drugs.
Collapse
Affiliation(s)
- David Lynch
- Department of Internal Medicine, University of North Carolina Hospitals, Chapel Hill, North Carolina, USA
| | - Adrian Murphy
- Department Medical Oncology, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, Maryland, USA
| |
Collapse
|
97
|
Qian DC, Xiao X, Byun J, Suriawinata AA, Her SC, Amos CI, Barth RJ. PI3K/Akt/mTOR Signaling and Plasma Membrane Proteins Are Implicated in Responsiveness to Adjuvant Dendritic Cell Vaccination for Metastatic Colorectal Cancer. Clin Cancer Res 2016; 23:399-406. [PMID: 27435399 DOI: 10.1158/1078-0432.ccr-16-0623] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Revised: 06/20/2016] [Accepted: 07/12/2016] [Indexed: 12/13/2022]
Abstract
PURPOSE We have previously demonstrated that patients with metastatic colorectal cancer who exhibit immune responses to a dendritic cell (DC) vaccine have superior recurrence-free survival following surgery, compared with patients in whom responses do not occur. We sought to characterize the patterns of T-lymphocyte infiltration and somatic mutations in metastases that are associated with and predictive of response to the DC vaccine. EXPERIMENTAL DESIGN Cytotoxic, memory, and regulatory T cells in resected metastases and surrounding normal liver tissue from 22 patients (11 responders and 11 nonresponders) were enumerated by immunohistochemistry prior to vaccine administration. In conjunction with tumor sequencing, the combined multivariate and collapsing method was used to identify gene mutations that are associated with vaccine response. We also derived a response prediction score for each patient using his/her tumor genotype data and variant association effect sizes computed from the other 21 patients; greater weighting was placed on gene products with cell membrane-related functions. RESULTS There was no correlation between vaccine response and intratumor, peritumor, or hepatic densities of T-cell subpopulations. Associated genes were found to be enriched in the PI3K/Akt/mTOR signaling axis (P < 0.001). Applying a consistent prediction score cutoff over 22 rounds of leave-one-out cross-validation correctly inferred vaccine response in 21 of 22 patients (95%). CONCLUSIONS Adjuvant DC vaccination has shown promise as a form of immunotherapy for patients with metastatic colorectal cancer. Its efficacy may be influenced by somatic mutations that affect pathways involving PI3K, Akt, and mTOR, as well as tumor surface proteins. Clin Cancer Res; 23(2); 399-406. ©2016 AACR.
Collapse
Affiliation(s)
- David C Qian
- Department of Biomedical Data Science, Dartmouth Geisel School of Medicine, Lebanon, New Hampshire
| | - Xiangjun Xiao
- Department of Biomedical Data Science, Dartmouth Geisel School of Medicine, Lebanon, New Hampshire
| | - Jinyoung Byun
- Department of Biomedical Data Science, Dartmouth Geisel School of Medicine, Lebanon, New Hampshire
| | - Arief A Suriawinata
- Department of Pathology, Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire
| | - Stephanie C Her
- Department of Computer Science, Dartmouth College, Hanover, New Hampshire
| | - Christopher I Amos
- Department of Biomedical Data Science, Dartmouth Geisel School of Medicine, Lebanon, New Hampshire.
| | - Richard J Barth
- Department of Surgery, Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire.
| |
Collapse
|
98
|
Li X, Nie J, Mei Q, Han WD. MicroRNAs: Novel immunotherapeutic targets in colorectal carcinoma. World J Gastroenterol 2016; 22:5317-5331. [PMID: 27340348 PMCID: PMC4910653 DOI: 10.3748/wjg.v22.i23.5317] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Revised: 04/23/2016] [Accepted: 05/04/2016] [Indexed: 02/06/2023] Open
Abstract
Colorectal carcinoma (CRC) is one of the most common types of cancer worldwide and the prognosis for CRC patients with recurrence or metastasis is extremely poor. Although chemotherapy and radiation therapy can improve survival, there are still numerous efforts to be performed. Immunotherapy is frequently used, either alone or in combination with other therapies, for the treatment of CRC and is a safe and feasible way to improve CRC treatment. Furthermore, the significance of the immune system in the biology of CRC has been demonstrated by retrospective assessments of immune infiltrates in resected CRC tumors. MicroRNAs (miRNAs) are short, non-coding RNAs that can regulate multiple target genes at the post-transcriptional level and play critical roles in cell proliferation, differentiation and apoptosis. MiRNAs are required for normal immune system development and function. Nevertheless, aberrant expression of miRNAs is often observed in various tumor types and leads to immune disorders or immune evasion. The immunomodulatory function of miRNAs indicates that miRNAs may ultimately be part of the portfolio of anti-cancer targets. Herein, we will review the potential roles of miRNAs in the regulation of the immune response in CRC and then move on to discuss how to utilize different miRNA targets to treat CRC. We also provide an overview of the major limitations and challenges of using miRNAs as immunotherapeutic targets.
Collapse
|
99
|
Reduced Expression of Galectin-9 Contributes to a Poor Outcome in Colon Cancer by Inhibiting NK Cell Chemotaxis Partially through the Rho/ROCK1 Signaling Pathway. PLoS One 2016; 11:e0152599. [PMID: 27028892 PMCID: PMC4814049 DOI: 10.1371/journal.pone.0152599] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Accepted: 03/16/2016] [Indexed: 12/15/2022] Open
Abstract
Galectin-9 is a widely expressed protein that is involved in immune regulation and tumorpathogenesis and serves as a marker of a poor prognosis in various types of cancers. However, the clinical impact and the precise mechanism by which this protein contributes to colon tumor progression are unclear. In the present study, we detected the expression of galectin-9 and CD56 cells using immunohistochemistry. Spearman's rank correlation was used to clarify the association between galectin-9 expression and natural killer (NK) cell infiltration. The influence of galectin-9 on NK-92 cell migration was evaluated in vitro using transwell chemotaxis assays. The role of rh-galectin-9 in F-actin polarization in NK-92 cells was investigated using laser scanning confocal microscopy. We showed that galectin-9 was expressed in 101 (78.91%) colon tumor tissues and that was expressed at lower levels in these tissues than in para-tumor tissues. Low levels of galectin-9 expression were positively correlated with a poor histological grade and lymph node metastasis (P<0.05). A Kaplan-Meier method and Cox proportional hazards regression analysis showed that overall survival was longer in patients with high galectin-9 expression in an 8-year follow-up (P<0.05). Spearman's rank correlation indicated that there was a linear correlation between galectin-9 expression and CD56+ NK cell infiltration (R(2) = 0.658; P<0.0001). Galectin-9 stimulated migration in human NK-92 cells by affecting F-actin polarization through the Rho/ROCK1 signaling pathway. These results suggest that galectin-9 expression potentially represents a novel mechanism for tumors to escape immune surveillance in colon tumors.
Collapse
|
100
|
Immune Checkpoint Modulation in Colorectal Cancer: What's New and What to Expect. J Immunol Res 2015; 2015:158038. [PMID: 26605342 PMCID: PMC4641952 DOI: 10.1155/2015/158038] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Accepted: 10/11/2015] [Indexed: 02/07/2023] Open
Abstract
Colorectal cancer (CRC), as one of the most prevalent types of cancer worldwide, is still a leading cause of cancer related mortality. There is an urgent need for more efficient therapies in metastatic disease. Immunotherapy, a rapidly expanding field of oncology, is designed to boost the body's natural defenses to fight cancer. Of the many approaches currently under study to improve antitumor immune responses, immune checkpoint inhibition has thus far been proven to be the most effective. This review will outline the treatments that take advantage of our growing understanding of the role of the immune system in cancer, with a particular emphasis on immune checkpoint molecules, involved in CRC pathogenesis.
Collapse
|