Abstract

Simple SummaryRectal cancer occurs in the lower part of the bowel, and approximately half of all rectal cancer patients receive chemoradiotherapy before surgery. In ~22% of cases the tumour is eradicated, but the reasons for different response rates between patients are largely unknown. Inflammation and the immune system are important players in the response to cancer treatment, but we do not fully understand the role they play in this clinical setting. We examined the levels of 54 inflammatory markers in normal (non-cancerous) rectal tissue and rectal cancer tissue, and we found that rectal cancer tissue was more inflammatory, and the levels of inflammatory markers correlated with obesity status. We found that irradiating rectal cancer tissue enhanced the ability of immune cells to induce an anti-tumour immune response.Locally advanced rectal cancer is treated with neoadjuvant-chemoradiotherapy; however, only ~22% of patients achieve a complete response, and resistance mechanisms are poorly understood. The role of inflammation and immune cell biology in this setting is under-investigated. In this study, we profiled the inflammatory protein secretome of normal (non-cancer) (n = 8) and malignant rectal tissue (n = 12) pre- and post-radiation in human ex vivo explant models and examined the influence of these untreated and treated secretomes on dendritic cell biology (n = 8 for cancer and normal). These resultant profiles were correlated with patient clinical characteristics. Nineteen factors were secreted at significantly higher levels from the rectal cancer secretome when compared to the normal rectal secretome; Flt-1, P1GF, IFN-γ, IL-6, IL-10, CCL20, CCL26, CCL22, CCL3, CCL4, CCL17, GM-CSF, IL-12/IL-23p40, IL-17A, IL-1α, IL-17A/F, IL-1RA, TSLP and CXCL10 (p < 0.05). Radiation was found to have differential effects on normal rectal tissue and rectal cancer tissue with increased IL-15 and CCL22 secretion following radiation from normal rectal tissue explants (p < 0.05), while no significant alterations were observed in the irradiated rectal cancer tissue. Interestingly, however, the irradiated rectal cancer secretome induced the most potent effect on dendritic cell maturation via upregulation of CD80 and PD-L1. Patient’s visceral fat area correlated with secreted factors including CCL20, suggesting that obesity status may alter the tumour microenvironment (TME). These results suggest that radiation does not have a negative effect on the ability of the rectal cancer TME to induce an immune response. Understanding these responses may unveil potential therapeutic targets to enhance radiation response and mitigate normal tissue injury. Tumour irradiation in this cohort enhances innate immune responses, which may be harnessed to improve patient treatment outcome.

Highlights

  • Rectal cancer is a malignancy that occurs in the lower part of the large intestine

  • In order to assess the differences in the inflammatory secretome between normal rectal tissue and rectal cancer tissue, we cultured 12 rectal cancer biopsies and 8 normal rectal biopsies for 24 h and used a multiplex ELISA platform to quantify the expression of 54 inflammatory proteins in the resultant normal conditioned media (NCM) and tumour conditioned media (TCM)

  • We identified 19 factors that were secreted at significantly higher levels in the rectal cancer secretome compared to the normal rectal secretome: Flt-1 (p = 0.001), P1GF (p = 0.01), IFN-γ (p = 0.04), IL-6 (p = 0.02), IL-10 (p = 0.0002), CCL20 (p = 0.005), CCL26 (p = 0.009), CCL22 (p = 0.007), CCL3 (p = 0.002), CCL4 (p = 0.0007), CCL17 (p = 0.02), GMCSF (p < 0.0001), IL-12/IL-23p40 (p = 0.01), IL-17A (p = 0.0003), IL-1α (p = 0.003), IL-17A/F (p = 0.003), IL-1RA (p = 0.03), TSLP (p = 0.007) and CXCL10 (p = 0.001) (Figure 3A–S)

Read more

Summary

Introduction

Rectal cancer is a malignancy that occurs in the lower part of the large intestine. Approximately 700,000 cases of rectal cancer are diagnosed globally each year, and the annual death toll is approximately 310,000 [1]. The role of the tumour microenvironment (TME) and the biological mechanisms underlying these responses to treatment are poorly understood. The TME describes the milieu of cancer cells, infiltrating immune cells, secreted factors and the extracellular matrix. The interaction between the tumour cells and the surrounding microenvironment profoundly affects tumour progression and treatment response [5]. The interplay between the TME, secreted inflammatory mediators and immune cell function and how it may be altered by radiation is poorly understood in the context of rectal cancer. To fully understand the alterations occurring in the rectal cancer microenvironment and its interaction with the immune system, it is important to gain an understanding of the microenvironment in a non-cancerous rectal tissue model

Objectives
Results
Discussion
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call