Emerging roles of non‑coding RNAs in the response of rectal cancer to radiotherapy (Review)

Corrigendum in: /10.3892/ijo.2021.5247

  • Authors:
    • Huyen Trang Ha Thi
    • Hong‑Quan Duong
    • Suntaek Hong
  • View Affiliations

  • Published online on: January 22, 2021     https://doi.org/10.3892/ijo.2021.5175
  • Pages: 344-358
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Radiotherapy (RT) followed by radical surgery is an effective standard treatment strategy for various types of cancer, including rectal cancer. The response to RT varies among patients, and the radiosensitivity of cancer cells determines the clinical outcome of patients. However, the application of RT to patients with radioresistant tumors may result in radiation‑induced toxicity without clinical benefits. Currently, there are no effective methods to predict the response to RT. The limitations of the methods currently used to evaluate tumor radiosensitivity, which are mainly based on clinical and radiological features, are low sensitivity and specificity. Non‑coding RNAs (ncRNAs) have emerged as a class of biomarkers for predicting radiosensitivity. In particular, the expression pattern of ncRNAs can predict the response to RT in patients with rectal cancer. Thus, ncRNAs may be used as potential biomarkers and therapeutic targets to improve the diagnosis and treatment outcome of patients with rectal cancer. In the present review, the current knowledge on the limitations of RT for rectal cancer and the association between ncRNA expression and sensitivity of rectal cancer to RT are presented. Additionally, the potential of ncRNAs as predictive biomarkers and therapeutic targets to mitigate resistance of rectal cancer to RT is discussed.

1. Introduction

Neoadjuvant chemoradiotherapy (nCRT) followed by radical surgery has been the standard treatment modality for patients with locally advanced rectal cancer (LARC); this treatment modality has improved the local control rate and alleviated the radiation-induced toxicity in patients with rectal cancer (1). However, the major challenges for enhancing the efficacy of radiotherapy (RT) are the development of radioresistance in the tumor and the radiation-induced complications and toxicities. Therefore, the efficacy of RT in rectal cancer may be improved with the identification of radioresistance markers and predictive biomarkers of radiosensitivity, as well as the elucidation of the mechanisms underlying radioresistance and radiation-induced toxicity and complications.

Radiosensitivity and radiation-induced toxicity are predicted based on the clinical factors and irradiation doses; however, these parameters have low predictive accuracy (2-4). Therefore, there is a need to identify biomarkers that can predict the response to RT in patients with LARC to optimize the radiation doses, minimize the radiation-induced complications and select the optimal treatment strategy. Previous studies have demonstrated that the expression patterns of non-coding RNAs (ncRNAs) can predict the response to RT in patients with rectal cancer (5-7). ncRNAs can be classified as short ncRNAs and long ncRNAs (lncRNAs). Short ncRNAs are further classified into several classes, including piwi-associated RNAs, short-interfering RNAs and microRNAs (miRNAs/miRs). miRNAs are short (18-25 nucleotides in length) highly conserved RNAs that regulate gene expression at the post-transcriptional level through the degradation and/or translational repression of their mRNA targets (8). Currently, various miRNAs are reported to function as master regulators of several biological processes, including cell proliferation, apoptosis and cancer development and progression (9). Several miRNAs are present in the extracellular microenvironment (10,11). Some miRNAs are stable in biological fluids (12,13). The stability of circulating miRNAs can be attributed to their ability to form complexes with proteins in fluids, which prevents RNase-mediated miRNA degradation (14). Therefore, miRNAs are considered promising biomarkers and clinical therapeutic targets for rectal cancer.

lncRNAs contain >200 nucleotides with limited or no protein-coding capacity (15). Although experimental studies have identified only a small number of lncRNAs, previous studies have suggested that lncRNAs mediate several biological processes through the regulation of gene expression and alternative splicing (16,17). Additionally, a subset of lncRNAs is reported to be involved in maintaining pluripotency of stem cells (18). Further studies have indicated that dysregulated expression levels of lncRNAs are involved in tumorigenesis and promoting resistance to therapeutics (19-22). The present review summarizes the current findings on the ability of ncRNAs to predict the response of rectal cancer to RT and emphasizes their potential as therapeutic targets to enhance the radiosensitivity of rectal cancer.

Studies on the ability of ncRNAs to predict the response of rectal cancer to RT were searched in the following databases: Medline through Pubmed (https://pubmed.ncbi.nlm.nih.gov/) and Cochrane database (https://www.cochranelibrary.com/) of systemic reviews and reference lists from the retrieved papers (January 2008-October 2020). The search terms and groups of words used were: 'Rectal cancer', 'neoadjuvant radiotherapy', 'miRNAs in rectal cancers', 'lncRNAs' or 'non-coding RNA in rectal cancer'. The type of articles included in the present review were systemic reviews, meta-analyses, randomised controlled trials and clinical trials. Published conference extracts or non-peer reviewed manuscripts were excluded. A total of 229 related articles were identified and 23 studies were included in the final review (Fig. 1). Due to the small number of studies yielded by the search criteria, and the heterogeneity of outcomes, a narrative review has been performed.

2. Incidence of rectal cancer

Globally, colorectal cancer (CRC) is the third most common cancer and the second most common cause of cancer-associated mortality (23). Rectal cancer accounts for ~30% of all CRC cases (24). Among the rectal cancer cases, ~40% cases are LARC, which is associated with poor clinical outcomes (25). In the United States of America (USA), 147,950 new cases of CRC were estimated to be diagnosed in 2020, including 104,610 cases of colon cancer and 43,340 cases of rectal cancer, with high mortality expected among CRC cases (24).

Age and environmental and lifestyle factors contribute to the development of CRC; hence, several studies have demonstrated that dietary habits, sedentary lifestyles and the increased prevalence of obesity have increased the risk of developing CRC among younger populations (<40 years old) compared with older populations (>40 years old) (26,27). Previous studies have suggested that occupational exposure to toxins may increase the risks of developing CRC (28-30). Long-term exposure to industrial fumes and wood or metal dust and the consumption of deep-fried and barbecued foods in young adulthood have been markedly associated with rectal cancer (31). Compared with individuals residing in urban areas and involved in non-farming occupations, individuals residing in rural areas where pesticides are not used and involved in farming occupations were associated with a lower risk of developing rectal cancer (32,33). Consistently, the history of exposure to industrial toxins, unhealthy dietary habits and sedentary lifestyle have been positively associated with an increased incidence of rectal cancer in young patients (26,34).

3. Current treatment paradigms for rectal cancer

Recent advances in cancer therapeutics have increased the therapeutic efficacy in patients with rectal cancer. Surgery is the only curative therapy for patients with rectal cancer, especially for those with rectal adenocarcinoma (35,36). The development of combined modality treatment strategies, such as the combination of RT, chemotherapy and total mesorectal excision (TME), has improved the clinical outcomes and decreased the local failure rates in patients with LARC (37). The development of improved staging and surgical techniques, and the utilization of targeted combinatorial therapies have resulted in a significant increase in loco regional control and slightly improved overall survival in trials (37-39).

Recent advances in the field of anatomy and physiology have contributed to the development of advanced surgical procedures. Heald et al (40) developed a breakthrough procedure called TME, which is now the standard surgical procedure for patients with LARC. TME involves the complete surgical resection of discontinuous tumor remaining in the mesorectum, which may cause local treatment failure, with the preservation of the pelvic nerves using sharp instruments under direct vision (41). Subsequently, MacFarlane et al (42) demonstrated that the local recurrence rates in patients with rectal cancer decreased from 30-40% without TME to <5% with TME. Another study reported that the local recurrence rates of 1,411 patients with rectal cancer decreased from 32-35% with conventional surgeries to 4-9% with TME (43). These results confirmed that TME is an optimal surgical procedure for patients with rectal cancer, in which it markedly improves the clinical outcome and decreases the local recurrence.

Although TME can markedly decrease the local recurrence rate in patients with early-stage rectal cancer (42), the local regional recurrence rate in patients with stage III rectal cancer is high (~30%), with a high frequency of lymph node metastasis in lower rectal cancer (44,45). However, extensive tumor resection is not prescribed due to the severe side effects associated with lateral dissection, including impaired urinary system, loss of sexual ability and decreased 5-year survival rate (46). Therefore, additional treatment strategies have been developed to decrease the tumor volume before surgery. nCRT followed by TME has been developed and recommended by the National Comprehensive Cancer Network (NCCN) guidelines (47) as the standard therapy for stage II/III RC treatment. It can be used to shrink the volume of primary or metastatic tumors, resulting in decreasing the tumor stages with reduced loco-regional recurrence and distant metastasis rate (48). Most patients receiving nCRT before an operation are in better physical condition and have a better tolerance compared with those receiving CRT after operation (38). In case of patients with unresectable local advance RC, chemo-radiotherapy has become the only standard therapy until the patients are available for surgery (39). Except for patients with contraindications, recommended radiotherapy schedules for LARC include two radiation treatment paradigms: Short-course (SC)- and long-course (LC)-RT. SC-RT is given as 5 daily fractions of 5 Gy (5 Gy x 5 fractions) over 1 week or 25 Gy total followed by immediate surgical resection within 1 week (49). LC-RT has been shown as treatment of 45-50.4 Gy in 25-28 fractions with concurrent chemotherapy followed by delayed TME from 4 to 8 weeks after CRT (50). Both SC-RT and LC-CRT are effective therapies and multiple trials have demonstrated the equal efficacy and safety of SC-RT and LC-RT therapy, with no significant differences in long-term outcome with regards to local recurrence rate, survival and late toxicity between SC-RT and LC-CRT (51-53). However, the choice of the optimal approach remains controversial. In the USA, LC-RT has been used as the standard of care for treatment of patients with LARC, while preoperative SC-RT has been more used by radiation oncologists in Western Europe (54,55). Radiation oncologists in the USA have emphasized that LC-RT improves the complete response rate of patients, with lower surgical morbidity, less incidence of positive radial resection margins and increased tumor down-staging due to a longer delay between irradiation and surgical resection (56). SC-RT has just been prescribed for patients not receiving chemotherapy or having a geographic barrier to receiving LC-CRT (54). Conversely, supporters of SC-RT have suggested that patients treated with SC-RT received more benefits of lower cost and less acute radiation toxicity without different long-term outcomes compared with LC-RT (57). Although the positive outcomes of nCRT have been widely demonstrated and used in Western Europe and the USA, nCRT is still not a standard treatment in Japan (58,59). The application of nCRT to Japanese patients with LARC has met several obstacles. Normally, Japanese people are thinner and with less visceral fat than individuals in Western countries and the USA, therefore making surgery a favorable choice (60). Another reason is the lack of radiation oncologists, RT technologists and medical physicists in Japan, resulting in the rate of new patients with cancer who receive RT being less than half of that in Western countries (61,62). Evidence on the efficacy of preoperative RT in Japan is extremely limited (61,62).

CRT before TME is currently used as the standard therapy for patients with stage II/III rectal cancer. This strategy is used to shrink the volume of primary or metastatic tumors, which downgrades the tumor stage and decreases the loco-regional recurrence and distant metastasis rates (48). Patients who undergo CRT before surgery exhibit better physical health and therapy tolerance than those who receive CRT after surgery (38). In patients with unresectable LARC, CRT is the only standard therapy until the patients are eligible for surgery (63,64).

Additionally, other treatment strategies have been developed for special cases of rectal cancer. The 'watch and wait' strategy has been used for patients with rectal cancer who achieved a complete clinical response after nCRT without surgery; the advantages of this approach include the reduction in the local recurrence rate, which was similar to that of surgical resection, and the preservation of organ (65). The toxicities associated with RT and the ineligibility of some patients to undergo RT due to their tumor location have led to the development of neoadjuvant chemotherapy without radiation as an alternative therapeutic strategy for patients with rectal cancer (66). However, this treatment paradigm remains under consideration and is not yet recommended for use according to the NCCN guidelines (47), which recommend total neoadjuvant therapy, involving the combination of both chemotherapy and CRT before surgery, for patients with rectal cancer. This therapeutic strategy downgrades the tumor stage and improves the overall survival rate (67,68). Moreover, patients who receive total neoadjuvant therapy are eligible for the 'watch and wait' therapeutic strategy (69). Thus, this approach provides a viable treatment strategy for patients with rectal cancer as per the NCCN guidelines.

4. Limitations of RT for rectal cancer

The advantages of the therapeutic combination of preoperative RT and surgery for patients with LARC have been well documented in the last few decades. Preoperative RT markedly improves the outcome of patients with rectal cancer (70). However, the radiation-induced adverse effects and the development of radioresistance in the tumors have been the major limitations for the clinical application of this strategy (71). Radiation-induced toxicities are classified as acute and late toxicity based on the time of symptom onset (72). The symptoms of acute toxicities appear during or right after treatment (73). By contrast, the symptoms of late toxicity appear after prolonged treatment ranging from several months to years (73). The symptoms of acute toxicities include skin erythema, fatigue, nausea, diarrhea and neurological pain, as well as wound healing complications for patients undergoing surgical resection (74,75). Gastrointestinal dysfunction, urinary dysfunction, permanent neurological toxicities and increased risk of secondary cancers are the major complications associated with late toxicities (76).

In a TME trial, the combination of SC-RT and TME markedly decreased the recurrence rates of LARC from 30-50% to <15% (78) and improved the 10-year survival rate in patients with advanced rectal cancer compared with surgery alone (78). However, SC-RT markedly increased the acute toxicity-associated symptoms, such as lumbosacral plexopathy, stiff pain in the back and legs during RT, or chronic neurological pain 75). Additionally, a series of long-term follow-up randomized studies of Stockholm trials (79), Swedish rectal cancer trial (80) and Dutch TME trial (81) indicated that the frequency of late toxicity-associated symptoms, such as gastrointestinal dysfunctions with impaired bowel movements and fecal incontinence, was higher and more severe in patients who underwent RT compared with that in patients who under-went surgery alone (80,82). In particular, bowel dysfunction was observed in ~62% of patients who underwent RT, but in only 38% of patients who underwent TME alone in the Dutch TME trial (81). Daily activities were significantly decreased and sexual activity was impaired in patients who underwent preoperative SC-RT (83). Although the aforementioned Dutch TME study (81) suggested the negative effects of RT on the quality of life and sexual function, patients who underwent RT exhibited downgraded tumor stages compared with those who underwent only surgery. Moreover, a follow-up study based on the Uppsala trial revealed that the risk of secondary cancer in patients who underwent RT was two times higher than that in patients who did not undergo RT (84). RT-associated secondary pelvic cancers mostly appeared adjacent to other organs, such as the colon, prostate, bladder and ureter, in patients who underwent RT (77).

In addition to radiation-induced toxicities, resistance to preoperative RT is a major clinical obstacle for treating LARC. The biological complexities and heterogeneities of tumors, and the presence of cancer stem cells or accumulated mutations in the tumors (85,86) render the tumors radioresistant. Radioresistance is associated with tumor recurrence, metastasis and poor prognosis, which affect the therapeutic outcomes (87,88). Although a pathological complete response is obtained in numerous cases, several cases are resistant to RT, which is the main cause of rectal cancer-associated death (89,90). The pathological response to preoperative RT varies markedly among patients with LARC. Approximately 8-20% of patients with LARC respond to preoperative RT (91,92). However, >40% of patients have a partial response and ~20% of patients are resistant to preoperative RT (93). These different responses to preoperative RT are directly associated with local recurrence, distant metastasis and overall survival of patients with rectal cancer.

RT is an expensive and time-consuming procedure that is associated with a high risk of perioperative morbidity. Thus, the prediction of the response to RT has direct implications on the clinical decisions regarding treatment strategy. Patients who are sensitive to CRT must undergo a manual 'wait and see' procedure or transanal local excision treatment therapy (94,95). Conversely, patients with poor response to CRT require an alternative therapeutic strategy. The accurate prediction of the pathological and clinical response of patients with rectal cancer to RT may allow clinicians to select the optimal therapeutic strategy for each case and improve the clinical outcome of RT.

5. ncRNAs as predictive biomarkers for RT response in patients with rectal cancer

The response in patients with rectal cancer to preoperative CRT is a critical predictor of local recurrence and patient survival. However, there are no methods to predict the response to RT. Previous studies have reported that ncRNAs, which are key regulators of multiple biological signaling path-ways, are involved in tumorigenesis and in the regulation of tumor sensitivity to RT (5,96,97). Aberrant ncRNA expression induces radioresistance by modulating the signaling pathways associated with cell cycle regulation, DNA repair, cell proliferation/apoptosis, hypoxia or self-renewal, and differentiation of cancer stem cells (98). Since ncRNAs are not digested by RNases, they are stable in fluids, such as blood, saliva and urine (99). Therefore, ncRNAs can be potential predictors of RT response in patients with cancer. However, there are limited studies that have examined the potential of ncRNA profiles as predictors of RT response in patients with rectal cancer. The following section will summarize the published studies on the ability of various ncRNAs in predicting the RT response of patients with rectal cancer (Table I).

Table I

Association between specific ncRNAs and response to therapy in studies of patients with rectal cancer.

Table I

Association between specific ncRNAs and response to therapy in studies of patients with rectal cancer.

A, lncRNA
First author, yearSpecimenPatients in T set, nPatients in V set, nTreatment dose (RT/CRT)pCR rate, %ncRNA platformDifferentially expressed ncRNAs, nIdentified ncRNAs and outcome(Refs.)
Li et al, 2019Frozen tumor tissues6NoNot specifiedNot specifiedlncRNA array2823 lncRNAs: PRDM11, RPB10 and CWC15. Aberrant expression between responders and non-responders(100)
Ferrando et al, 2020Frozen tumor tissues30No50.4 Gy in 28 fractions with capecitabine40lncRNA microarray112 upregulated lncRNAs (LINC00324 and lncKLF7-1) and 1 downregulated lncRNA (lncMAB21L2-1) in responders. Extremely high overall sensitivity and specificity: 91 and 94%, respectively(103)

B, Individual miRNAs
First author, yearSpecimenPatients in T set, nPatients in V set, nTreatment dose (RT/CRT)pCR rate, %ncRNA platformDifferentially expressed ncRNAs, nIdentified ncRNAs and outcome(Refs.)

Drebber et al, 2011FFPE tissues40No50.4 Gy with 5-FU36.6RT-qPCR3Low expression levels of intratumoral miR-145 post-treatment associated with worse response to neoadjuvant CRT(107)
Lopes-Ramos et al, 2014Tumor biopsies271650.4-54 Gy with 5-FUT set: 25.9 V set: 43.8miRNA sequencing4Upregulated miR-21 expression was associated with complete responders. Predictive sensitivity and specificity were 78 and 86%, respectively(110)
Caramés et al, 2015FFPE tissues92NoRadiation dose unspecified with 5-FU/capecitabine13.2RT-qPCRHigh miR-21 expression associated with complete pathological response and good outcome. Positive and negative predictive values were 92 and 42.8%, respectively(111)
Svoboda et al, 2008Tumor biopsiesBR:35
AR:31
No45-50.4 Gy with capecitabine58.1RT-qPCR8Upregulation of miR125b and miR137 expression in post-treatment patients was associated with worse outcome(112)
D'Angelo et al, 2016Tumor biopsies and serum samples38
34
No45 Gy with 5-FU/capecitabine with or without oxaliplatin47.4miRNA microarray11High miR-125b expression in both tumor tissues and serum was associated with a poor response in patients with rectal cancer(113)
Yu et al, 2016Tumor biopsies and serum samples20
87
20
42
50.4-54 Gy50
38
miRNA microarray16High miR-345 expression was significantly associated with a poor response to neoadjuvant CRT and poor locoregional control in patients with rectal cancer(114)
Caramés et al, 2016FFPE tissues78NoRadiation dose unspecified with 5-FU12.2RT-qPCROverexpression of miR-31 significantly predicted a poor pathological response and worse overall survival(115)
D'Angelo et al, 2018Tumor biopsies38Not specified21miRNA microarrayHigh miR-194 expression was closely associated with radiation- sensitive patients(119)

C, miRNA signatures
First author, yearSpecimenPatients in T set, nPatients in V set, nTreatment dose (RT/CRT)pCR rate, %ncRNA platformDifferentially expressed ncRNAs, nIdentified ncRNAs and outcome(Refs.)

Della Vittoria Scarpati et al, 2012Frozen tumor biopsies35No45 Gy with capecitabine- oxaliplatin21.6miRNA microarray5311 miRNAs were upregulated (miR-1183, miR-622, miR-483-5p, miR-125a-3p, miR-1224-5p, miR-1471miR-188-5p, miR-671-5p, miR-1909*, miR-630 and miR-765) and 2 miRNAs were downregulated (miR-1274b and miR-720) in complete responders. Predictive sensitivity and specificity were significant(122)
Hotchi et al, 2013Tumor biopsies222140 Gy in 22 fractions with S-1, a novel oral fluoropyrimidineT set: 31.8 V set: 42.8miRNA microarrayThe response to CRT was evaluated using three parameters: i) in responders by histopathological examination (2 upregulated miRNAs, miR-223 and miR-142-3p); ii) in responders by RECIST (1 upregulated miRNA, miR-223, and 8 downregulated miRNAs, miR-20b, miR-92a, let-7a*, miR-20a, miR-17*, miR-106a, miR-17 and miR-20a); iii) in responders by downstaging (3 upregulated miRNAs, miR-223, miR-630 and miR-126*)(123)
Azizian et al, 2016Tumor biopsies4514750.4 Gy in 28 fractions with capecitabine/oxaliplatinNot specifiedmiRNA microarray19High expression of four-miRNA signature (miR-515-5p, miR-573, miR-579 and miR-802) was associated with a poor outcome(127)
Campayo et al, 2018Tumor biopsies129645-50 Gy with 5-FUV set: 25miRNA microarray377Low expression of three-miRNA signature (miR-21, miR-99b and miR-375) was positively associated with complete responders. Predictive sensitivity and specificity were 60 and 82.9%, respectively(129)
Machackova et al, 2020Tumor biopsies4087Radiation dose unspecified with 5-FUV set: 17Small RNA sequencing69High expression of five-miRNA signature (miR-324-3p, miR-15b-5p, miR-146a-5p, miR-193a-5p and miR- 487a-3p) was positively associated with non-responders. miR-487a-3p was considered as the best candidate among 5 selected miRNAs(130)
Li et al, 2018Peripheral blood samplesBR: 38
AR: 31
BR: 24
AR: 24
(2 Gy/day) from 3-6 monthsNot specifiedqPCR array22The three-miRNA signature with higher levels of miR-374-5p and lower levels of miR-342-5p and miR-519d-3p in plasma was associated with a worse prognosis(131)

[i] ncRNA, non-coding RNA; lncRNA, long ncRNA; miR/miRNA, microRNA; T set, training set; V set, validation set; FFPE, formalin-fixed paraffin-embedded; BR, before radiation; AR, after radiation; RT, radiotherapy; CRT, chemoradiotherapy; pCR, pathological complete response; 5-FU, 5-fluorouracil; RT-qPCR, reverse transcription-quantitative PCR.

lncRNAs

Some studies have examined the expression profile of lncRNAs in patients with rectal cancer who have undergone preoperative RT. Li et al (100) examined six patients with LARC who had undergone preoperative CRT and demonstrated that the lncRNA-miRNA-mRNA regulatory network was associated with the nCRT response. This analysis revealed that 282 lncRNAs were differentially expressed between the two groups (100). Previous studies have reported that lncRNAs may regulate gene expression through direct interaction with the target gene or interaction with miRNAs (101,102). Therefore, lncRNAs and their targets that were differentially expressed between the two groups were identified by constructing the lncRNA-miRNA-mRNA regulatory network with lncRNAs as key players, and the interaction pairs of lncRNA-miRNA and miRNA-mRNA were extracted (100). Specific lncRNAs (PRDM11, RPB10 and CWC15) were positively associated with the response to nCRT (100).

A comprehensive analysis of lncRNA profile was also performed using lncRNA-specific microarrays (103). Among the 8,127 lncRNAs represented in the microarrays, 11 were differentially expressed (two-fold change in expression) between responders and non-responders (103). Of these 11 lncRNAs, five (LINC00261, lncKIF3A-1, LINC00324, lncKLF7-1 and LINC00511) were upregulated and six (lncWAPAL-1, lncFGF10-3, lncMAB21L2-1, lncGALC-6, lncZNF-366-6 and lncHDAC2-2) were downregulated in the responders (103). Additionally, these differentially expressed lncRNAs were associated with the molecular mechanism underlying tumor progression and resistance to therapeutics (104-106).

Although the comprehensive analysis identified differentially expressed lncRNAs in patients with rectal cancer, the aforementioned studies had several limitations, such as the small size of the study cohort and the lack of functional analyses of lncRNA signatures. Therefore, these differentially expressed lncRNAs must be validated and further analyzed in vitro and in vivo to determine their ability to predict the response to CRT in patients with LARC.

Individual miRNAs

A study examining the ability of miRNAs to predict the nCRT response in patients with rectal cancer reported that miR-145 expression in the tumor tissues after therapy was upregulated compared with that in the tumor tissues before therapy. Therefore, the downregulated expression of intratumoral miRNA-145 post-treatment was positively associated with a poor response to neoadjuvant therapy in patients with rectal cancer (107). miR-21 is one of the most promising predictors of response to RT in different types of cancer, such as nasopharyngeal carcinoma and non-small-cell lung cancer (108,109). Several clinical trials have examined the roles of miR-21 in patients with rectal cancer undergoing CRT. Sequencing analysis of 27 rectal tumor biopsies before CRT revealed that miR-21 was a predictive biomarker in complete responders and that the sensitivity and specificity of miR-21 to predict therapeutic response in responders were 78 and 86%, respectively (110). Caramés et al (111) examined 92 patients with LARC undergoing CRT to investigate the roles of miR-21 in clinical and molecular characteristics of the tumor, pathological response and clinical outcome. The aforementioned study indicated that the preoperative expression levels of miR-21 could distinguish the responders from the non-responders with 92% positive predictive value and 42.8% negative predictive value (111).

In addition to miR-21, there are other miRNAs that can predict the response to therapy in patients with rectal cancer undergoing CRT. Svoboda et al (112) observed 35 patients with LARC undergoing CRT and identified that several miRNAs (miR-10a, miR-21, miR-145, miR-212, miR-339 and miR-361) were differentially expressed after RT. Among these, miR-125 and miR-137 were markedly upregulated after the initiation of therapy in most samples (112). The upregulated expression levels of both miR-125 and miR-137 have been associated with a poor response to neoadjuvant therapy (112). Additionally, serum miR-125 levels in the non-responders are significantly upregulated compared with those in the responders, and are associated with an excellent discriminating power (112,113). Additionally, the upregulated expression levels of miR-345 in both tissues and serum have been significantly associated with a poor response to neoadjuvant CRT (114).

Caramés et al (115) also examined 78 patients diagnosed with LARC who were undergoing nCRT and demonstrated that miR-31 may be used as a novel predictive marker for both pathological response and clinical outcome of the patients. miR-31, which was markedly upregulated in 32.4% of cases, was positively associated with a lack of pathological response and worse overall survival (115). By contrast, downregulated miR-31 expression was positively associated with improved responses to neoadjuvant therapy (115). Previous studies reported that miR-194, which is frequently downregulated in colorectal cancer (116,117), is a potential tumor suppressor (117,118). Therefore, D'Angelo et al (119) investigated the potential of miR-194 to predict the response to CRT using biopsy samples collected from 38 patients diagnosed with LARC. Patients with radiosensitive LARC were associated with upregulated miR-194 expression (119). These findings suggest that miR-194 may serve as a radiosensitive marker in patients with rectal cancer. Additionally, the results of the aforementioned studies support the hypothesis that CRT may modulate the miRNA expression profiles in both tissue and serum samples. Thus, the expression levels of miRNAs have the potential to predict tumor response to treatment and may be used as potential prognostic biomarkers.

miRNA signature

A group of miRNAs can be a reliable biomarker that can accurately predict tumor response to RT. Most studies have included pathological staging analysis before and after preoperative CRT as the tumor stage is associated with prognosis and performed large-scale analysis of the expression levels of several miRNAs (120,121). Previous miRNA expression profile analysis has revealed the differential expression patterns of miRNA signatures between responders and non-responders after preoperative CRT (122,123). Della Vittoria Scarpati et al (122) examined the changes in the expression levels of selected miRNAs in rectal cancer biopsies from 38 patients diagnosed with T3-4/N+ rectal cancer who were treated with capecitabine-oxaliplatin and underwent RT followed by surgery. Microarray was performed to analyze the expression of 373 miRNAs in frozen biopsies obtained before treatment (122). In total, 13 differentially expressed miRNAs were identified between good responders and non-responders (122). Of these 13 miRNAs, 11 (miR-1183, miR-483-5p, miR-622, miR-125a-3p, miR-1224-5p, miR-188-5p, miR-1471, miR-671-5p, miR-1909*, miR-630 and miR-765) were significantly upregulated in good responders, while 2 (miR-1274b and miR-720) were downregulated (122). The specificity and sensitivity of differentially expressed miRNAs indicated that they could predict complete response cases, especially two miRNAs (miR-622 and miR-630) that are involved in DNA repair mechanisms (122).

To identify the expression pattern of miRNAs to predict the response of rectal cancer to CRT, Hotchi et al (123) analyzed 43 patients with rectal cancer before preoperative CRT using miRNA microarray. The response to CRT was evaluated based on the following three parameters: Histopathological exami-nation of surgically resected specimens, response evaluation criteria in solid tumors (RECIST) and downstaging (123). Histopathological examination of surgically resected specimens revealed that the expression levels of two miRNAs (miR-223 and miR-142-3p) in the responders were significantly higher than those in the non-responders (123). Based on the RECIST parameter, nine miRNAs were identified as predictive biomarkers of RT response (123). Compared with that in the non-responders, the expression of one miRNA (miR-223) was upregulated, while that of eight miRNAs (miR-20b, miR-92a, let-7a*, miR-20a, miR-17*, miR-106a, miR-17 and miR-20a) was downregulated in the responders (123). The downstaging evaluation revealed that the expression levels of three miRNAs (miR-223, miR-630 and miR-126*) were associated with response to RT (123). These results indicated that the upregulated expression levels of these miRNA signatures had the potential to predict the response of patients with rectal cancer to CRT with high sensitivity and specificity (123).

The ability of miRNA signatures to predict the response to treatment has piqued the interest of the scientific community. Most studies involve a small number of samples with no validation group (123-125). The investigations involving small cohorts are associated with a high risk of false-positive cases, which has hindered or delayed the clinical application of diagnostic and prognostic biomarkers (126). To overcome these limitations, Azizian et al (127) performed microarray analysis of a training set comprising 45 preoperative biopsies from patients with rectal cancer to identify potential miRNAs that can predict tumor regression grade and other clinical parameters. The selected miRNAs were then validated using reverse transcription-quantitative PCR (RT-qPCR) with an independent set of 147 patients with rectal cancer (127). The upregulated expression levels of four miRNAs (miR-515-5p, miR-573, miR-579 and miR-802) were significantly associated with overall survival and cancer-specific survival in patients with rectal cancer (123,128). The miRNA signature comprising the four aforementioned miRNAs may predict the treatment response before RT in patients with rectal cancer.

Campayo et al (129) analyzed the miRNA expression levels in a training group comprising 12 selected patients (six responders and six non-responders) and a validation set comprising 96 pretreatment biopsies from patients with rectal cancer. Among the 377 miRNAs, eight (let-7b, let-7e, miR-21, miR-99b, miR-183, miR-328, miR-375 and miR-483-5p) could predict the response to CRT (129). Of these eight miRNAs, three (miR-21, miR-99b and miR-375) could predict the response to CRT in the cohort comprising 96 patients (129). The downregulated expression levels of miR-21, miR-99b and miR-375 could distinguish the responders from the non-responders (129). Recently, a large-scale miRNA analysis of 40 tumor biopsy samples using small RNA sequencing revealed that five miRNAs (miR-324-3p, miR-15b-5p, miR-146a-5p, miR-193a-5p and miR-487a-3p) were differen-tially expressed between responders and non-responders (130); among the selected miRNAs, miR-487a-3p expression in the non-responders was significantly upregulated compared with that in the responders and had the highest potential to predict CRT response (130).

miRNA signature in the plasma may also potentially predict tumor response to CRT. Li et al (131) screened the miRNA profile of plasma samples derived from 26 patients with rectal cancer before and after RT. The selected miRNAs in the testing and training groups were validated using RT-qPCR (131). A group of three miRNAs comprising miR-374a-5p, miR-342-5p and miR-519d-3p emerged as potential predictive markers for distinguishing the responders from the non-responders before RT (131). In particular, the downregulated expression levels of miR-342-5p and miR-519d-3p in the plasma were associated with a worse prognosis and a short 5-year survival in patients with rectal cancer (131). These results demonstrate the clinical importance of both tissue and blood miRNA signatures in predicting the clinical outcomes of rectal cancer and emphasize the necessity for validation studies in a clinical setting.

From the aforementioned studies, it is clear that miRNAs are becoming involved in the response to RT. They are not only shown as a predictor of RT to select responding or non-responding patients, but also used to predict the survival and post-radiation toxicity. Although emerging evidence has indicated promising roles of miRNAs as valuable clinical tools for personalized care of rectal cancer in the future, any potential biomarker requires multiple and large cohorts to validate its reproducibility. Therefore, statistically rigorous independent validation studies in different cohorts and different types of cancer are critical to confirm the prognostic or predictive value of these emerging findings.

6. ncRNAs are promising targets to enhance the radiosensitivity of rectal cancer

Previous preliminary studies have demonstrated that ncRNAs can be potential therapeutic targets for cancer (132,133). ncRNAs can function as oncogenes or tumor suppressors and are involved in cancer initiation, progression and resistance to therapeutics (134-136). Hence, various strategies have been developed to utilize ncRNAs as therapeutic targets in cancer. Recent targeting strategies consist of the application of ncRNA inhibitors, such as ncRNA mimics, ncRNA vectors, ncRNA sponges, antisense oligonucleotides, ncRNA-mask oligonucleotides or clustered regularly interspaced short palindromic repeat-Cas9 technology (137). The modulation of ncRNA expression levels may be also achieved by transferring ncRNAs using various delivery systems. Currently, different strategies have been used to deliver ncRNAs, such as viral-based vector systems (lentiviruses, retroviruses, adenoviruses or adeno-associated viruses) and non-viral delivery systems (physical and chemical approaches) (138,139). The advantages of non-viral systems include decreased toxicity, low immunogenicity and the ability to transfer large-sized passenger ncRNAs (139). The viral-based vector systems are associated with high transfection efficiency, long half-life and high biodegradability (140). Therefore, designing a precise delivery system to ensure the stability of synthetic ncRNAs in circulation, a high transfection efficiency and tumor-specific targeting is indispensable to translate ncRNA therapy to the clinic. The modulation of ncRNAs or ncRNA-regulated signaling pathways can be a potential therapeutic strategy to enhance the radiosensitivity of rectal cancer (Table II).

Table II

ncRNAs as potential therapeutic targets in rectal cancer.

Table II

ncRNAs as potential therapeutic targets in rectal cancer.

A, miRNAs
First author, yearncRNAsTargetEffect(Refs.)
Lopes-Ramos et al, 2014miR-21-5pSATB1Modulating the malignant behavior and multidrug resistance(110)
Ha Thi et al, 2019miR-130aSOX4Reducing the irradiation-induced cell invasion and damaged DNA repair(144)
Ruhl et al, 2018miR-451EMSY, CAB39Inhibiting cell proliferation and DNA damage repair(146)
Luo et al, 2018miR-519b-3pARID4BInhibiting cell proliferation, migration and invasion(147)

B, lncRNAs
First author, yearncRNAsTargetEffect(Refs.)

Yang et al, 2017lincRNA-RORmiR-145Inducing radiation-mediated cell death(149)
Zou et al, 2018lncRNA OIP5-AS1miR-363-3pInducing radiation-mediated cell death(150)
Wang et al, 2014lincRNA-p21Wnt/β-catenin pathwayInducing radiation-mediated apoptosis(151)

[i] ncRNA, non-coding RNA; lncRNA, long ncRNA; miR/miRNA, microRNA.

Lopes-Ramos et al (110) reported that miR-21-5p, which was upregulated in complete responders of RT, may be a promising therapeutic candidate for rectal cancer. Upregulated miR-21-5p expression in rectal cancer cells (compared with that in control cells) increased the sensitivity of cancer cells to CRT (110). Conversely, the inhibition of miR-21-5p decreased the sensitivity of rectal cancer cells to CRT (110). Mechanistically, miR-21-5p negatively regulated the mRNA expression levels of SATB1, which is reported to induce malignant behavior and confer multidrug resistance (110). Additionally, miR-21-5p expression was positively associated with a poor prognosis in several types of cancer, including rectal cancer (110,141). miR-205 and miR-95 enhance the sensitivity of tumors to RT by targeting specific mRNAs, such as ZEB1, Ubc13 or SGPP1 (142,143). Recently, Ha Thi et al (144) have identified miR-130a as a promising candidate to modulate the radioresistance of rectal cancer. Mechanistically, miR-130a suppressed the repair pathway of irradiation-induced DNA damage by directly targeting SOX4, which encodes a transcription factor (144). Additionally, upregulated miR-451a expression enhanced the radiosensitivity of rectal cancer by modulating the mRNA expression levels of EMSY and CAB39, which are involved in cell proliferation and DNA damage repair (145,146), and are positively correlated with the upregulation of the SHP2 and RAF signaling pathways (145). Therefore, pretreatment with SHP2 and RAF inhibitors may benefit patients with rectal cancer who have a limited response to CRT and exhibit low miR-451a expression. Additionally, miR-519b-3p was reported to be a promising candidate to over-come the radioresistance of rectal cancer (147). miR-519b-3p expression was upregulated in good responders among patients with LARC and was inversely correlated with the mRNA expression levels of ARID4B, which is associated with tumor growth, migration and invasion (148).

Several lncRNAs can modulate radiosensitivity through multiple mechanisms in rectal cancer. The suppression of lincRNA-ROR sensitizes rectal cancer cells by inducing cell death after irradiation (149). lincRNA-ROR regulates radiosensitivity by promoting the expression levels of p53 and miR-145, and consequently modulating the expression levels of p21 and Myc (149). lncRNA OIP5-AS1 inhibits miR-363-3p expression and promotes DYRK1A expression (150). Ectopic expression of lncRNA OIP5-AS1 promotes radiation-induced cell death and enhances the radiosensitivity of rectal cancer by targeting DYRK1A (150). Additionally, lincRNA-p21 promotes the radiosensitivity of rectal cancer by targeting the Wnt/β-catenin signaling pathway (151), and lincRNA-p21 overexpression enhances radiation-induced apoptosis by inducing Noxa expression, a pro-apoptotic gene (151).

7. Conclusion

Previous studies have demonstrated that ncRNAs mediate tumor response against radiation. Several ncRNAs may enhance tumor radiosensitivity, while others confer resistance to radiation by modulating the irradiation-associated signaling pathways. Additionally, experimental studies have demonstrated that the modulation of ncRNA expression levels may markedly enhance the radiosensitivity of rectal cancer cells in vitro and in vivo. These findings suggest that ncRNAs may serve as potential therapeutic targets to improve the outcome of RT. Additionally, ncRNAs can be targeted in combination therapy for cancer.

The ncRNA-based therapies for cancer have yielded promising results in preclinical studies. However, the implementation of these techniques in a clinical setting is associated with several challenges. The expression levels of ncRNAs vary among different types of human cancer and even among the same tumor type in different studies. The tumor microenvironment serves an essential role in regulating tumor biological behavior and may hinder the quantification of ncRNA expression. Tumor progression during RT is a dynamic process that is accompanied by dynamic changes in ncRNA expression. To enhance the therapeutic efficacy of ncRNA-based RT, the delivery system of ncRNAs should be modified to increase their stability and specificity. Therefore, further technical and scientific development and clinical investigation are warranted to overcome these challenges.

Funding

The present review was supported by the National Research Foundation of Korea grant funded by the Korean government (grant no. 2020R1F1A1061122).

Availability of data and materials

Not applicable.

Authors' contributions

HTHT and SH were involved in the conceptualization of the study, and confirmed the authenticity of the data. HTHT and HQD wrote the manuscript, and SH edited and revised it. All authors have read and approved the final version of the manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Acknowledgments

Not applicable.

References

1 

Miller KD, Siegel RL, Lin CC, Mariotto AB, Kramer JL, Rowland JH, Stein KD, Alteri R and Jemal A: Cancer treatment and survivorship statistics, 2016. CA Cancer J Clin. 66:271–289. 2016. View Article : Google Scholar : PubMed/NCBI

2 

Giuliani ME, Lindsay PE, Kwan JY, Sun A, Bezjak A, Le LW, Brade A, Cho J, Leighl NB, Shepherd FA and Hope AJ: Correlation of dosimetric and clinical factors with the development of esophagitis and radiation pneumonitis in patients with limited-stage small-cell lung carcinoma. Clin Lung Cancer. 16:216–220. 2015. View Article : Google Scholar

3 

Han S, Gu F, Lin G, Sun X, Wang Y, Wang Z, Lin Q, Weng D, Xu Y and Mao W: Analysis of clinical and dosimetric factors influencing radiation-induced lung injury in patients with lung cancer. J Cancer. 6:1172–1178. 2015. View Article : Google Scholar : PubMed/NCBI

4 

Briere TM, Krafft S, Liao Z and Martel MK: Lung size and the risk of radiation pneumonitis. Int J Radiat Oncol Biol Phys. 94:377–384. 2016. View Article : Google Scholar

5 

Podralska M, Ciesielska S, Kluiver J, van den Berg A, Dzikiewicz-Krawczyk A and Slezak-Prochazka I: Non-coding RNAs in cancer radiosensitivity: MicroRNAs and lncRNAs as regulators of radiation-induced signaling pathways. Cancers (Basel). 12:16622020. View Article : Google Scholar

6 

Dreussi E, Pucciarelli S, De Paoli A, Polesel J, Canzonieri V, Agostini M, Friso ML, Belluco C, Buonadonna A, Lonardi S, et al: Predictive role of microRNA-related genetic polymorphisms in the pathological complete response to neoadjuvant chemoradiotherapy in locally advanced rectal cancer patients. Oncotarget. 7:19781–19793. 2016. View Article : Google Scholar : PubMed/NCBI

7 

Fanale D, Castiglia M, Bazan V and Russo A: Involvement of non-coding RNAs in chemo- and radioresistance of colorectal cancer. Adv Exp Med Biol. 937:207–228. 2016. View Article : Google Scholar : PubMed/NCBI

8 

Bartel DP: MicroRNAs: Genomics, biogenesis, mechanism, and function. Cell. 116:281–297. 2004. View Article : Google Scholar : PubMed/NCBI

9 

Chen CZ: MicroRNAs as oncogenes and tumor suppressors. N Engl J Med. 353:1768–1771. 2005. View Article : Google Scholar : PubMed/NCBI

10 

Tan S, Xia L, Yi P, Han Y, Tang L, Pan Q, Tian Y, Rao S, Oyang L, Liang J, et al: Exosomal miRNAs in tumor microenvironment. J Exp Clin Cancer Res. 39:672020. View Article : Google Scholar : PubMed/NCBI

11 

Patil N, Allgayer H and Leupold JH: MicroRNAs in the tumor microenvironment. Adv Exp Med Biol. 1277:1–31. 2020. View Article : Google Scholar : PubMed/NCBI

12 

Kosaka N, Iguchi H and Ochiya T: Circulating microRNA in body fluid: A new potential biomarker for cancer diagnosis and prognosis. Cancer Sci. 101:2087–2092. 2010. View Article : Google Scholar : PubMed/NCBI

13 

Movahedpour A, Ahmadi N, Ghasemi Y, Savardashtaki A and Shabaninejad Z: Circulating microRNAs as potential diagnostic biomarkers and therapeutic targets in prostate cancer: Current status and future perspectives. J Cell Biochem. 120:16316–16329. 2019. View Article : Google Scholar : PubMed/NCBI

14 

Sohel MH: Extracellular/circulating MicroRNAs: Release mechanisms, functions and challenges. Achiev Life Sci. 10:175–186. 2016.

15 

Mercer TR, Dinger ME and Mattick JS: Long non-coding RNAs: Insights into functions. Nat Rev Genet. 10:155–159. 2009. View Article : Google Scholar : PubMed/NCBI

16 

Fernandes JCR, Acuña SM, Aoki JI, Floeter-Winter LM and Muxel SM: Long non-coding RNAs in the regulation of gene expression: Physiology and disease. Noncoding RNA. 5:172019.

17 

Statello L, Guo CJ, Chen LL and Huarte M: Gene regulation by long non-coding RNAs and its biological functions. Nat Rev Mol Cell Biol. Dec 22–2020.Epub ahead of print. PubMed/NCBI

18 

Spizzo R, Almeida MI, Colombatti A and Calin GA: Long non-coding RNAs and cancer: A new frontier of translational research? Oncogene. 31:4577–4587. 2012. View Article : Google Scholar : PubMed/NCBI

19 

Schmitt AM and Chang HY: Long noncoding RNAs in cancer pathways. Cancer Cell. 29:452–463. 2016. View Article : Google Scholar : PubMed/NCBI

20 

Liu K, Gao L, Ma X, Huang JJ, Chen J, Zeng L, Ashby CR Jr, Zou C and Chen ZS: Long non-coding RNAs regulate drug resistance in cancer. Mol Cancer. 19:542020. View Article : Google Scholar : PubMed/NCBI

21 

Zhu Y, Zhao Y, Dong S, Liu L, Tai L and Xu Y: Systematic identification of dysregulated lncRNAs associated with platinum-based chemotherapy response across 11 cancer types. Genomics. 112:1214–1222. 2020. View Article : Google Scholar

22 

Huang L, Zeng L, Chu J, Xu P, Lv M, Xu J, Wen J, Li W, Wang L, Wu X, et al: Chemoresistance-related long non-coding RNA expression profiles in human breast cancer cells. Mol Med Rep. 18:243–253. 2018.PubMed/NCBI

23 

Arnold M, Sierra MS, Laversanne M, Soerjomataram I, Jemal A and Bray F: Global patterns and trends in colorectal cancer incidence and mortality. Gut. 66:683–691. 2017. View Article : Google Scholar

24 

Siegel RL, Miller KD, Goding Sauer A, Fedewa SA, Butterly LF, Anderson JC, Cercek A, Smith RA and Jemal A: Colorectal cancer statistics, 2020. CA Cancer J Clin. 70:145–164. 2020. View Article : Google Scholar : PubMed/NCBI

25 

Siegel R, Desantis C and Jemal A: Colorectal cancer statistics, 2014. CA Cancer J Clin. 64:104–117. 2014. View Article : Google Scholar : PubMed/NCBI

26 

Lo AC, Soliman AS, Khaled HM, Aboelyazid A and Greenson JK: Lifestyle, occupational, and reproductive factors and risk of colorectal cancer. Dis Colon Rectum. 53:830–837. 2010. View Article : Google Scholar : PubMed/NCBI

27 

Basen-Engquist K and Chang M: Obesity and cancer risk: Recent review and evidence. Curr Oncol Rep. 13:71–76. 2011. View Article : Google Scholar

28 

De Roos AJ, Ray RM, Gao DL, Fitzgibbons ED, Ziding F, Astrakianakis G, Thomas DB and Checkoway H: Colorectal cancer incidence among female textile workers in Shanghai, China: A case-cohort analysis of occupational exposures. Cancer Causes Control. 16:1177–1188. 2005. View Article : Google Scholar : PubMed/NCBI

29 

Oddone E, Modonesi C and Gatta G: Occupational exposures and colorectal cancers: A quantitative overview of epidemiological evidence. World J Gastroenterol. 20:12431–12444. 2014. View Article : Google Scholar : PubMed/NCBI

30 

Rattray NJW, Charkoftaki G, Rattray Z, Hansen JE, Vasiliou V and Johnson CH: Environmental influences in the etiology of colorectal cancer: The premise of metabolomics. Curr Pharmacol Rep. 3:114–125. 2017. View Article : Google Scholar : PubMed/NCBI

31 

Peters RK, Garabrant DH, Yu MC and Mack TM: A case-control study of occupational and dietary factors in colorectal cancer in young men by subsite. Cancer Res. 49:5459–5468. 1989.PubMed/NCBI

32 

Wang Y, Lewis-Michl EL, Hwang SA, Fitzgerald EF and Stark AD: Cancer incidence among a cohort of female farm residents in New York State. Arch Environ Health. 57:561–567. 2002. View Article : Google Scholar

33 

Wesseling C, Antich D, Hogstedt C, Rodríguez AC and Ahlbom A: Geographical differences of cancer incidence in Costa Rica in relation to environmental and occupational pesticide exposure. Int J Epidemiol. 28:365–374. 1999. View Article : Google Scholar : PubMed/NCBI

34 

Rushton L, Hutchings S and Brown T: The burden of cancer at work: Estimation as the first step to prevention. Occup Environ Med. 65:789–800. 2008. View Article : Google Scholar

35 

Toiyama Y and Kusunoki M: Changes in surgical therapies for rectal cancer over the past 100 years: A review. Ann Gastroenterol Surg. 4:331–342. 2020. View Article : Google Scholar : PubMed/NCBI

36 

Knol J and Keller DS: Total mesorectal excision technique-past, present, and future. Clin Colon Rectal Surg. 33:134–143. 2020. View Article : Google Scholar : PubMed/NCBI

37 

Kapiteijn E, Marijnen CA, Nagtegaal ID, Putter H, Steup WH, Wiggers T, Rutten HJ, Pahlman L, Glimelius B, van Krieken JH, et al: Preoperative radiotherapy combined with total mesorectal excision for resectable rectal cancer. N Engl J Med. 345:638–646. 2001. View Article : Google Scholar : PubMed/NCBI

38 

Sauer R, Becker H, Hohenberger W, Rödel C, Wittekind C, Fietkau R, Martus P, Tschmelitsch J, Hager E, Hess CF, et al: Preoperative versus postoperative chemoradiotherapy for rectal cancer. N Engl J Med. 351:1731–1740. 2004. View Article : Google Scholar : PubMed/NCBI

39 

Roeder F, Meldolesi E, Gerum S, Valentini V and Rödel C: Recent advances in (chemo-)radiation therapy for rectal cancer: A comprehensive review. Radiat Oncol. 15:2622020. View Article : Google Scholar : PubMed/NCBI

40 

Heald RJ, Husband EM and Ryall RD: The mesorectum in rectal cancer surgery-the clue to pelvic recurrence? Br J Surg. 69:613–616. 1982. View Article : Google Scholar : PubMed/NCBI

41 

Hermanek P, Hermanek P, Hohenberger W, Klimpfinger M, Köckerling F and Papadopoulos T: The pathological assessment of mesorectal excision: Implications for further treatment and quality management. Int J Colorectal Dis. 18:335–341. 2003. View Article : Google Scholar : PubMed/NCBI

42 

MacFarlane JK, Ryall RD and Heald RJ: Mesorectal excision for rectal cancer. Lancet. 341:457–460. 1993. View Article : Google Scholar : PubMed/NCBI

43 

Havenga K, Enker WE, Norstein J, Moriya Y, Heald RJ, van Houwelingen HC and van de Velde CJ: Improved survival and local control after total mesorectal excision or D3 lymph-adenectomy in the treatment of primary rectal cancer: An international analysis of 1411 patients. Eur J Surg Oncol. 25:368–374. 1999. View Article : Google Scholar : PubMed/NCBI

44 

Randomised trial of surgery alone versus radiotherapy followed by surgery for potentially operable locally advanced rectal cancer. Medical research council rectal cancer working party Lancet. 348:1605–1610. 1996.

45 

Kim SH, Lee JM, Hong SH, Kim GH, Lee JY, Han JK and Choi BI: Locally advanced rectal cancer: Added value of diffusion-weighted MR imaging in the evaluation of tumor response to neoadjuvant chemo- and radiation therapy. Radiology. 253:116–125. 2009. View Article : Google Scholar : PubMed/NCBI

46 

Christou N, Meyer J, Toso C, Ris F and Buchs NC: Lateral lymph node dissection for low rectal cancer: Is it necessary? World J Gastroenterol. 25:4294–4299. 2019. View Article : Google Scholar : PubMed/NCBI

47 

Benson AB, Venook AP, Al-Hawary MM, Arain MA, Chen YJ, Ciombor KK, Cohen S, Cooper HS, Deming D, Garrido-Laguna I, et al: NCCN Guidelines insights: Rectal cancer, version 6.2020. J Natl Compr Canc Netw. 18:806–815. 2020. View Article : Google Scholar : PubMed/NCBI

48 

Zhang S, Bai W, Tong X, Bu P, Xu J and Xi Y: Correlation between tumor microenvironment-associated factors and the efficacy and prognosis of neoadjuvant therapy for rectal cancer. Oncol Lett. 17:1062–1070. 2019.PubMed/NCBI

49 

Ma B, Gao P, Song Y, Huang X, Wang H, Xu Q, Zhao S and Wang Z: Short-course radiotherapy in neoadjuvant treatment for rectal cancer: A systematic review and meta-analysis. Clin Colorectal Cancer. 17:320–330.e5. 2018. View Article : Google Scholar : PubMed/NCBI

50 

Bujko K, Wyrwicz L, Rutkowski A, Malinowska M, Pietrzak L, Kryński J, Michalski W, Olędzki J, Kuśnierz J, Zając L, et al: Long-course oxaliplatin-based preoperative chemoradiation versus 5×5 Gy and consolidation chemotherapy for cT4 or fixed cT3 rectal cancer: Results of a randomized phase III study. Ann Oncol. 27:834–842. 2016. View Article : Google Scholar : PubMed/NCBI

51 

From the American Association of Neurological Surgeons (AANS); American Society of Neuroradiology (ASNR); Canadian Interventional Radiology Association (CIRA); Congress of Neurological Surgeons (CNS); European Society of Minimally Invasive Neurological Therapy (ESMINT); European Society of Neuroradiology (ESNR); European Stroke Organization (ESO); Society for Cardiovascular Angiography; Interventions (SCAI); Society of Interventional Radiology (SIR); Society of Interventional Radiology (SIR); et al: Multisociety consensus quality improvement revised consensus statement for endovascular therapy of acute ischemic stroke. Int J Stroke. 13:612–632. 2018.PubMed/NCBI

52 

Ngan SY, Burmeister B, Fisher RJ, Solomon M, Goldstein D, Joseph D, Ackland SP, Schache D, McClure B, McLachlan SA, et al: Randomized trial of short-course radiotherapy versus long-course chemoradiation comparing rates of local recurrence in patients with T3 rectal cancer: Trans-Tasman radiation oncology group trial 01.04. J Clin Oncol. 30:3827–3833. 2012. View Article : Google Scholar : PubMed/NCBI

53 

McLachlan SA, Fisher RJ, Zalcberg J, Solomon M, Burmeister B, Goldstein D, Leong T, Ackland SP, McKendrick J, McClure B, et al: The impact on health-related quality of life in the first 12 months: A randomised comparison of preoperative short-course radiation versus long-course chemoradiation for T3 rectal cancer (Trans-Tasman Radiation Oncology Group Trial 01.04). Eur J Cancer. 55:15–26. 2016. View Article : Google Scholar : PubMed/NCBI

54 

Mowery YM, Salama JK, Zafar SY, Moore HG, Willett CG, Czito BG, Hopkins MB and Palta M: Neoadjuvant long-course chemoradiation remains strongly favored over short-course radiotherapy by radiation oncologists in the United States. Cancer. 123:1434–1441. 2017. View Article : Google Scholar

55 

Glynne-Jones R, Wyrwicz L, Tiret E, Brown G, Rödel C, Cervantes A and Arnold D; ESMO Guidelines Committee: Rectal cancer: ESMO clinical practice guidelines for diagnosis, treatment and follow-up. Ann Oncol. 29(Suppl 4): iv2632018. View Article : Google Scholar : PubMed/NCBI

56 

Sineshaw HM, Jemal A, Thomas CR Jr and Mitin T: Changes in treatment patterns for patients with locally advanced rectal cancer in the United States over the past decade: An analysis from the national cancer data base. Cancer. 122:1996–2003. 2016. View Article : Google Scholar : PubMed/NCBI

57 

Wang X, Zheng B, Lu X, Bai R, Feng L, Wang Q, Zhao Y and He S: Preoperative short-course radiotherapy and long-course radiochemotherapy for locally advanced rectal cancer: Meta-analysis with trial sequential analysis of long-term survival data. PLoS One. 13:e02001422018. View Article : Google Scholar : PubMed/NCBI

58 

Otero de Pablos J and Mayol J: Controversies in the management of lateral pelvic lymph nodes in patients with advanced rectal cancer: East or west? Front Surg. 6:792019. View Article : Google Scholar

59 

Watanabe T, Muro K, Ajioka Y, Hashiguchi Y, Ito Y, Saito Y, Hamaguchi T, Ishida H, Ishiguro M, Ishihara S, et al: Japanese society for cancer of the colon and rectum (JSCCR) guidelines 2016 for the treatment of colorectal cancer. Int J Clin Oncol. 23:1–34. 2018. View Article : Google Scholar :

60 

Tokuhara K, Ueyama Y, Nakatani K, Yoshioka K and Kon M: Outcomes of neoadjuvant chemoradiotherapy in Japanese locally advanced rectal carcinoma patients. World J Surg Oncol. 14:1362016. View Article : Google Scholar : PubMed/NCBI

61 

Numasaki H, Shibuya H, Nishio M, Ikeda H, Sekiguchi K, Kamikonya N, Koizumi M, Tago M, Ando Y, Tsukamoto N, et al: Japanese structure survey of radiation oncology in 2007 with special reference to designated cancer care hospitals. Strahlenther Onkol. 187:167–174. 2011. View Article : Google Scholar : PubMed/NCBI

62 

Teshima T, Numasaki H, Shibuya H, Nishio M, Ikeda H, Sekiguchi K, Kamikonya N, Koizumi M, Tago M, Ando Y, et al: Japanese structure survey of radiation oncology in 2007 based on institutional stratification of patterns of care study. Int J Radiat Oncol Biol Phys. 78:1483–1493. 2010. View Article : Google Scholar : PubMed/NCBI

63 

Minsky BD, Cohen AM, Enker WE, Sigurdson E and Harrison LB: Radiation therapy for unresectable rectal cancer. Int J Radiat Oncol Biol Phys. 21:1283–1289. 1991. View Article : Google Scholar : PubMed/NCBI

64 

Wang G, Wang W, Jin H, Dong H, Chen W, Li X, Li G and Li L: The effect of primary tumor radiotherapy in patients with unresectable stage IV rectal or rectosigmoid cancer: A propensity score matching analysis for survival. Radiat Oncol. 15:1262020. View Article : Google Scholar : PubMed/NCBI

65 

van der Valk MJM, Hilling DE, Bastiaannet E, Meershoek-Klein Kranenbarg E, Beets GL, Figueiredo NL, Habr-Gama A, Perez RO, Renehan AG and van de Velde CJH: IWWD Consortium: Long-term outcomes of clinical complete responders after neoadjuvant treatment for rectal cancer in the international watch & wait database (IWWD): An international multicentre registry study. Lancet. 391:2537–2545. 2018. View Article : Google Scholar : PubMed/NCBI

66 

Zhao F, Wang J, Yu H, Cheng X, Li X, Zhu X, Xu X, Lin J, Chen X and Yan S: Neoadjuvant radiotherapy improves overall survival for T3/4N+M0 rectal cancer patients: A population-based study of 20300 patients. Radiat Oncol. 15:492020. View Article : Google Scholar : PubMed/NCBI

67 

Petrelli F, Trevisan F, Cabiddu M, Sgroi G, Bruschieri L, Rausa E, Ghidini M and Turati L: Total neoadjuvant therapy in rectal cancer: A systematic review and meta-analysis of treatment outcomes. Ann Surg. 271:440–448. 2020. View Article : Google Scholar

68 

Cercek A, Roxburgh CSD, Strombom P, Smith JJ, Temple LKF, Nash GM, Guillem JG, Paty PB, Yaeger R, Stadler ZK, et al: Adoption of total neoadjuvant therapy for locally advanced rectal cancer. JAMA Oncol. 4:e1800712018. View Article : Google Scholar : PubMed/NCBI

69 

Gilshtein H, Ghuman A, Dawoud M, Yellinek S, Kent I, Sharp SP, Nagarajan A and Wexner SD: Total neoadjuvant treatment for rectal cancer: Preliminary experience. Am Surg. Nov 10–2020.Epub ahead of print. View Article : Google Scholar : PubMed/NCBI

70 

den Dulk M, Krijnen P, Marijnen CA, Rutten HJ, van de Poll-Franse LV, Putter H, Meershoek-Klein Kranenbarg E, Jansen-Landheer ML, Coebergh JW and van de Velde CJ: Improved overall survival for patients with rectal cancer since 1990: The effects of TME surgery and pre-operative radio-therapy. Eur J Cancer. 44:1710–1716. 2008. View Article : Google Scholar : PubMed/NCBI

71 

Birgisson H, Påhlman L, Gunnarsson U and Glimelius B: Late adverse effects of radiation therapy for rectal cancer-a systematic overview. Acta Oncol. 46:504–516. 2007. View Article : Google Scholar

72 

Bruheim K, Guren MG, Skovlund E, Hjermstad MJ, Dahl O, Frykholm G, Carlsen E and Tveit KM: Late side effects and quality of life after radiotherapy for rectal cancer. Int J Radiat Oncol Biol Phys. 76:1005–1011. 2010. View Article : Google Scholar

73 

Joye I and Haustermans K: Early and late toxicity of radiotherapy for rectal cancer. Recent Results Cancer Res. 203:189–201. 2014. View Article : Google Scholar : PubMed/NCBI

74 

Marijnen CA, Kapiteijn E, van de Velde CJ, Martijn H, Steup WH, Wiggers T, Kranenbarg EK and Leer JW; Cooperative Investigators of the Dutch Colorectal Cancer Group: Acute side effects and complications after short-term preoperative radio-therapy combined with total mesorectal excision in primary rectal cancer: Report of a multicenter randomized trial. J Clin Oncol. 20:817–825. 2002. View Article : Google Scholar : PubMed/NCBI

75 

Frykholm GJ, Isacsson U, Nygård K, Montelius A, Jung B, Påhlman L and Glimelius B: Preoperative radiotherapy in rectal carcinoma-aspects of acute adverse effects and radiation technique. Int J Radiat Oncol Biol Phys. 35:1039–1048. 1996. View Article : Google Scholar : PubMed/NCBI

76 

Sipaviciute A, Sileika E, Burneckis A and Dulskas A: Late gastrointestinal toxicity after radiotherapy for rectal cancer: A systematic review. Int J Colorectal Dis. 35:977–983. 2020. View Article : Google Scholar : PubMed/NCBI

77 

Swedish Rectal Cancer Trial; Cedermark B, Dahlberg M, Glimelius B, Påhlman L, Rutqvist LE and Wilking N: Improved survival with preoperative radiotherapy in resectable rectal cancer. N Engl J Med. 336:980–987. 1997. View Article : Google Scholar : PubMed/NCBI

78 

van Gijn W, Marijnen CA, Nagtegaal ID, Kranenbarg EM, Putter H, Wiggers T, Rutten HJ, Påhlman L, Glimelius B and van de Velde CJ; Dutch Colorectal Cancer Group: Preoperative radiotherapy combined with total mesorectal excision for resectable rectal cancer: 12-year follow-up of the multicentre, randomised controlled TME trial. Lancet Oncol. 12:575–582. 2011. View Article : Google Scholar : PubMed/NCBI

79 

Randomized study on preoperative radiotherapy in rectal carcinoma. Stockholm colorectal cancer study group. Ann Surg Oncol. 3:423–430. 1996. View Article : Google Scholar : PubMed/NCBI

80 

Birgisson H, Påhlman L, Gunnarsson U and Glimelius B; Swedish Rectal Cancer Trial Group: Adverse effects of preoperative radiation therapy for rectal cancer: Long-term follow-up of the Swedish rectal cancer trial. J Clin Oncol. 23:8697–8705. 2005. View Article : Google Scholar : PubMed/NCBI

81 

Peeters KC, van de Velde CJ, Leer JW, Martijn H, Junggeburt JM, Kranenbarg EK, Steup WH, Wiggers T, Rutten HJ and Marijnen CA: Late side effects of short-course preoperative radiotherapy combined with total mesorectal excision for rectal cancer: Increased bowel dysfunction in irradiated patients-a Dutch colorectal cancer group study. J Clin Oncol. 23:6199–6206. 2005. View Article : Google Scholar : PubMed/NCBI

82 

Pollack J, Holm T, Cedermark B, Altman D, Holmström B, Glimelius B and Mellgren A: Late adverse effects of short-course preoperative radiotherapy in rectal cancer. Br J Surg. 93:1519–1525. 2006. View Article : Google Scholar : PubMed/NCBI

83 

Marijnen CA, van de Velde CJ, Putter H, van den Brink M, Maas CP, Martijn H, Rutten HJ, Wiggers T, Kranenbarg EK, Leer JW and Stiggelbout AM: Impact of short-term preoperative radiotherapy on health-related quality of life and sexual functioning in primary rectal cancer: Report of a multicenter randomized trial. J Clin Oncol. 23:1847–1858. 2005. View Article : Google Scholar : PubMed/NCBI

84 

Frykholm GJ, Glimelius B and Påhlman L: Preoperative or postoperative irradiation in adenocarcinoma of the rectum: Final treatment results of a randomized trial and an evaluation of late secondary effects. Dis Colon Rectum. 36:564–572. 1993. View Article : Google Scholar : PubMed/NCBI

85 

Shi Y, Liu N, Lai W, Yan B, Chen L, Liu S, Liu S, Wang X, Xiao D, Liu X, et al: Nuclear EGFR-PKM2 axis induces cancer stem cell-like characteristics in irradiation-resistant cells. Cancer Lett. 422:81–93. 2018. View Article : Google Scholar : PubMed/NCBI

86 

Yan B, Liu S, Shi Y, Liu N, Chen L, Wang X, Xiao D, Liu X, Mao C, Jiang Y, et al: Activation of AhR with nuclear IKKα regulates cancer stem-like properties in the occurrence of radio-resistance. Cell Death Dis. 9:4902018. View Article : Google Scholar

87 

Rycaj K and Tang DG: Cancer stem cells and radioresistance. Int J Radiat Biol. 90:615–621. 2014. View Article : Google Scholar : PubMed/NCBI

88 

Li F, Zhou K, Gao L, Zhang B, Li W, Yan W, Song X, Yu H, Wang S, Yu N and Jiang Q: Radiation induces the generation of cancer stem cells: A novel mechanism for cancer radioresistance. Oncol Lett. 12:3059–3065. 2016. View Article : Google Scholar : PubMed/NCBI

89 

Hartley A, Ho KF, McConkey C and Geh JI: Pathological complete response following pre-operative chemoradiotherapy in rectal cancer: Analysis of phase II/III trials. Br J Radiol. 78:934–938. 2005. View Article : Google Scholar : PubMed/NCBI

90 

Sinukumar S, Patil P, Engineer R, Desouza A and Saklani A: Clinical outcome of patients with complete pathological response to neoadjuvant chemoradiotherapy for locally advanced rectal cancers: The Indian scenario. Gastroenterol Res Pract. 2014:8678412014. View Article : Google Scholar

91 

Valentini V, Aristei C, Glimelius B, Minsky BD, Beets-Tan R, Borras JM, Haustermans K, Maingon P, Overgaard J, Pahlman L, et al: Multidisciplinary rectal cancer management: 2nd European rectal cancer consensus conference (EURECA-CC2). Radiother Oncol. 92:148–163. 2009. View Article : Google Scholar : PubMed/NCBI

92 

Gérard JP, Conroy T, Bonnetain F, Bouché O, Chapet O, Closon-Dejardin MT, Untereiner M, Leduc B, Francois E, Maurel J, et al: Preoperative radiotherapy with or without concurrent fluorouracil and leucovorin in T3-4 rectal cancers: Results of FFCD 9203. J Clin Oncol. 24:4620–4625. 2006. View Article : Google Scholar : PubMed/NCBI

93 

Yoon WH, Kim HJ, Kim CH, Joo JK, Kim YJ and Kim HR: Oncologic impact of pathologic response on clinical outcome after preoperative chemoradiotherapy in locally advanced rectal cancer. Ann Surg Treat Res. 88:15–20. 2015. View Article : Google Scholar : PubMed/NCBI

94 

Pozo ME and Fang SH: Watch and wait approach to rectal cancer: A review. World J Gastrointest Surg. 7:306–312. 2015. View Article : Google Scholar : PubMed/NCBI

95 

Renehan AG, Malcomson L, Emsley R, Gollins S, Maw A, Myint AS, Rooney PS, Susnerwala S, Blower A, Saunders MP, et al: Watch-and-wait approach versus surgical resection after chemoradiotherapy for patients with rectal cancer (the OnCoRe project): A propensity-score matched cohort analysis. Lancet Oncol. 17:174–183. 2016. View Article : Google Scholar

96 

Chan N, Milosevic M and Bristow RG: Tumor hypoxia, DNA repair and prostate cancer progression: New targets and new therapies. Future Oncol. 3:329–341. 2007. View Article : Google Scholar : PubMed/NCBI

97 

Anastasiadou E, Jacob LS and Slack FJ: Non-coding RNA networks in cancer. Nat Rev Cancer. 18:5–18. 2018. View Article : Google Scholar

98 

Zhu J, Chen S, Yang B, Mao W, Yang X and Cai J: Molecular mechanisms of lncRNAs in regulating cancer cell radiosensitivity. Biosci Rep. 39:BSR201905902019. View Article : Google Scholar : PubMed/NCBI

99 

Sole C, Arnaiz E, Manterola L, Otaegui D and Lawrie CH: The circulating transcriptome as a source of cancer liquid biopsy biomarkers. Semin Cancer Biol. 58:100–108. 2019. View Article : Google Scholar : PubMed/NCBI

100 

Li N, Yu J, Luo A, Tang Y, Liu W, Wang S, Liu Y, Song Y, Fang H, Chen B, et al: LncRNA and mRNA signatures associated with neoadjuvant chemoradiotherapy downstaging effects in rectal cancer. J Cell Biochem. 120:5207–5217. 2019. View Article : Google Scholar

101 

Liz J and Esteller M: lncRNAs and microRNAs with a role in cancer development. Biochim Biophys Acta. 1859:169–176. 2016. View Article : Google Scholar

102 

Cao MX, Jiang YP, Tang YL and Liang XH: The crosstalk between lncRNA and microRNA in cancer metastasis: Orchestrating the epithelial-mesenchymal plasticity. Oncotarget. 8:12472–12483. 2017. View Article : Google Scholar :

103 

Ferrando L, Cirmena G, Garuti A, Scabini S, Grillo F, Mastracci L, Isnaldi E, Marrone C, Gonella R, Murialdo R, et al: Development of a long non-coding RNA signature for prediction of response to neoadjuvant chemoradiotherapy in locally advanced rectal adenocarcinoma. PLoS One. 15:e02265952020. View Article : Google Scholar : PubMed/NCBI

104 

Wang ZK, Yang L, Wu LL, Mao H, Zhou YH, Zhang PF and Dai GH: Long non-coding RNA LINC00261 sensitizes human colon cancer cells to cisplatin therapy. Braz J Med Biol Res. 51:e67932017. View Article : Google Scholar : PubMed/NCBI

105 

Liao J and Dong LP: Linc00261 suppresses growth and metastasis of non-small cell lung cancer via repressing epithelial-mesenchymal transition. Eur Rev Med Pharmacol Sci. 23:3829–3837. 2019.PubMed/NCBI

106 

Fang Q, Sang L and Du S: Long noncoding RNA LINC00261 regulates endometrial carcinoma progression by modulating miRNA/FOXO1 expression. Cell Biochem Funct. 36:323–330. 2018. View Article : Google Scholar : PubMed/NCBI

107 

Drebber U, Lay M, Wedemeyer I, Vallböhmer D, Bollschweiler E, Brabender J, Mönig SP, Hölscher AH, Dienes HP and Odenthal M: Altered levels of the onco-microRNA 21 and the tumor-supressor microRNAs 143 and 145 in advanced rectal cancer indicate successful neoadjuvant chemoradiotherapy. Int J Oncol. 39:409–415. 2011.PubMed/NCBI

108 

Zhu H, Zhu X, Cheng G, Zhou M and Lou W: Downregulation of microRNA-21 enhances radiosensitivity in nasopharyngeal carcinoma. Exp Ther Med. 9:2185–2189. 2015. View Article : Google Scholar : PubMed/NCBI

109 

Huang Q: Predictive relevance of ncRNAs in non-small-cell lung cancer patients with radiotherapy: A review of the published data. Biomark Med. 12:1149–1159. 2018. View Article : Google Scholar : PubMed/NCBI

110 

Lopes-Ramos CM, Habr-Gama A, Quevedo Bde S, Felício NM, Bettoni F, Koyama FC, Asprino PF, Galante PA, Gama-Rodrigues J, Camargo AA, et al: Overexpression of miR-21-5p as a predictive marker for complete tumor regression to neoadjuvant chemoradiotherapy in rectal cancer patients. BMC Med Genomics. 7:682014. View Article : Google Scholar : PubMed/NCBI

111 

Caramés C, Cristóbal I, Moreno V, del Puerto L, Moreno I, Rodriguez M, Marín JP, Correa AV, Hernández R, Zenzola V, et al: MicroRNA-21 predicts response to preoperative chemoradiotherapy in locally advanced rectal cancer. Int J Colorectal Dis. 30:899–906. 2015. View Article : Google Scholar : PubMed/NCBI

112 

Svoboda M, Izakovicova Holla L, Sefr R, Vrtkova I, Kocakova I, Tichy B and Dvorak J: Micro-RNAs miR125b and miR137 are frequently upregulated in response to capecitabine chemoradiotherapy of rectal cancer. Int J Oncol. 33:541–547. 2008.PubMed/NCBI

113 

D'Angelo E, Fassan M, Maretto I, Pucciarelli S, Zanon C, Digito M, Rugge M, Nitti D and Agostini M: Serum miR-125b is a non-invasive predictive biomarker of the pre-operative chemoradiotherapy responsiveness in patients with rectal adenocarcinoma. Oncotarget. 7:28647–28657. 2016. View Article : Google Scholar : PubMed/NCBI

114 

Yu J, Li N, Wang X, Ren H, Wang W, Wang S, Song Y, Liu Y, Li Y, Zhou X, et al: Circulating serum microRNA-345 correlates with unfavorable pathological response to preoperative chemo-radiotherapy in locally advanced rectal cancer. Oncotarget. 7:64233–64243. 2016. View Article : Google Scholar : PubMed/NCBI

115 

Caramés C, Cristobal I, Moreno V, Marín JP, González-Alonso P, Torrejón B, Minguez P, Leon A, Martín JI, Hernández R, et al: MicroRNA-31 emerges as a predictive biomarker of pathological response and outcome in locally advanced rectal cancer. Int J Mol Sci. 17:8782016. View Article : Google Scholar :

116 

Chiang Y, Song Y, Wang Z, Liu Z, Gao P, Liang J, Zhu J, Xing C and Xu H: microRNA-192, -194 and -215 are frequently down-regulated in colorectal cancer. Exp Ther Med. 3:560–566. 2012. View Article : Google Scholar : PubMed/NCBI

117 

Wang B, Shen ZL, Gao ZD, Zhao G, Wang CY, Yang Y, Zhang JZ, Yan YC, Shen C, Jiang KW, et al: MiR-194, commonly repressed in colorectal cancer, suppresses tumor growth by regulating the MAP4K4/c-Jun/MDM2 signaling pathway. Cell Cycle. 14:1046–1058. 2015. View Article : Google Scholar : PubMed/NCBI

118 

Zhao Y, Li F, Zhang X, Liu A, Qi J, Cui H and Zhao P: MicroRNA-194 acts as a prognostic marker and inhibits proliferation in hepatocellular carcinoma by targeting MAP4K4. Int J Clin Exp Pathol. 8:12446–12454. 2015.

119 

D'Angelo E, Zanon C, Sensi F, Digito M, Rugge M, Fassan M, Scarpa M, Pucciarelli S, Nitti D and Agostini M: miR-194 as predictive biomarker of responsiveness to neoadjuvant chemoradiotherapy in patients with locally advanced rectal adenocarcinoma. J Clin Pathol. 71:344–350. 2018. View Article : Google Scholar

120 

Slotta-Huspenina J, Drecoll E, Feith M, Habermehl D, Combs S, Weichert W, Bettstetter M, Becker K and Langer R: MicroRNA expression profiling for the prediction of resistance to neoadjuvant radiochemotherapy in squamous cell carcinoma of the esophagus. J Transl Med. 16:1092018. View Article : Google Scholar : PubMed/NCBI

121 

Du B, Wang X, Wu D, Wang T, Yang X, Wang J, Shi X, Chen L and Zhang W: MicroRNA expression profiles identify biomarkers for predicting the response to chemoradiotherapy in rectal cancer. Mol Med Rep. 18:1909–1916. 2018.PubMed/NCBI

122 

Della Vittoria Scarpati G, Falcetta F, Carlomagno C, Ubezio P, Marchini S, De Stefano A, Singh VK, D'Incalci M, De Placido S and Pepe S: A specific miRNA signature correlates with complete pathological response to neoadjuvant chemoradiotherapy in locally advanced rectal cancer. Int J Radiat Oncol Biol Phys. 83:1113–1119. 2012. View Article : Google Scholar

123 

Hotchi M, Shimada M, Kurita N, Iwata T, Sato H, Morimoto S, Yoshikawa K, Higashijima J and Miyatani T: microRNA expression is able to predict response to chemoradiotherapy in rectal cancer. Mol Clin Oncol. 1:137–142. 2013.PubMed/NCBI

124 

Kheirelseid EA, Miller N, Chang KH, Curran C, Hennessey E, Sheehan M, Newell J, Lemetre C, Balls G and Kerin MJ: miRNA expressions in rectal cancer as predictors of response to neoadjuvant chemoradiation therapy. Int J Colorectal Dis. 28:247–260. 2013. View Article : Google Scholar

125 

Svoboda M, Sana J, Fabian P, Kocakova I, Gombosova J, Nekvindova J, Radova L, Vyzula R and Slaby O: MicroRNA expression profile associated with response to neoadjuvant chemoradiotherapy in locally advanced rectal cancer patients. Radiat Oncol. 7:1952012. View Article : Google Scholar : PubMed/NCBI

126 

Calanzani N, Druce PE, Snudden C, Milley KM, Boscott R, Behiyat D, Saji S, Martinez-Gutierrez J, Oberoi J, Funston G, et al: Identifying novel biomarkers ready for evaluation in low-prevalence populations for the early detection of upper gastrointestinal cancers: A systematic review. Adv Ther. Dec 11–2020.Epub ahead of print. View Article : Google Scholar : PubMed/NCBI

127 

Azizian A, Epping I, Kramer F, Jo P, Bernhardt M, Kitz J, Salinas G, Wolff HA, Grade M, Beißbarth T, et al: Prognostic value of MicroRNAs in preoperative treated rectal cancer. Int J Mol Sci. 17:5682016. View Article : Google Scholar : PubMed/NCBI

128 

Toyota M, Suzuki H, Sasaki Y, Maruyama R, Imai K, Shinomura Y and Tokino T: Epigenetic silencing of microRNA-34b/c and B-cell translocation gene 4 is associated with CpG island methylation in colorectal cancer. Cancer Res. 68:4123–4132. 2008. View Article : Google Scholar : PubMed/NCBI

129 

Campayo M, Navarro A, Benítez JC, Santasusagna S, Ferrer C, Monzó M and Cirera L: miR-21, miR-99b and miR-375 combination as predictive response signature for preoperative chemoradiotherapy in rectal cancer. PLoS One. 13:e02065422018. View Article : Google Scholar : PubMed/NCBI

130 

Machackova T, Trachtova K, Prochazka V, Grolich T, Farkasova M, Fiala L, Sefr R, Kiss I, Skrovina M, Dosoudil M, et al: Tumor microRNAs identified by small RNA sequencing as potential response predictors in locally advanced rectal cancer patients treated with neoadjuvant chemoradiotherapy. Cancer Genomics Proteomics. 17:249–257. 2020. View Article : Google Scholar : PubMed/NCBI

131 

Li AL, Chung TS, Chan YN, Chen CL, Lin SC, Chiang YR, Lin CH, Chen CC and Ma N: microRNA expression pattern as an ancillary prognostic signature for radiotherapy. J Transl Med. 16:3412018. View Article : Google Scholar : PubMed/NCBI

132 

Rupaimoole R and Slack FJ: MicroRNA therapeutics: Towards a new era for the management of cancer and other diseases. Nat Rev Drug Discov. 16:203–222. 2017. View Article : Google Scholar : PubMed/NCBI

133 

Renganathan A and Felley-Bosco E: Long noncoding RNAs in cancer and therapeutic potential. Adv Exp Med Biol. 1008:199–222. 2017. View Article : Google Scholar : PubMed/NCBI

134 

Guzel E, Okyay TM, Yalcinkaya B, Karacaoglu S, Gocmen M and Akcakuyu MH: Tumor suppressor and oncogenic role of long non-coding RNAs in cancer. North Clin Istanb. 7:81–86. 2019.

135 

Zhang H, Chen Z, Wang X, Huang Z, He Z and Chen Y: Long non-coding RNA: A new player in cancer. J Hematol Oncol. 6:372013. View Article : Google Scholar : PubMed/NCBI

136 

Rasool M, Malik A, Zahid S, Basit Ashraf MA, Qazi MH, Asif M, Zaheer A, Arshad M, Raza A and Jamal MS: Non-coding RNAs in cancer diagnosis and therapy. Noncoding RNA Res. 1:69–76. 2016. View Article : Google Scholar : PubMed/NCBI

137 

Dizaji BF: Strategies to target long non-coding RNAs in cancer treatment: Progress and challenges. Egypt J Med Hum Genet. 21:412020. View Article : Google Scholar

138 

Yang N: An overview of viral and nonviral delivery systems for microRNA. Int J Pharm Investig. 5:179–181. 2015. View Article : Google Scholar : PubMed/NCBI

139 

Labatut AE and Mattheolabakis G: Non-viral based miR delivery and recent developments. Eur J Pharm Biopharm. 128:82–90. 2018. View Article : Google Scholar : PubMed/NCBI

140 

Zhang Y, Wang Z and Gemeinhart RA: Progress in microRNA delivery. J Control Release. 172:962–974. 2013. View Article : Google Scholar : PubMed/NCBI

141 

Meng WJ, Yan H, Zhou B, Zhang W, Kong XH, Wang R, Zhan L, Li Y, Zhou ZG and Sun XF: Correlation of SATB1 overexpression with the progression of human rectal cancer. Int J Colorectal Dis. 27:143–150. 2012. View Article : Google Scholar

142 

Zhang P, Wang L, Rodriguez-Aguayo C, Yuan Y, Debeb BG, Chen D, Sun Y, You MJ, Liu Y, Dean DC, et al: miR-205 acts as a tumour radiosensitizer by targeting ZEB1 and Ubc13. Nat Commun. 5:56712014. View Article : Google Scholar : PubMed/NCBI

143 

Huang X, Taeb S, Jahangiri S, Emmenegger U, Tran E, Bruce J, Mesci A, Korpela E, Vesprini D, Wong CS, et al: miRNA-95 mediates radioresistance in tumors by targeting the sphingolipid phosphatase SGPP1. Cancer Res. 73:6972–6986. 2013. View Article : Google Scholar : PubMed/NCBI

144 

Ha Thi HT, Kim HY, Kim YM and Hong S: MicroRNA-130a modulates a radiosensitivity of rectal cancer by targeting SOX4. Neoplasia. 21:882–892. 2019. View Article : Google Scholar : PubMed/NCBI

145 

Kelley KA, Ruhl RA, Rana SR, Dewey E, Espinosa C, Thomas CR Jr, Martindale RG, Anand S and Tsikitis VL: Understanding and resetting radiation sensitivity in rectal cancer. Ann Surg. 266:610–616. 2017. View Article : Google Scholar : PubMed/NCBI

146 

Ruhl R, Rana S, Kelley K, Espinosa-Diez C, Hudson C, Lanciault C, Thomas CR Jr, Liana Tsikitis V and Anand S: microRNA-451a regulates colorectal cancer proliferation in response to radiation. BMC Cancer. 18:5172018. View Article : Google Scholar : PubMed/NCBI

147 

Luo J, Liu L, Zhou N, Shen J, Sun Q, Zhu Y and Chen M: miR-519b-3p promotes responsiveness to preoperative chemo-radiotherapy in rectal cancer patients by targeting ARID4B. Gene. 655:84–90. 2018. View Article : Google Scholar : PubMed/NCBI

148 

Winter SF, Lukes L, Walker RC, Welch DR and Hunter KW: Allelic variation and differential expression of the mSIN3A histone deacetylase complex gene Arid4b promote mammary tumor growth and metastasis. PLoS Genet. 8:e10027352012. View Article : Google Scholar : PubMed/NCBI

149 

Yang P, Yang Y, An W, Xu J, Zhang G, Jie J and Zhang Q: The long noncoding RNA-ROR promotes the resistance of radiotherapy for human colorectal cancer cells by targeting the p53/miR-145 pathway. J Gastroenterol Hepatol. 32:837–845. 2017. View Article : Google Scholar

150 

Zou Y, Yao S, Chen X, Liu D, Wang J, Yuan X, Rao J, Xiong H, Yu S, Yuan X, et al: LncRNA OIP5-AS1 regulates radioresistance by targeting DYRK1A through miR-369-3p in colorectal cancer cells. Eur J Cell Biol. 97:369–378. 2018. View Article : Google Scholar : PubMed/NCBI

151 

Wang G, Li Z, Zhao Q, Zhu Y, Zhao C, Li X, Ma Z, Li X and Zhang Y: LincRNA-p21 enhances the sensitivity of radiotherapy for human colorectal cancer by targeting the Wnt/β-catenin signaling pathway. Oncol Rep. 31:1839–1845. 2014. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

March-2021
Volume 58 Issue 3

Print ISSN: 1019-6439
Online ISSN:1791-2423

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Ha Thi H, Duong HQ and Hong S: Emerging roles of non‑coding RNAs in the response of rectal cancer to radiotherapy (Review) Corrigendum in /10.3892/ijo.2021.5247. Int J Oncol 58: 344-358, 2021
APA
Ha Thi, H., Duong, H., & Hong, S. (2021). Emerging roles of non‑coding RNAs in the response of rectal cancer to radiotherapy (Review) Corrigendum in /10.3892/ijo.2021.5247. International Journal of Oncology, 58, 344-358. https://doi.org/10.3892/ijo.2021.5175
MLA
Ha Thi, H., Duong, H., Hong, S."Emerging roles of non‑coding RNAs in the response of rectal cancer to radiotherapy (Review) Corrigendum in /10.3892/ijo.2021.5247". International Journal of Oncology 58.3 (2021): 344-358.
Chicago
Ha Thi, H., Duong, H., Hong, S."Emerging roles of non‑coding RNAs in the response of rectal cancer to radiotherapy (Review) Corrigendum in /10.3892/ijo.2021.5247". International Journal of Oncology 58, no. 3 (2021): 344-358. https://doi.org/10.3892/ijo.2021.5175