Next Article in Journal
The Role of Cytokines in Epithelial–Mesenchymal Transition in Gynaecological Cancers: A Systematic Review
Next Article in Special Issue
Plasma-Derived Exosome Proteins as Novel Diagnostic and Prognostic Biomarkers in Neuroblastoma Patients
Previous Article in Journal
Autophagy as a Therapeutic Target for Chronic Kidney Disease and the Roles of TGF-β1 in Autophagy and Kidney Fibrosis
Previous Article in Special Issue
Cell Pair Algorithm-Based Immune Infiltrating Cell Signature for Improving Outcomes and Treatment Responses in Patients with Hepatocellular Carcinoma
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Promises and Challenges of Predictive Blood Biomarkers for Locally Advanced Rectal Cancer Treated with Neoadjuvant Chemoradiotherapy

by
Joao Victor Machado Carvalho
1,2,3,
Valérie Dutoit
1,2,
Claudia Corrò
1,2,3,† and
Thibaud Koessler
1,2,3,*,†
1
Translational Research Center in Onco-Hematology, Department of Medicine, Faculty of Medicine, University of Geneva, 1205 Geneva, Switzerland
2
Swiss Cancer Center Léman, 1005 Lausanne, Switzerland
3
Department of Oncology, Geneva University Hospital, 1205 Geneva, Switzerland
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Cells 2023, 12(3), 413; https://doi.org/10.3390/cells12030413
Submission received: 13 January 2023 / Accepted: 24 January 2023 / Published: 26 January 2023
(This article belongs to the Special Issue Early Biomarkers of Cancer: Diagnosis and Progression)

Abstract

:
The treatment of locally advanced rectal cancer (LARC) requires a multimodal approach combining neoadjuvant radiotherapy or chemoradiotherapy (CRT) and surgery. Predicting tumor response to CRT can guide clinical decision making and improve patient care while avoiding unnecessary toxicity and morbidity. Circulating biomarkers offer both the advantage to be easily accessed and followed over time. In recent years, biomarkers such as proteins, blood cells, or nucleic acids have been investigated for their predictive value in oncology. We conducted a comprehensive literature review with the aim to summarize the status of circulating biomarkers predicting response to CRT in LARC. Forty-nine publications, of which forty-seven full-text articles, one review and one systematic review, were retrieved. These studies evaluated circulating markers (CEA and CA 19-9), inflammatory biomarkers (CRP, albumin, and lymphocytes), hematologic markers (hemoglobin and thrombocytes), lipids and circulating nucleic acids (cell-free DNA [cfDNA], circulating tumor DNA [ctDNA], and microRNA [miRNA]). Post-CRT CEA levels had the most consistent association with tumor response, while cfDNA integrity index, MGMT promoter methylation, ERCC-1, miRNAs, and miRNA-related SNPs were identified as potential predictive markers. Although circulating biomarkers hold great promise, inconsistent results, low statistical power, and low specificity and sensibility prevent them from reliably predicting tumor response following CRT. Validation and standardization of methods and technologies are further required to confirm results.

1. Introduction

According to the European Society for Medical Oncology (ESMO) in 2017, rectal cancer (RC) represents 35% of colorectal cancers (CRC); in the European Union, the incidence of RC is 125,000 per year (15–20 cases/100,000 habitants per year), and the mortality is 4–10 deaths/100,000 habitants [1]. It is suggested that these numbers may rise in the future [1,2]. Locally advanced RC (LARC) corresponds to stage II or III RC and is the most prevalent stage at diagnosis in Europe [1,3].
For LARC, European and American medical oncology guidelines recommend curative surgery with total mesorectal excision (TME) and removal of the mesorectal nodes en-bloc [1,4]. To decrease the risk of local recurrence, neo-adjuvant radiotherapy (RT) or chemo-RT (CRT) is used [1]. CRT combines 45–50.4 Gy radiation in 25 fractions of 1.8–2 Gy with radio-sensitizing chemotherapy using capecitabin or fluorouracil (5-FU) for 6 weeks [1]. RT alone consists of a short-course radiotherapy (SCRT) of 25 Gy in 5 fractions of 5 Gy. Surgery takes place between 1 and 12 weeks after completion of neoadjuvant treatment [1]. It has been shown that longer waiting times do not improve local control but increase the chances of pathological complete response (pCR) [5,6]. The ideal time to surgery is currently unknown [7]. To date, CRT and SCRT with delayed surgery (at least 4 weeks after RT) are considered standard of care and interchangeable [6,8]. Treatment intensification with total neoadjuvant treatment (TNT)—adding consolidation or induction chemotherapy to standard of care—further improves distant control and pCR (up to 30%) as shown in the RAPIDO [9] and PRODIGE-23 [10] trials.
In LARC, despite neoadjuvant treatments, pCR occurs in only 6 to 39% of patients [11,12,13,14]. It means that a large proportion of the patients do not benefit from neoadjuvant treatment while experiencing side effects from it [15]. Therefore, being able to predict treatment response is of utmost importance as it can determine patients who would benefit from neoadjuvant treatment and minimize the harmful effects of it when it is not appropriate [16]. Another major goal is to avoid removing the rectum and losing the organ, losing its function, and introducing the associated surgical complications [15]. For patients not to be operated on, they need to achieve clinical complete response (cCR) or near cCR which could be complemented with local excision. Patients in cCR usually enter a “watch and wait” program with regular clinical, radiological, and endoscopical assessments [16].
Despite the existence of well-known measurable circulating biomarkers, none are available in clinical use to predict the tumor response to neoadjuvant treatment in LARC patients [1]. Our review aims at clarifying the performance and clinical utility of the circulating biomarkers for predicting the tumor response in the context of LARC patients undergoing CRT (Figure 1).

2. Materials and Methods

We performed a systematic literature search of the Embase database on 18 November 2021, using the following keywords: “locally advanced rectal cancer”, “rectal cancer”, “blood biomarker”, “biological marker”, “tumor marker”, “tumor response”, “cancer regression”, “complete response”,” preoperative treatment”, “neoadjuvant”, and “chemoradiotherapy” with the Boolean AND/OR.
We only included studies evaluating tumor response as an endpoint based on the pCR, tumor regression grade (TRG), or tumor downstaging. Studies evaluating overall survival (OS) and/or disease-free survival (DFS) only were not included. Patients needed to have stage II or III RC treated with CRT (CRT) using capecitabine or 5-FU chemotherapy and a total radiation dosage of 45 to 50.4 Gy. Notably, other long-course CRT with RT schemes of 39.6 to 63 Gy were also included (Table S1).
When possible, sensitivity (Sn), specificity (Sp), positive predictive value (PPV), and negative predictive value (NPV) were calculated based on the cut-off values available.

3. Results

3.1. Literature Search Results

Taken together, the literature search resulted in the selection of fifteen full text articles. In addition to these studies, thirty-four articles were added after analyzing the cited literature from the fifteen selected articles. All added articles met the inclusion criteria. In total forty-nine studies were included. An overview of the selection process is provided in Figure 2.
Twenty-five of these studies investigated the following biomarkers: carcinoembryonic antigen (CEA), carbohydrate antigen (CA 19-9), thrombocytes, hemoglobin (Hb), leukocytes, lymphocytes, c-reactive protein (CRP), albumin, and lipids, while twenty-four studies focused on acid nucleic liquid biopsies, analyzing cell free DNA (cfDNA), circulating tumor DNA (ctDNA), and microRNA (miRNA) (Table S1).

3.2. Protein Tumor Markers

3.2.1. CEA

CEA is a glycoprotein expressed on the apical part of the normal epithelial cell membrane. In tumor cells, CEA loses its polarization, which increases its expression and concentration in the circulation [17]. CEA is a prognostic marker in CRC and is used to detect tumor relapse after surgery [1] and progression during treatment [18]. No specific cut-off value exists for local recurrence in RC [19], and its predictive value for tumor relapse pre-surgery is controversial [20]. We identified twenty-four studies investigating CEA levels as a predictor of tumor regression at different time points: pre-CRT and post-CRT (Table 1).
Pre-CRT CEA levels. Twenty-one studies investigated pre-CRT CEA levels using different cut-offs: 2.5 ng/mL [21], 2.7 ng/mL [22], 2.85 ng/mL [23], 3.5 ng/mL [24], 4.4 ng/mL [25], 5 ng/mL [26,27,28,29,30,31,32,33,34,35,36,37,38,39], and 6 ng/mL [40]. Eleven studies found that CEA values lower than the cut-off in pre-CRT was an independent and positive predictive factor of tumor response to CRT in univariate analyses [21,22,23,25,26,28,30,31,34,35,41]. In multivariate analyses, low CEA value pre-CRT remained an independent predictive factor of tumor regression in eight studies [25,26,28,30,31,34,35,41]. The ten other studies found no statistical correlation between pre-CRT CEA level and tumor regression [24,27,29,32,33,36,37,38,39,40].
Post-CRT pre-surgery levels. Ten studies explored post-CRT but pre-surgery CEA levels with different cut-offs: 2 ng/mL [32], 2.45 ng/mL [23], 2.61 ng/mL [40], 2.7 ng/mL [24], and 5 ng/mL [29,31,36,39,42,43]. Seven studies found that CEA value lower than the cut-off in the post-CRT setting is predictive of tumor regression [23,24,31,32,39,40,42] in univariate and multivariate analysis, and three studies reported no association [29,36,43].
Two studies [23,39] considered the ratio between pre- and post-CRT CEA levels. Cai et al. [39] evaluated a post-CRT/pre-CRT CEA ratio, whereas Song et al. [23] evaluated a pre-CRT/post-CRT ratio. Cai et al. [39] use 0.23 as a cut-off value and demonstrated its predictive value for tumor regression in univariate and multivariate analyses for patients with values lower than 0.23. Song et al. [23] found that a value lower than 1.07 was an independent predictive factor of pCR in univariate and multivariate analyses.
To reduce the effect that tumor size can have on CEA levels, Gago et al. [33] consider the ratio between the pre-operative CEA level and the maximum tumor diameter (measured by MRI). Using a cut-off value of 2.429 ng per mL per cm, they demonstrated that patients having a lower value after neoadjuvant CRT had a higher chance of reaching pCR (66.3% versus 14.3%), but it was not significant in multivariate analysis.
Finally, Hu et al. [43] measured CEA levels at several time points: baseline (pre-CRT) and then at 2, 6, and 12 weeks after the start of treatment. They measured the CEA clearance pattern by drawing exponential curves based on trend lines. Then, they calculated the R2 value which represent the “correlation coefficient between the trend line illustrating the exponential decrease and the measured CEA values” [43]. If the R2 is equal or close to 1, it means that the patient had an adequate tumor response, and, in fact, they find that the clearance pattern with and exponential decrease in CEA during treatment (R2 ≧ 0.9) is predictive of pCR in univariate and multivariate analysis in a cohort of 146 patients derived from the two arms of the prospective trial FOWARC [44] comparing standard CRT to modified FOLFOX6 with or without RT.

3.2.2. CA 19-9

CA 19-9 is a cell surface glycoprotein complex secreted by a variety of secretory cells including pancreatic, biliary ductal, gastric, colon, endometrial, and salivary cells as part of mucous secretions [45,46]. CA 19-9 is prognostic factor in pancreatic adenocarcinoma [47,48]. It can be falsely negative in patients Lewis (Le) negative blood group, which represent 7–10% of the global population [48].
In RC, seven studies measured CA 19-9 levels using cut-off values of 3.5 U/mL [22], 9 U/mL [26], 10 U/mL [26], 12.6 U/mL [23], 35 U/mL [34], or 37 U/mL [28,30,39]. None of these studies showed significant results using CA 19-9 levels in pre- or post-CRT settings (Table S2). When evaluating tumor downstaging, two studies showed that pre-CRT CA 19-9 level were predictive of response in univariate analysis [23,26], but only Yeo et al. [26] observed a significant association in multivariate analysis.
Yeo et al. [26] studied the predictive value of tumor downstaging for post-CRT CA 19.9 but failed to show a significant association. They evaluated the pre- and post-CA 19-9 ratio using a cut-off value of 1.28 (for downstaging) and 0.92 (for pCR). They found that lower values were predictive of tumor downstaging and pCR in univariate analyses, whereas this held true in multivariate analyses for downstaging only.
Finally, Yang et al. [30] looked at the predictive value of a decreased pre- and post-CRT CA 19.9 ratio using a cut-off value of 37 U/mL but did not observe association with tumor response.

3.3. Hematological Markers

3.3.1. Thrombocytes

It is thought that thrombocytes can promote cancer through several mechanisms: shielding tumor cells from the immune system, supporting tumor cell extravasation, and stimulating angiogenesis through the secretion of proangiogenic cytokines such as VEGF or PDGF [49]. In turn, tumor cells can promote thrombocytosis by secreting thrombopoietin, which is correlated with poorer outcome in solid cancers such as pancreatic adenocarcinoma [50] and non-small cell lung cancer [51]. In the context of LARC, four studies looked at the predictive value of thrombocyte level in pre-CRT or post-CRT with cut-off values ranging from 253 G/L to 370 G/L [22,28,41,52] (Table 2). Only two studies demonstrated that high pre-CRT thrombocyte levels were predictive of poor likelihood of response in univariate analyses [22,28], whereas only one study remained significant for predicting pCR after multivariate analyses [28]. A single study looked at post-CRT thrombocyte levels with no significant result [41].

3.3.2. Hemoglobin (Hb)

Anemia is associated with poor outcomes in several cancers [53] probably due to intratumoral hypoxia [54], which in turn stimulates angiogenesis at the tumor site via secretion of HIF and VEGF [55]. Hypoxia can further modify tumor cell metabolism and induce cell quiescence, which can increase treatment resistance, especially in the context of RT [56,57]. In LARC, ten studies explored association of pre-CRT Hb levels with tumor response after CRT [22,24,25,26,29,30,32,41,52,58]; one study considered post-CRT Hb levels [41], and one considered Hb variation during treatment [32]. Several cut-off values are investigated, including 9 g/dL [30], 10 g/dL [29,32], 12 g/dL [52,58], 12.2 g/dL [22], 12.5 g/dL [26], 13.2 g/dL [25], and 13.5 g/dL [24] (Table S3). Regarding pre-CRT Hb level, three studies [22,41,58] demonstrate an association with tumor response in univariate analysis, but none demonstrate an association with tumor response in multivariate analysis. Post-CRT Hb levels were not associated with tumor response in any of the studies. Finally, Huang et al. showed that patients without anemia (Hb > 10 g/dL) during CRT were more likely to achieve pCR compared to those with anemia; these results remained significant in multivariate analyses [32].

3.4. Leukocytes and Inflammatory Markers

3.4.1. Albumin and C-Reactive Protein

Inflammation is a well-known hallmark of cancer [59]. It is measured in peripheral blood using c-reactive protein (CRP), albumin, or circulating leukocytes levels [60]. We found ten studies looking at inflammatory markers as predictive markers in LARC patients treated with CRT [22,25,27,31,32,38,41,52,61,62] (Tables S4 and S5). Three studies looked at CRP or albumin [22,41,52]; four studies measured leukocytes levels [25,27,32,41]; and seven investigated the ratio between leukocytes and/or CRP and/or albumin [22,31,38,41,52,61,62] or its combinations [38].
Albumin is the most abundant serum protein [63]. Its level decreases during an inflammatory event, this being usually associated with poor outcomes [64,65]. Two studies [41,52] evaluating the predictive value of albumin in LARC patients failed to show any association of pre- or post-CRT albumin levels with tumor response.
CRP is an acute-phase protein whose concentration in plasma rises during inflammation and is routinely measured as a diagnostic marker for inflammatory events [66,67]. High CRP levels are associated with poor outcomes [68,69]. In LARC, Aires et al. [22] found that pre-treatment CRP level below 3.5 mg/L was an independent predictive factor of tumor regression in univariate and multivariate analyses.
CRP and albumin have been combined in the Glasgow prognostic score (GPS) and the modified GPS (mGPS). GPS scores from 0 to 2; one score is given for CRP levels above 10 mg/L or albumin levels below 35 g/L [70,71]. In the mGPS, isolated hypoalbuminemia (<35 g/L) does not score [72]; therefore, a mGPS score of 1 represents an isolated elevation of CRP (>10 mg/L), and a mGPS score of 2 represents an elevation of CRP with hypoalbuminemia [73]. Dreyer et al. [62] showed that a pre-CRT mGPS score of 0 was associated with increased tumor regression and predicted TRG in univariate and in multivariate analyses.

3.4.2. Leukocytes

Eight studies investigated circulating leukocyte levels as a predictor of tumor response after CRT in LARC [25,27,31,32,38,41,52,61] (Table S5). Three studies evaluated leukocyte counts before [25], during [32], or before and after [41] CRT treatment, but none showed significant association with tumor response. On the contrary, Kitayama et al. [27] showed that high lymphocyte counts pre-CRT was significantly associated with response in univariate and multivariate analysis. Likewise, Heo et al. [25] found that a high-sustained lymphocyte count after 4 weeks of CRT compared to the pre-CRT count was predictive of pCR in univariate and multivariate analyses. Noticeably, results at other time points (after 8 or 12 weeks) were not significant (Table S5).
The neutrophil to lymphocyte ratio (NLR) reflects the dynamic interaction between innate and adaptative immunity in inflammatory events such as sepsis or cancer [74]. NLR is a prognostic marker in several solid tumor types with high ratios being associated with poorer outcomes (e.g., reduced overall survival or disease-free survival) [75,76]. In LARC, seven studies evaluated its predictive value [22,31,38,41,52,61,62], all measuring NLR pre-CRT with a cut-off ranging from 1.7 to 5 (Table S5). In univariate analysis, two studies reached statistical significance [31,41]. One identified NLR < 2.8 associated with pCR in multivariate analysis [31]. No association could be demonstrated for NLR in post-CRT [41].
The neutrophils to albumin ratio (NAR) is predictive of mortality or has a prognostic value in several tumor types [77,78,79]. In LARC, two studies measured its predictive value for tumor response to CRT [38,41]. Out of the two studies, only Tawfik et al. [41] found that an elevated pre-CRT NAR was associated with decreased likelihood of pCR in univariate and multivariate analysis, while post-CRT NAR was not.
The lymphocyte to monocyte ratio (LMR) is associated with worse tumor control in breast or lung cancer [80,81]. Three studies investigated LMR pre- or post-CRT in LARC [38,41,61], but none showed significant results in prediction of treatment response.
The platelet to lymphocyte ratio (PLR) has been shown to be predictive of worse overall survival (OS) in several solid tumors [82]. Regarding the response to CRT in LARC, none of four studies investigating PLR showed a significant association between PLR measured pre- or post-CRT and response [31,41,61,62].
Finally, Sawada et al. [38] evaluated seven other ratios and combinations: the lymphocyte to CRP ratio (LCR), neutrophil and CRP product (N×C), monocyte and CRP product (M×C), neutrophil and monocyte product (N×M), monocyte to albumin ratio (MAR), CRP to Albumin ratio (CAR), and the prognostic nutritional index (PNI) which is calculated by using an equation combining serum albumin levels and the lymphocyte count [83]. They show that pre-CRT high LCR and low (N×M) correlate with better TRG in univariate and multivariate analysis.

3.5. Lipid Marker

Apolipoprotein A-1

Lipids can promote oncogenesis by allowing tumor cells to meet their increased metabolic demand, by modulating being part of tumor-modulating pathways, by inducing the recruitment of inflammatory cells, and by playing immuno-modulatory roles [84,85,86]. One study [34] looked at the predictive value of different lipids in relation to tumor regression following CRT. Only pre-treatment apolipoprotein A–I levels ≤ 1.20 g/L correlated with a lower proportion of responders, the association remaining significant in multivariate analysis.

3.6. Nucleic Acids Marker

The liquid biopsy technique is the analysis of non-solid tissues such as blood or other bodily fluids such as saliva, cerebrospinal fluid, or urine [87]. Analyzed components of liquid biopsies such as circulating tumor cells (CTC) [88] or nucleic acids (cfDNA, ctDNA, or miRNA) show great potential for clinical application in oncology for tumor early diagnosis, detection of tumor relapse after surgery, or identification of treatment targets [89]. We identified twenty-two studies evaluating cfDNA, ctDNA, or miRNA as predictor of the tumor response after neoadjuvant treatment in LARC patients. Out of twenty-two studies, twenty-one are discussed in two reviews [90,91].

3.6.1. Cell Free DNA

The uncovering of cfDNA dates back to 1948 when Mandel and Metais et al. observed presence of DNA and RNA in the blood of healthy and diseased patients [92]. cfDNA is now detected under physiological conditions (e.g., physical activity) or pathological conditions such auto-immune, inflammatory, or oncologic diseases [93,94]. cfDNA is either released following tissue damage [95,96] or is actively secreted [97]. In the bloodstream, its half-life is 4 to 30 min [98]. cfDNA is detectable in different bodily fluids such as urine or blood [99]. In healthy individuals, cfDNA blood concentration ranges from 1 to 10 ng/mL [94].
Five studies evaluated the predictive value of cfDNA in LARC [100,101,102,103,104] with four reporting significant results (Table 3). Zitt et al. [100] showed a significant association between cfDNA levels and tumor downstaging following CRT with cfDNA decrease after surgery significantly associated with tumor response in the multivariate analyses. Using cfDNA integrity index [105]—the ratio between long and short cfDNA fragments—Agostini et al. [101] found that a lower cfDNA integrity index post-CRT was associated with increased tumor response in multivariate analysis. Sun et al. [102] showed that long cfDNA fragments (≈400 bp) abundancy and its ratio to short cfDNA fragments (100 bp) in pre-CRT were significantly associated with tumor response. They also showed that higher MGMT promoter methylation pre-CRT predicted better TRG. Similarly, Shalaby et al. [104] studied the pre-CRT methylation of MGMT and ERCC-1 promoters, with MGMT and ERCC-1 playing an important role in DNA repair mechanisms. In both cases, a hypermethylation of MGMT and ERCC-1 promoter was associated with decreased tumor regression.

3.6.2. Circulating Tumor DNA

In 1989, Stroun et al. discovered the presence of ctDNA as part of the cfDNA [106]. ctDNA detection in blood is achieved using targeted or untargeted approaches [89,107]. Targeted approaches are PCR-based and require prior knowledge of specific mutation(s) of interest that will be researched for in the blood [107]. Untargeted approaches use next generation sequencing (NGS) to detect unknown mutations than can thereafter be followed [107]. This method has the advantage of not requiring knowledge of mutations before performing the test but is less sensitive than the targeted approach [107].
Nine studies evaluated ctDNA as predictor of tumor response following CRT in LARC [37,108,109,110,111,112,113,114,115]. Eight studies [37,108,109,110,111,112,113,115] considered ctDNA levels; one study used BRAF and KRAS mutations in ctDNA [113]; and one used neuropeptide Y (NPY) methylation status in ctDNA [114] (Table 4). The detection rate of ctDNA in these studies ranged from 20.5% to 75% in pre-CRT, 8.3% to 22.3% in post-CRT, and 6.7% to 13% in post-surgery samples (Table 4). Two studies showed significant results regarding post-CRT DNA levels. Khakoo et al. [112] and Zhou et al. [110] demonstrated that post-CRT ctDNA levels was associated with TRG (mrTRG) [112] or pCR [110] and TRG (CAP) [110]. Finally, Murahashi et al. [37] showed that a loss of >80% between post-CRT ctDNA and baseline ctDNA (pre-CRT) predicted TRG in univariate and multivariate analyses.

3.6.3. MicroRNA

microRNAs (miRNAs) are non-coding RNA molecules which regulate gene expression and cell-cell interactions in autocrine, paracrine, or endocrine ways [116,117]. In general, onco-miRNAs are overexpressed in cancer cells and increase degradation of tumor suppressor messenger RNA (mRNA), while tumor suppressive miRNAs are usually less expressed in tumor patients [118,119,120]. In blood, miRNAs are also shed in exosomes in all bodily fluids, and recent studies suggest that they are associated with tumorigenesis [120]. Exosomal miRNAs are resistant against degradation which makes them potential biomarkers of interest [120,121].
Several studies investigating several different kinds of miRNA of pre-treatment formalin-fixed and paraffin embedded (FFPE) biopsy samples demonstrated an association with tumor response in LARC patients undergoing CRT (pCR/TRG) [122,123,124]. Eight studies investigated blood circulating miRNA predictive value of tumor response following CRT in RC [36,125,126,127,128,129,130,131] (Table 5).
Yu et al. [127] identified low pre-CRT levels of miR-345 as a predictor of TRG (Mandard). D’Angelo et al. [126] showed that pre-CRT miR-125b was preferentially overexpressed in non-responders. Hiyoshi et al. [131] showed that low pre-CRT levels of miR-43 were predictive of response to CRT; however, they did not reproduce Yu’s or D’Angelo’s results regarding miR-125b and miR-345. Azizian et al. [128] showed that reduced miR-20a and miR-18b during CRT was predictor of negative postoperative nodal status; however, no correlation with TRG was found. Wada et al. [36] identified a panel of 8 pre-CRT miRNA (miR-30e-5p, miR-33a-5p, miR-130a-5p, miR-210-3p, miR-214-3p, miR-320a, miR-338-3p, and miR-1260a) for which variations were predictors of tumor response (TRG) to CRT in LARC. Looking specifically at exosomal miRNAs, Baek et al. [130] identified pre-CRT miR-199b-5p upregulation as predictor of response in multivariate analyses. Finally, Dreussi et al. [125] investigated a series of 114 single nucleotide polymorphisms (SNPs) related to miRNA and found that DROSHA-rs10719, SMAD3-rs17228212, SMAD3 rs744910, and SMAD3-rs745103 SNPs were associated with non-pCR, whereas TRBP-rs6088619 mutation was associated with pCR.

4. Discussion

This review explores the potential value of several circulating biomarkers to predict LARC regression following CRT. The forty-nine selected studies looked at twelve single biomarkers (CEA, CA 19-9, Hb, thrombocytes, leukocytes, lymphocytes, albumin, CRP, apoA1, cfDNA, ctDNA, and selected miRNA) and twelve multiple biomarkers (mGPS, NLR, NAR, LMR, PLR, LCR, MAR, CAR, PNI, N×C, M×C, and N×M).
Pre-treatment (pre-CRT) is the most relevant time point when predicting neoadjuvant treatment effect as it can guide the clinician in choosing the right approach prior any treatment is administrated. In this setting, CEA is the most studied circulating marker. It shows significant association in univariate and multivariate analyses with tumor response in eight out of twenty-one studies. The most widely used cut-off is 5 ng/mL. With this cut-off, sensitivity ranges from 61.7% to 92.6%, and specificity ranges from 41.8% to 63%. These characteristics do not allow CEA to be a clinically relevant biomarker, and it is therefore not currently used to guide clinical decision. A second circulating biomarker associated with poor tumor response to CRT is detection in cfDNA of MGMT promoter hypermethylation [102,104]. However, these results originate from two small studies and warrant replication in a larger cohort of patients. Other markers studied during the pre-CRT window show either inconsistent results (e.g., CA 19-9, thrombocytes), no association (e.g., hemoglobin, albumin, or ctDNA) or positive association (e.g., CRP, apolipoprotein A-I, or miR-125b). The latest were shown in only one study and failed to be reproduced.
Single liquid biomarkers tested at a post-CRT pre-surgery time point are more consistently associated with tumor response. Some markers such as ctDNA are associated with tumor response only when measured during this period [110,112]. This observation illustrates the importance of selecting the right time point of analysis for each biomarker. Out of all biomarkers tested, CEA showed frequent and significant association with tumor response in eight studies out of eleven. Improvement in the test sensitivity, ranging from 90.9% to 100% while specificity ranged from 25% to 32.5% at a cut-off set at 5 ng/mL, was observed when this time point is used. However, this time point (post-CRT but pre-surgery) is of less clinical relevance, as neo-adjuvant CRT has already been given. It could, however, complement clinical, radiological, and endoscopic assessments for patients on a watch and wait track. Nevertheless, the true added value of these biomarkers in the watch and wait setting must be specifically studied, as none of the studies reviewed addresses this topic.
To improve on the predicting value of circulating biomarkers, authors used two different strategies: testing at multiple time points to assess biomarker change over time and ratio or testing of multiple biomarkers. For change over time and ratios measurement, usually one time point before CRT and one time point before surgery is selected. Authors use different mathematical analyses, ratios between post- and pre-CRT levels being most commonly used. Most of these analyses showed no predictive impact, with the noticeable exception of Song et al. [23] showing that CEA and CA19-9 pre-CRT over post-CRT ratios were independently and statistically associated with downstaging in a large patient cohort (n = 674). On the other hand, most authors exploring multiple markers were equally unsuccessful, apart from Sawada’s neutrophil × monocytes value and lymphocytes to CRP ratio [38]. Significant associations with the tumor response have also been shown by Dreussi et al. [125] using 6 SNPs and Wada et al. [36] using a panel of 8 miRNAs. However, despite a large sample size [125] or validation cohort [36], these finding have currently never been replicated. Wada et al. [36] showed that the predictive value of a miRNA panel is improved when combined with the CEA levels, highlighting the fact that multiple markers should be used as they interrogate different tumor components. Indeed, a possible explanation for the failure of other miRNA studies is that miRNA transcriptional profile is modified by the tumor microenvironment and other parameters such as the oxygenation levels [132], which CEA is less sensitive to.
Several factors might be responsible for the lack of a predictive value from the studied circulating biomarkers. First, most studies have small sample size, with on average 184 patients per study, which leaves 92 patients in each group based on a dichotomous outcome—pCR or not pCR. Limited sample size may be the most important factor as it limits statistical power to detect moderate effects. Second, variations in the pre-analytic setting (preparation prior to sampling, sampling collection method, sample conservation method, time between sampling and measurement, etc.) may hinder the comparison of the findings between different studies. Numerous studies reviewed here are based on patient records with no possibility to assess the used technic [24,41]. Third, variations in the determination of the cut-off values are present between the studies due to the lack of established cut-off value. This variability may be increased by the fact that some used technologies are still in development (e.g., ctDNA and cfDNA [133,134]) with lack of standardization leading to inconsistencies in their threshold. All these variations can lead to either false negative or positive results. Fourth, for authors using multiple time points, the time at which the second value is measured, post-CRT but pre-surgery, is of critical importance as biomarker clearance dynamic post-CRT is not standardized and, in many cases, not studied [135]. Fifth, heterogeneity in the treatments administrated in these studies can be observed, with variations in radiotherapy (total dose, boost technic, and dose per fraction) and chemotherapy (dosage) schedule, as well as time to surgery, with all these factors being known to influence pCR rate [5,136]. Sixth, confounding factors such as gender, ethnicity, age, concomitant disease, or treatment may influence the measurement of the circulating biomarkers and may induce biases in the results. This a possible explanation for some discrepancies between univariate and multivariate analyses [22,33]. Furthermore, the presence of multivariate analyses does not guarantee that all the confounding factors have been taken into account. Lastly, there is a possible heterogenicity in the tumor response assessment in the listed studies as some use a pathological score (pCR) while other use radiological downstaging. This discrepancy into the assessment method and score can lead to discrepancies between the studies [137].
This review faces several limitations. First, as it is based on studies retrieved the EMBASE dataset, it is possible that studies have been missed during the selection process.
Second, this review focuses on CRT, which is the most common neoadjuvant treatment used. However, other treatment regimens such as short-course radiotherapy (SCRT) or total neoadjuvant therapy (TNT) exist and possibly have different impacts on the tumor or its microenvironment and therefore a different impact on circulating biomarkers. Biomarker studies using these different treatments are soon to be available due to the high interest for TNT and its increased rate of pCR (up to 30%) and for SCRT due to its convenience for patients and similar outcome compared to CRT.
Currently, no single circulating biomarker is able to guide clinical decision. Multimarker models such as the Glasgow Prognostic Score (albumin and CRP) seem to have some potential at predicting tumor response to neoadjuvant CRT [62,138]. Furthermore, others circulating biomarkers (e.g., long non-coding RNA, circulating RNA, or methylated DNA) deserve to be studied [139,140,141].
Ultimately, the prediction accuracy of these models may be improved by the addition of other types of biomarkers associated with the tumor characteristics, tumor microenvironment composition, or the microbiota which could influence it [142,143,144,145,146]. The Immunoscore®—the combination of CD3 and CD8 T-cell densities in the tumor and its invasive margin [147]—was positively correlated with the degree of histologic response after neoadjuvant CRT [148]. More recently Chatila et al. conducted the largest (n = 738) genomic and transcriptomic study looking at determinants of tumor response to neoadjuvant therapy in rectal cancer [149]. If no somatic alterations had significant associations the tumor response, overexpression of IGF2 and L1CAM was associated with decreased response. Furthermore, Chatila et al. discovered a subset of microsatellite-stable tumors with an immune hot transcriptomic profile and increased response [149].

5. Conclusions

Although circulating biomarkers hold great promises, inconsistent results, low statistical power, and low specificity and sensibility prevent them from reliably predicting tumor response to CRT. Further validation and standardization of methods and technologies are required to confirm early results. Ultimately, a multimarker model incorporating circulating as well as tissue (tumor and microenvironment) biomarkers seems to hold the highest promise for the future clinical involvement of circulating biomarker for LARC patients.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/cells12030413/s1; Table S1: Overview of the studies on predictive value of circulating biomarker in LARC; Table S2: Studies exploring CA19-9 as a biomarker predictive of tumor regression following CRT; Table S3: Studies exploring hemoglobin level as a biomarker predictive of tumor regression following CRT; Table S4: Studies exploring albumin or CRP levels as biomarkers predictive of tumor regression following CRT; Table S5: Studies exploring leukocyte and lymphocyte counts as well as NLR, NAR, LMR and PLR ratios as biomarkers predictive of tumor regression following CRT.

Author Contributions

Conceptualization, J.V.M.C., C.C. and T.K.; literature search, J.V.M.C.; analysis and data curation, J.V.M.C.; writing—original draft preparation, J.V.M.C.; writing—review and editing, J.V.M.C., V.D., C.C. and T.K.; supervision, C.C. and T.K. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

We thank Laurence Zulianello (University of Geneva) for providing Figure 1.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Glynne-Jones, R.; Wyrwicz, L.; Tiret, E.; Brown, G.; Rödel, C.; Cervantes, A.; Arnold, D.; Committee, E.G. Rectal cancer: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann. Oncol. 2017, 28, iv22–iv40. [Google Scholar] [CrossRef] [PubMed]
  2. Araghi, M.; Soerjomataram, I.; Jenkins, M.; Brierley, J.; Morris, E.; Bray, F.; Arnold, M. Global trends in colorectal cancer mortality: Projections to the year 2035. Int. J. Cancer 2019, 144, 2992–3000. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Minicozzi, P.; Innos, K.; Sánchez, M.-J.; Trama, A.; Walsh, P.M.; Marcos-Gragera, R.; Dimitrova, N.; Botta, L.; Visser, O.; Rossi, S.; et al. Quality analysis of population-based information on cancer stage at diagnosis across Europe, with presentation of stage-specific cancer survival estimates: A EUROCARE-5 study. Eur. J. Cancer 2017, 84, 335–353. [Google Scholar] [CrossRef]
  4. Benson, A.B.; Venook, A.P.; Al-Hawary, M.M.; Cederquist, L.; Chen, Y.-J.; Ciombor, K.K.; Cohen, S.; Cooper, H.S.; Deming, D.; Engstrom, P.F.; et al. Rectal Cancer, Version 2.2018, NCCN Clinical Practice Guidelines in Oncology. J. Natl. Compr. Cancer Netw. 2018, 16, 874–901. [Google Scholar] [CrossRef] [Green Version]
  5. Macchia, G.; Gambacorta, M.A.; Masciocchi, C.; Chiloiro, G.; Mantello, G.; di Benedetto, M.; Lupattelli, M.; Palazzari, E.; Belgioia, L.; Bacigalupo, A.; et al. Time to surgery and pathologic complete response after neoadjuvant chemoradiation in rectal cancer: A population study on 2094 patients. Clin. Transl. Radiat. Oncol. 2017, 4, 8–14. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Erlandsson, J.; Holm, T.; Pettersson, D.; Berglund, A.; Cedermark, B.; Radu, C.; Johansson, H.; Machado, M.; Hjern, F.; Hallböök, O.; et al. Optimal fractionation of preoperative radiotherapy and timing to surgery for rectal cancer (Stockholm III): A multicentre, randomised, non-blinded, phase 3, non-inferiority trial. Lancet Oncol. 2017, 18, 336–346. [Google Scholar] [CrossRef]
  7. Narang, A.K.; Meyer, J. Neoadjuvant Short-Course Radiation Therapy for Rectal Cancer: Trends and Controversies. Curr. Oncol. Rep. 2018, 20, 68. [Google Scholar] [CrossRef]
  8. Qiaoli, W.; Yongping, H.; Wei, X.; Guoqiang, X.; Yunhe, J.; Qiuyan, L.; Cheng, L.; Mengling, G.; Jiayi, L.; Yi, Y. Preoperative short-course radiotherapy (5 × 5 Gy) with delayed surgery versus preoperative long-course radiotherapy for locally resectable rectal cancer: A meta-analysis. Int. J. Color. Dis. 2019, 34, 2171–2183. [Google Scholar] [CrossRef] [PubMed]
  9. Bahadoer, R.R.; Dijkstra, E.A.; van Etten, B.; Marijnen, C.A.M.; Putter, H.; Kranenbarg, E.M.-K.; Roodvoets, A.G.H.; Nagtegaal, I.D.; Beets-Tan, R.G.H.; Blomqvist, L.K.; et al. Short-course radiotherapy followed by chemotherapy before total mesorectal excision (TME) versus preoperative chemoradiotherapy, TME, and optional adjuvant chemotherapy in locally advanced rectal cancer (RAPIDO): A randomised, open-label, phase 3 trial. Lancet Oncol. 2021, 22, 29–42. [Google Scholar] [CrossRef]
  10. Conroy, T.; Bosset, J.-F.; Etienne, P.-L.; Rio, E.; François, E.; Mesgouez-Nebout, N.; Vendrely, V.; Artignan, X.; Bouché, O.; Gargot, D.; et al. Neoadjuvant chemotherapy with FOLFIRINOX and preoperative chemoradiotherapy for patients with locally advanced rectal cancer (UNICANCER-PRODIGE 23): A multicentre, randomised, open-label, phase 3 trial. Lancet Oncol. 2021, 22, 702–715. [Google Scholar] [CrossRef]
  11. Oronsky, B.; Reid, T.; Larson, C.; Knox, S.J. Locally advanced rectal cancer: The past, present, and future. Semin. Oncol. 2020, 47, 85–92. [Google Scholar] [CrossRef] [PubMed]
  12. Hoendervangers, S.; Couwenberg, A.M.; Intven, M.P.; van Grevenstein, W.M.; Verkooijen, H.M. Comparison of pathological complete response rates after neoadjuvant short-course radiotherapy or chemoradiation followed by delayed surgery in locally advanced rectal cancer. Eur. J. Surg. Oncol. 2018, 44, 1013–1017. [Google Scholar] [CrossRef] [PubMed]
  13. Li, J.-L.; Ji, J.-F.; Cai, Y.; Li, X.-F.; Li, Y.-H.; Wu, H.; Xu, B.; Dou, F.-Y.; Li, Z.-Y.; Bu, Z.-D.; et al. Preoperative concomitant boost intensity-modulated radiotherapy with oral capecitabine in locally advanced mid-low rectal cancer: A phase II trial. Radiother. Oncol. 2012, 102, 4–9. [Google Scholar] [CrossRef] [PubMed]
  14. Petrelli, F.; Trevisan, F.; Cabiddu, M.; Sgroi, G.; Bruschieri, L.; Rausa, E.; Ghidini, M.; Turati, L. Total Neoadjuvant Therapy in Rectal Cancer: A Systematic Review and Meta-analysis of Treatment Outcomes. Ann. Surg. 2020, 271, 440–448. [Google Scholar] [CrossRef]
  15. Chessin, D.B.; Enker, W.; Cohen, A.M.; Paty, P.B.; Weiser, M.R.; Saltz, L.; Minsky, B.D.; Wong, W.D.; Guillem, J.G. Complications after Preoperative Combined Modality Therapy and Radical Resection of Locally Advanced Rectal Cancer: A 14-Year Experience from a Specialty Service. J. Am. Coll. Surg. 2005, 200, 876–882. [Google Scholar] [CrossRef]
  16. Dewdney, A.; Cunningham, D. Toward the Non-surgical Management of Locally Advanced Rectal Cancer. Curr. Oncol. Rep. 2012, 14, 267–276. [Google Scholar] [CrossRef]
  17. Hammarström, S. The carcinoembryonic antigen (CEA) family: Structures, suggested functions and expression in normal and malignant tissues. Semin. Cancer Biol. 1999, 9, 67–81. [Google Scholar] [CrossRef]
  18. Tiernan, J.P.; Perry, S.L.; Verghese, E.T.; West, N.; Yeluri, S.; Jayne, D.G.; Hughes, T. Carcinoembryonic antigen is the preferred biomarker for in vivo colorectal cancer targeting. Br. J. Cancer 2013, 108, 662–667. [Google Scholar] [CrossRef] [Green Version]
  19. Saito, G.; Sadahiro, S.; Kamata, H.; Miyakita, H.; Okada, K.; Tanaka, A.; Suzuki, T. Monitoring of Serum Carcinoembryonic Antigen Levels after Curative Resection of Colon Cancer: Cutoff Values Determined according to Preoperative Levels Enhance the Diagnostic Accuracy for Recurrence. Oncology 2017, 92, 276–282. [Google Scholar] [CrossRef]
  20. Shen, D.; Wang, X.; Wang, H.; Xu, G.; Xie, Y.; Zhuang, Z.; Li, J.; Lin, J.; Wang, P.; Huang, M.; et al. Current Surveillance After Treatment is Not Sufficient for Patients With Rectal Cancer With Negative Baseline CEA. J. Natl. Compr. Cancer Netw. 2022, 20, 653–662.e3. [Google Scholar] [CrossRef]
  21. Engel, R.M.; Oliva, K.; Koulis, C.; Yap, R.; McMurrick, P.J. Predictive factors of complete pathological response in patients with locally advanced rectal cancer. Int. J. Color. Dis. 2020, 35, 1759–1767. [Google Scholar] [CrossRef] [PubMed]
  22. Aires, F.; Rodrigues, D.; Lamas, M.P.; Herdeiro, M.T.; Figueiras, A.; Oliveira, M.J.; Marques, M.; Pinto, A.T. C-Reactive Protein as Predictive Biomarker for Response to Chemoradiotherapy in Patients with Locally Advanced Rectal Cancer: A Retrospective Study. Cancers 2022, 14, 491. [Google Scholar] [CrossRef] [PubMed]
  23. Song, J.; Huang, X.; Chen, Z.; Chen, M.; Lin, Q.; Li, A.; Chen, Y.; Xu, B. Predictive value of carcinoembryonic antigen and carbohydrate antigen 19-9 related to downstaging to stage 0–I after neoadjuvant chemoradiotherapy in locally advanced rectal cancer. Cancer Manag. Res. 2018, 10, 3101–3108. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Jang, N.Y.; Kang, S.-B.; Kim, D.-W.; Kim, J.H.; Lee, K.-W.; Kim, I.A.; Kim, J.-S. The Role of Carcinoembryonic Antigen After Neoadjuvant Chemoradiotherapy in Patients With Rectal Cancer. Dis. Colon Rectum 2011, 54, 245–252. [Google Scholar] [CrossRef] [PubMed]
  25. Heo, J.; Chun, M.; Noh, O.K.; Oh, Y.-T.; Suh, K.W.; Park, J.E.; Cho, O. Sustaining Blood Lymphocyte Count during Preoperative Chemoradiotherapy as a Predictive Marker for Pathologic Complete Response in Locally Advanced Rectal Cancer. Cancer Res. Treat. 2016, 48, 232–239. [Google Scholar] [CrossRef]
  26. Yeo, S.-G.; Kim, D.Y.; Kim, T.H.; Kim, S.Y.; Baek, J.Y.; Chang, H.J.; Park, J.W.; Oh, J.H. Carbohydrate antigen 19-9 levels associated with pathological responses to preoperative chemoradiotherapy in rectal cancer. Asian Pac. J. Cancer Prev. 2014, 15, 5383–5387. [Google Scholar] [CrossRef] [Green Version]
  27. Kitayama, J.; Yasuda, K.; Kawai, K.; Sunami, E.; Nagawa, H. Circulating lymphocyte number has a positive association with tumor response in neoadjuvant chemoradiotherapy for advanced rectal cancer. Radiat. Oncol. 2010, 5, 47. [Google Scholar] [CrossRef] [Green Version]
  28. Kim, H.J.; Choi, G.-S.; Park, J.S.; Park, S.; Kawai, K.; Watanabe, T. Clinical Significance of Thrombocytosis Before Preoperative Chemoradiotherapy in Rectal Cancer: Predicting Pathologic Tumor Response and Oncologic Outcome. Ann. Surg. Oncol. 2015, 22, 513–519. [Google Scholar] [CrossRef]
  29. Choi, E.; Kim, J.H.; Kim, O.B.; Kim, M.Y.; Oh, Y.K.; Baek, S.G. Predictors of pathologic complete response after preoperative concurrent chemoradiotherapy of rectal cancer: A single center experience. Radiat. Oncol. J. 2016, 34, 106–112. [Google Scholar] [CrossRef]
  30. Yang, J.; Ling, X.; Tang, W.; Hu, D.; Zhou, H.; Yin, G. Analyses of predictive factors for pathological complete remission in neoadjuvant therapy for locally advanced rectal cancer. J. B. U. ON. 2019, 24, 77–83. [Google Scholar]
  31. Cheong, C.; Shin, J.S.; Suh, K.W. Prognostic value of changes in serum carcinoembryonic antigen levels for preoperative chemoradiotherapy response in locally advanced rectal cancer. World J. Gastroenterol. 2020, 26, 7022–7035. [Google Scholar] [CrossRef] [PubMed]
  32. Huang, C.-M.; Huang, C.-W.; Ma, C.-J.; Yeh, Y.-S.; Su, W.-C.; Chang, T.-K.; Tsai, H.-L.; Juo, S.-H.; Huang, M.-Y.; Wang, J.-Y. Predictive Value of FOLFOX-Based Regimen, Long Interval, Hemoglobin Levels and Clinical Negative Nodal Status, and Postchemoradiotherapy CEA Levels for Pathological Complete Response in Patients with Locally Advanced Rectal Cancer after Neoadjuvant Chemoradiotherapy. J. Oncol. 2020, 2020, 9437684. [Google Scholar] [CrossRef] [PubMed]
  33. Gago, T.; Caldeira, P.; Cunha, C.; Campelo, P.; Guerreiro, H. Can we optimize the CEA as a response marker in rectal cancer? Rev. Esp. Enferm. Dig. 2020, 113, 423–428. [Google Scholar] [CrossRef] [PubMed]
  34. Guo, S.-P.; Chen, C.; Zeng, Z.-F.; Wang, Q.-X.; Jiang, W.; Gao, Y.-H.; Chang, H. Serum Apolipoprotein A-I Predicts Response of Rectal Cancer to Neoadjuvant Chemoradiotherapy. Cancer Manag. Res. 2021, 13, 2623–2631. [Google Scholar] [CrossRef]
  35. Zhang, Q.; Liang, J.; Chen, J.; Mei, S.; Wang, Z. Predictive Factors for Pathologic Complete Response Following Neoadjuvant Chemoradiotherapy for Rectal Cancer. Asian Pac. J. Cancer Prev. 2021, 22, 1607–1611. [Google Scholar] [CrossRef]
  36. Wada, Y.; Shimada, M.; Morine, Y.; Ikemoto, T.; Saito, Y.; Zhu, Z.; Wang, X.; Etxart, A.; Park, Y.; Bujanda, L.; et al. Circulating miRNA Signature Predicts Response to Preoperative Chemoradiotherapy in Locally Advanced Rectal Cancer. JCO Precis. Oncol. 2021, 5, 1788–1801. [Google Scholar] [CrossRef]
  37. Murahashi, S.; Akiyoshi, T.; Sano, T.; Fukunaga, Y.; Noda, T.; Ueno, M.; Zembutsu, H. Serial circulating tumour DNA analysis for locally advanced rectal cancer treated with preoperative therapy: Prediction of pathological response and postoperative recurrence. Br. J. Cancer 2020, 123, 803–810. [Google Scholar] [CrossRef] [PubMed]
  38. Sawada, R.; Akiyoshi, T.; Kitagawa, Y.; Hiyoshi, Y.; Mukai, T.; Nagasaki, T.; Yamaguchi, T.; Konishi, T.; Yamamoto, N.; Ueno, M.; et al. Systemic Inflammatory Markers Combined with Tumor-Infiltrating Lymphocyte Density for the Improved Prediction of Response to Neoadjuvant Chemoradiotherapy in Rectal Cancer. Ann. Surg. Oncol. 2021, 28, 6189–6198. [Google Scholar] [CrossRef] [PubMed]
  39. Cai, Z.; Huang, L.; Chen, Y.; Xie, X.; Zou, Y.; Lan, P.; Wu, X. CEA Decline Predicts Tumor Regression and Prognosis in Locally Advanced Rectal Cancer Patients with Elevated Baseline CEA. J. Cancer 2020, 11, 6565–6570. [Google Scholar] [CrossRef] [PubMed]
  40. Yang, K.-L.; Yang, S.-H.; Liang, W.-Y.; Kuo, Y.-J.; Lin, J.-K.; Lin, T.-C.; Chen, W.-S.; Jiang, J.-K.; Wang, H.-S.; Chang, S.-C.; et al. Carcinoembryonic antigen (CEA) level, CEA ratio, and treatment outcome of rectal cancer patients receiving pre-operative chemoradiation and surgery. Radiat. Oncol. 2013, 8, 43. [Google Scholar] [CrossRef] [Green Version]
  41. Tawfik, B.; Mokdad, A.A.; Patel, P.M.; Li, H.C.; Huerta, S. The neutrophil to albumin ratio as a predictor of pathological complete response in rectal cancer patients following neoadjuvant chemoradiation. Anti-Cancer Drugs 2016, 27, 879–883. [Google Scholar] [CrossRef] [PubMed]
  42. Restivo, A.; Zorcolo, L.; Cocco, I.M.F.; Manunza, R.; Margiani, C.; Marongiu, L.; Casula, G. Elevated CEA Levels and Low Distance of the Tumor from the Anal Verge are Predictors of Incomplete Response to Chemoradiation in Patients with Rectal Cancer. Ann. Surg. Oncol. 2013, 20, 864–871. [Google Scholar] [CrossRef] [PubMed]
  43. Hu, H.; Huang, J.; Lan, P.; Wang, L.; Huang, M.; Wang, J.; Deng, Y. CEA clearance pattern as a predictor of tumor response to neoadjuvant treatment in rectal cancer: A post-hoc analysis of FOWARC trial. BMC Cancer 2018, 18, 1145. [Google Scholar] [CrossRef]
  44. Deng, Y.; Chi, P.; Lan, P.; Wang, L.; Chen, W.; Cui, L.; Chen, D.; Cao, J.; Wei, H.; Peng, X.; et al. Modified FOLFOX6 With or Without Radiation Versus Fluorouracil and Leucovorin With Radiation in Neoadjuvant Treatment of Locally Advanced Rectal Cancer: Initial Results of the Chinese FOWARC Multicenter, Open-Label, Randomized Three-Arm Phase III Trial. J. Clin. Oncol. 2016, 34, 3300–3307. [Google Scholar] [CrossRef]
  45. Scarà, S.; Bottoni, P.; Scatena, R. CA 19-9: Biochemical and Clinical Aspects. In Advances in Cancer Biomarkers: From Biochemistry to Clinic for a Critical Revision; Scatena, R., Ed.; Springer: Dordrecht, Netherlands, 2015; pp. 247–260. [Google Scholar]
  46. Lee, T.; Teng, T.Z.J.; Shelat, V.G. Carbohydrate antigen 19-9—tumor marker: Past, present, and future. World J. Gastrointest. Surg. 2020, 12, 468–490. [Google Scholar] [CrossRef]
  47. Goonetilleke, K.S.; Siriwardena, A.K. Systematic review of carbohydrate antigen (CA 19-9) as a biochemical marker in the diagnosis of pancreatic cancer. Eur. J. Surg. Oncol. 2007, 33, 266–270. [Google Scholar] [CrossRef] [PubMed]
  48. Ducreux, M.; Cuhna, A.S.; Caramella, C.; Hollebecque, A.; Burtin, P.; Goéré, D.; Seufferlein, T.; Haustermans, K.; Van Laethem, J.L.; Conroy, T.; et al. Cancer of the pancreas: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann. Oncol. 2015, 26 (Suppl. S5), v56–v68. [Google Scholar] [CrossRef] [PubMed]
  49. Buergy, D.; Wenz, F.; Groden, C.; Brockmann, M.A. Tumor-platelet interaction in solid tumors. Int. J. Cancer 2012, 130, 2747–2760. [Google Scholar] [CrossRef]
  50. Brown, K.M.; Domin, C.; Aranha, G.V.; Yong, S.; Shoup, M. Increased preoperative platelet count is associated with decreased survival after resection for adenocarcinoma of the pancreas. Am. J. Surg. 2005, 189, 278–282. [Google Scholar] [CrossRef]
  51. Tomita, M.; Shimizu, T.; Hara, M.; Ayabe, T.; Onitsuka, T. Prognostic impact of thrombocytosis in resectable non-small cell lung cancer. Interact. Cardiovasc. Thorac. Surg. 2008, 7, 613–615. [Google Scholar] [CrossRef] [Green Version]
  52. Krauthamer, M.; Rouvinov, K.; Ariad, S.; Man, S.; Walfish, S.; Pinsk, I.; Sztarker, I.; Charkovsky, T.; Lavrenkov, K. A Study of Inflammation-Based Predictors of Tumor Response to Neoadjuvant Chemoradiotherapy for Locally Advanced Rectal Cancer. Oncology 2013, 85, 27–32. [Google Scholar] [CrossRef] [PubMed]
  53. Robnett, T.J.; Machtay, M.; Hahn, S.M.; Shrager, J.B.; Friedberg, J.S.; Kaiser, L.R. Pathological Response to Preoperative Chemoradiation Worsens with Anemia in Non---Small Cell Lung Cancer Patients. Cancer J. 2002, 8, 263–267. [Google Scholar] [CrossRef] [PubMed]
  54. Rakotomalala, A.; Escande, A.; Furlan, A.; Meignan, S.; Lartigau, E. Hypoxia in Solid Tumors: How Low Oxygenation Impacts the “Six Rs” of Radiotherapy. Front. Endocrinol. 2021, 12, 742215. [Google Scholar] [CrossRef] [PubMed]
  55. Pakravan, N. Tumorigenesis: Cell defense against hypoxia? Oncol. Rev. 2013, 7, e1. [Google Scholar] [CrossRef] [Green Version]
  56. Muz, B.; de la Puente, P.; Azab, F.; Azab, A.K. The role of hypoxia in cancer progression, angiogenesis, metastasis, and resistance to therapy. Hypoxia 2015, 3, 83–92. [Google Scholar] [CrossRef] [Green Version]
  57. Harrison, L.B.; Chadha, M.; Hill, R.J.; Hu, K.; Shasha, D. Impact of Tumor Hypoxia and Anemia on Radiation Therapy Outcomes. Oncologist 2002, 7, 492–508. [Google Scholar] [CrossRef]
  58. Berardi, R.; Braconi, C.; Mantello, G.; Scartozzi, M.; Del Prete, S.; Luppi, G.; Martinelli, R.; Fumagalli, M.; Valeri, G.; Bearzi, I.; et al. Anemia may influence the outcome of patients undergoing neo-adjuvant treatment of rectal cancer. Ann. Oncol. 2006, 17, 1661–1664. [Google Scholar] [CrossRef]
  59. Hanahan, D.; Weinberg, R.A. Hallmarks of cancer: The next generation. Cell 2011, 144, 646–674. [Google Scholar] [CrossRef] [Green Version]
  60. Greten, F.R.; Grivennikov, S.I. Inflammation and Cancer: Triggers, Mechanisms, and Consequences. Immunity 2019, 51, 27–41. [Google Scholar] [CrossRef]
  61. Jung, S.W.; Park, I.J.; Oh, S.H.; Yeom, S.-S.; Lee, J.L.; Yoon, Y.S.; Kim, C.W.; Lim, S.-B.; Lee, J.B.; Yu, C.S.; et al. Association of immunologic markers from complete blood counts with the response to preoperative chemoradiotherapy and prognosis in locally advanced rectal cancer. Oncotarget 2017, 8, 59757–59765. [Google Scholar] [CrossRef] [Green Version]
  62. Dreyer, S.B.; Powell, A.G.M.T.; McSorley, S.T.; Waterston, A.; Going, J.J.; Edwards, J.; McMillan, D.C.; Horgan, P.G. The Pretreatment Systemic Inflammatory Response is an Important Determinant of Poor Pathologic Response for Patients Undergoing Neoadjuvant Therapy for Rectal Cancer. Ann. Surg. Oncol. 2017, 24, 1295–1303. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Moman, R.N.; Gupta, N.; Varacallo, M. Physiology, Albumin; StatPearls Publishing: Treasure Island, FL, USA, 2022. [Google Scholar]
  64. Akirov, A.; Masri-Iraqi, H.; Atamna, A.; Shimon, I. Low Albumin Levels Are Associated with Mortality Risk in Hospitalized Patients. Am. J. Med. 2017, 130, 1465.e11–1465.e19. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Caraceni, P.; Tufoni, M.; Bonavita, M.E. Clinical use of albumin. Blood Transfus. 2013, 11 (Suppl. S4), s18–s25. [Google Scholar] [CrossRef] [PubMed]
  66. Sproston, N.R.; Ashworth, J.J. Role of C-Reactive Protein at Sites of Inflammation and Infection. Front. Immunol. 2018, 9, 754. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Volanakis, J.E. Human C-reactive protein: Expression, structure, and function. Mol. Immunol. 2001, 38, 189–197. [Google Scholar] [CrossRef] [PubMed]
  68. Xiao, X.; Wang, S.; Long, G. C-reactive protein is a significant predictor of improved survival in patients with advanced non-small cell lung cancer. Medicine 2019, 98, e16238. [Google Scholar] [CrossRef] [PubMed]
  69. Okugawa, Y.; Toiyama, Y.; Yamamoto, A.; Shigemori, T.; Ide, S.; Kitajima, T.; Fujikawa, H.; Yasuda, H.; Hiro, J.; Yoshiyama, S.; et al. Lymphocyte-C-reactive Protein Ratio as Promising New Marker for Predicting Surgical and Oncological Outcomes in Colorectal Cancer. Ann. Surg. 2020, 272, 342–351. [Google Scholar] [CrossRef]
  70. Petrelli, F.; Barni, S.; Coinu, A.; Bertocchi, P.; Borgonovo, K.; Cabiddu, M.; Ghilardi, M.; Zaniboni, A. The Modified Glasgow Prognostic Score and Survival in Colorectal Cancer: A Pooled Analysis of the Literature. Rev. Recent Clin. Trials 2015, 10, 135–141. [Google Scholar] [CrossRef]
  71. Choi, K.W.; Hong, S.W.; Chang, Y.G.; Lee, W.Y.; Lee, B.; Paik, I.W.; Lee, H. Inflammation-based score (Glasgow prognostic score) as an independent prognostic factor in colorectal cancer patients. Ann. Surg. Treat. Res. 2014, 86, 309–313. [Google Scholar] [CrossRef] [Green Version]
  72. Tong, T.; Guan, Y.; Xiong, H.; Wang, L.; Pang, J. A Meta-Analysis of Glasgow Prognostic Score and Modified Glasgow Prognostic Score as Biomarkers for Predicting Survival Outcome in Renal Cell Carcinoma. Front. Oncol. 2020, 10, 1541. [Google Scholar] [CrossRef]
  73. Hu, X.; Wang, Y.; Yang, W.-X.; Dou, W.-C.; Shao, Y.-X.; Li, X. Modified Glasgow prognostic score as a prognostic factor for renal cell carcinomas: A systematic review and meta-analysis. Cancer Manag. Res. 2019, 11, 6163–6173. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Zahorec, R. Neutrophil-to-lymphocyte ratio, past, present and future perspectives. Bratisl Lek Listy 2021, 122, 474–488. [Google Scholar] [CrossRef] [PubMed]
  75. Bowen, R.C.; Little, N.A.B.; Harmer, J.R.; Ma, J.; Mirabelli, L.G.; Roller, K.D.; Breivik, A.M.; Signor, E.; Miller, A.B.; Khong, H.T. Neutrophil-to-lymphocyte ratio as prognostic indicator in gastrointestinal cancers: A systematic review and meta-analysis. Oncotarget 2017, 8, 32171–32189. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Guthrie, G.J.K.; Charles, K.A.; Roxburgh, C.S.D.; Horgan, P.G.; McMillan, D.C.; Clarke, S.J. The systemic inflammation-based neutrophil–lymphocyte ratio: Experience in patients with cancer. Crit. Rev. Oncol. Hematol. 2013, 88, 218–230. [Google Scholar] [CrossRef]
  77. Ferro, M.; Babă, D.-F.; de Cobelli, O.; Musi, G.; Lucarelli, G.; Terracciano, D.; Porreca, A.; Busetto, G.M.; Del Giudice, F.; Soria, F.; et al. Neutrophil percentage-to-albumin ratio predicts mortality in bladder cancer patients treated with neoadjuvant chemotherapy followed by radical cystectomy. Future Sci. OA 2021, 7, FSO709. [Google Scholar] [CrossRef]
  78. Li, R.; Sun, Z.; Song, S.; He, X.; Shi, X.; Li, Z.; Song, J. NARFIB: A Novel Prognostic Score Based on the Neutrophil-to-Albumin Ratio and Fibrinogen Can Predict the Prognosis of Gastrointestinal Stromal Tumors. Cancer Manag. Res. 2020, 12, 11183–11190. [Google Scholar] [CrossRef]
  79. Tingle, S.J.; Ma, G.R.S.; Goodfellow, M.; Moir, J.A.; White, S.A. NARCA: A novel prognostic scoring system using neutrophil-albumin ratio and Ca19-9 to predict overall survival in palliative pancreatic cancer. J. Surg. Oncol. 2018, 118, 680–686. [Google Scholar] [CrossRef]
  80. Goto, W.; Kashiwagi, S.; Asano, Y.; Takada, K.; Takahashi, K.; Hatano, T.; Takashima, T.; Tomita, S.; Motomura, H.; Hirakawa, K.; et al. Predictive value of lymphocyte-to-monocyte ratio in the preoperative setting for progression of patients with breast cancer. BMC Cancer 2018, 18, 1137. [Google Scholar] [CrossRef]
  81. Hu, P.; Shen, H.; Wang, G.; Zhang, P.; Liu, Q.; Du, J. Prognostic Significance of Systemic Inflammation-Based Lymphocyte- Monocyte Ratio in Patients with Lung Cancer: Based on a Large Cohort Study. PLoS ONE 2014, 9, e108062. [Google Scholar] [CrossRef]
  82. Templeton, A.J.; Ace, O.; McNamara, M.G.; Al-Mubarak, M.; Vera-Badillo, F.E.; Hermanns, T.; Šeruga, B.; Ocaña, A.; Tannock, I.F.; Amir, E. Prognostic Role of Platelet to Lymphocyte Ratio in Solid Tumors: A Systematic Review and Meta-Analysis. Cancer Epidemiol. Biomark. Prev. 2014, 23, 1204–1212. [Google Scholar] [CrossRef] [Green Version]
  83. Takamizawa, Y.; Shida, D.; Boku, N.; Nakamura, Y.; Ahiko, Y.; Yoshida, T.; Tanabe, T.; Takashima, A.; Kanemitsu, Y. Nutritional and inflammatory measures predict survival of patients with stage IV colorectal cancer. BMC Cancer 2020, 20, 1092. [Google Scholar] [CrossRef] [PubMed]
  84. Liu, R.; Huang, Y. Lipid Signaling in Tumorigenesis. Mol. Cell. Pharmacol. 2014, 6, 1–9. [Google Scholar] [PubMed]
  85. van Meeteren, L.A.; Moolenaar, W.H. Regulation and biological activities of the autotaxin–LPA axis. Prog. Lipid Res. 2007, 46, 145–160. [Google Scholar] [CrossRef] [PubMed]
  86. Corn, K.C.; Windham, M.A.; Rafat, M. Lipids in the tumor microenvironment: From cancer progression to treatment. Prog. Lipid Res. 2020, 80, 101055. [Google Scholar] [CrossRef]
  87. Reimers, N.; Pantel, K. Liquid biopsy: Novel technologies and clinical applications. Clin. Chem. Lab. Med. 2019, 57, 312–316. [Google Scholar] [CrossRef] [PubMed]
  88. Pantel, K.; Alix-Panabières, C. Circulating tumour cells in cancer patients: Challenges and perspectives. Trends Mol. Med. 2010, 16, 398–406. [Google Scholar] [CrossRef] [PubMed]
  89. Alix-Panabières, C.; Pantel, K. Liquid Biopsy: From Discovery to Clinical Application. Cancer Discov. 2021, 11, 858–873. [Google Scholar] [CrossRef]
  90. Morais, M.; Pinto, D.M.; Machado, J.C.; Carneiro, S. ctDNA on liquid biopsy for predicting response and prognosis in locally advanced rectal cancer: A systematic review. Eur. J. Surg. Oncol. 2021, 48, 218–227. [Google Scholar] [CrossRef]
  91. De Palma, F.D.E.; Luglio, G.; Tropeano, F.P.; Pagano, G.; D’Armiento, M.; Kroemer, G.; Maiuri, M.C.; De Palma, G.D. The Role of Micro-RNAs and Circulating Tumor Markers as Predictors of Response to Neoadjuvant Therapy in Locally Advanced Rectal Cancer. Int. J. Mol. Sci. 2020, 21, 7040. [Google Scholar] [CrossRef]
  92. Mandel, P.; Metais, P. Nuclear Acids In Human Blood Plasma. C. R. Seances Soc. Biol. Fil. 1948, 142, 241–243. [Google Scholar]
  93. Duvvuri, B.; Lood, C. Cell-Free DNA as a Biomarker in Autoimmune Rheumatic Diseases. Front. Immunol. 2019, 10, 502. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Fettke, H.; Kwan, E.M.; Azad, A.A. Cell-free DNA in cancer: Current insights. Cell. Oncol. 2019, 42, 13–28. [Google Scholar] [CrossRef] [PubMed]
  95. Nagata, S. Apoptotic DNA Fragmentation. Exp. Cell Res. 2000, 256, 12–18. [Google Scholar] [CrossRef] [PubMed]
  96. Fleischhacker, M.; Schmidt, B. Circulating nucleic acids (CNAs) and cancer—A survey. Biochim. Biophys. Acta 2007, 1775, 181–232. [Google Scholar] [CrossRef] [PubMed]
  97. Bronkhorst, A.J.; Wentzel, J.F.; Aucamp, J.; van Dyk, E.; du Plessis, L.; Pretorius, P.J. Characterization of the cell-free DNA released by cultured cancer cells. Biochim. Biophys. Acta 2016, 1863, 157–165. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  98. Lo, Y.M.D.; Zhang, J.; Leung, T.N.; Lau, T.K.; Chang, A.M.; Hjelm, N.M. Rapid Clearance of Fetal DNA from Maternal Plasma. Am. J. Hum. Genet. 1999, 64, 218–224. [Google Scholar] [CrossRef] [Green Version]
  99. Bryzgunova, O.E.; Laktionov, P.P. Extracellular Nucleic Acids in Urine: Sources, Structure, Diagnostic Potential. Acta Nat. 2015, 7, 48–54. [Google Scholar] [CrossRef] [Green Version]
  100. Zitt, M.; Müller, H.M.; Rochel, M.; Schwendinger, V.; Zitt, M.; Goebel, G.; DeVries, A.; Margreiter, R.; Oberwalder, M.; Zeillinger, R.; et al. Circulating Cell-Free DNA in Plasma of Locally Advanced Rectal Cancer Patients Undergoing Preoperative Chemoradiation: A Potential Diagnostic Tool for Therapy Monitoring. Dis. Markers 2008, 25, 159–165. [Google Scholar] [CrossRef] [Green Version]
  101. Agostini, M.; Pucciarelli, S.; Enzo, M.V.; Del Bianco, P.; Briarava, M.; Bedin, C.; Maretto, I.; Friso, M.L.; Lonardi, S.; Mescoli, C.; et al. Circulating Cell-Free DNA: A Promising Marker of Pathologic Tumor Response in Rectal Cancer Patients Receiving Preoperative Chemoradiotherapy. Ann. Surg. Oncol. 2011, 18, 2461–2468. [Google Scholar] [CrossRef]
  102. Sun, W.; Sun, Y.; Zhu, M.; Wang, Z.; Zhang, H.; Xin, Y.; Jiang, G.; Guo, X.; Zhang, Z.; Liu, Y. The role of plasma cell-free DNA detection in predicting preoperative chemoradiotherapy response in rectal cancer patients. Oncol. Rep. 2014, 31, 1466–1472. [Google Scholar] [CrossRef] [Green Version]
  103. Schou, J.; Larsen, F.; Sørensen, B.; Abrantes, R.; Boysen, A.; Johansen, J.; Jensen, B.; Nielsen, D.; Spindler, K. Circulating cell-free DNA as predictor of treatment failure after neoadjuvant chemo-radiotherapy before surgery in patients with locally advanced rectal cancer. Ann. Oncol. 2018, 29, 610–615. [Google Scholar] [CrossRef]
  104. Shalaby, S.M.; El-Shal, A.S.; Abdelaziz, L.A.; Abd-Elbary, E.; Khairy, M.M. Promoter methylation and expression of DNA repair genes MGMT and ERCC1 in tissue and blood of rectal cancer patients. Gene 2018, 644, 66–73. [Google Scholar] [CrossRef] [PubMed]
  105. Wang, B.; Huang, H.-Y.; Chen, Y.-C.; Bristow, R.E.; Kassauei, K.; Cheng, C.-C.; Roden, R.; Sokoll, L.J.; Chan, D.W.; Shih, I.-M. Increased plasma DNA integrity in cancer patients. Cancer Res 2003, 63, 3966–3968. [Google Scholar] [PubMed]
  106. Stroun, M.; Anker, P.; Maurice, P.; Lyautey, J.; Lederrey, C.; Beljanski, M. Neoplastic Characteristics of the DNA Found in the Plasma of Cancer Patients. Oncology 1989, 46, 318–322. [Google Scholar] [CrossRef] [PubMed]
  107. Volik, S.; Alcaide, M.; Morin, R.D.; Collins, C.C. Cell-free DNA (cfDNA): Clinical Significance and Utility in Cancer Shaped By Emerging Technologies. Mol. Cancer Res. 2016, 14, 898–908. [Google Scholar] [CrossRef] [Green Version]
  108. Pazdirek, F.; Minarik, M.; Benesova, L.; Halkova, T.; Belsanova, B.; Macek, M.; Stepanek, L.; Hoch, J. Monitoring of Early Changes of Circulating Tumor DNA in the Plasma of Rectal Cancer Patients Receiving Neoadjuvant Concomitant Chemoradiotherapy: Evaluation for Prognosis and Prediction of Therapeutic Response. Front. Oncol. 2020, 10, 1028. [Google Scholar] [CrossRef]
  109. Carpinetti, P.; Donnard, E.; Bettoni, F.; Asprino, P.; Koyama, F.; Rozanski, A.; Sabbaga, J.; Habr-Gama, A.; Parmigiani, R.B.; Galante, P.A.; et al. The use of personalized biomarkers and liquid biopsies to monitor treatment response and disease recurrence in locally advanced rectal cancer after neoadjuvant chemoradiation. Oncotarget 2015, 6, 38360–38371. [Google Scholar] [CrossRef] [Green Version]
  110. Zhou, J.; Wang, C.; Lin, G.; Xiao, Y.; Jia, W.; Xiao, G.; Liu, Q.; Wu, B.; Wu, A.; Qiu, H.; et al. Serial Circulating Tumor DNA in Predicting and Monitoring the Effect of Neoadjuvant Chemoradiotherapy in Patients with Rectal Cancer: A Prospective Multicenter Study. Clin. Cancer Res. 2021, 27, 301–310. [Google Scholar] [CrossRef]
  111. McDuff, S.G.R.; Hardiman, K.M.; Ulintz, P.J.; Parikh, A.R.; Zheng, H.; Kim, D.W.; Lennerz, J.K.; Hazar-Rethinam, M.; Van Seventer, E.E.; Fetter, I.J.; et al. Circulating Tumor DNA Predicts Pathologic and Clinical Outcomes Following Neoadjuvant Chemoradiation and Surgery for Patients With Locally Advanced Rectal Cancer. JCO Precis. Oncol. 2021, 5, 123–132. [Google Scholar] [CrossRef]
  112. Khakoo, S.; Carter, P.D.; Brown, G.; Valeri, N.; Picchia, S.; Bali, M.A.; Shaikh, R.; Jones, T.; Begum, R.; Rana, I.; et al. MRI Tumor Regression Grade and Circulating Tumor DNA as Complementary Tools to Assess Response and Guide Therapy Adaptation in Rectal Cancer. Clin. Cancer Res. 2020, 26, 183–192. [Google Scholar] [CrossRef]
  113. Sclafani, F.; Chau, I.; Cunningham, D.; Hahne, J.C.; Vlachogiannis, G.; Eltahir, Z.; Lampis, A.; Braconi, C.; Kalaitzaki, E.; De Castro, D.G.; et al. KRAS and BRAF mutations in circulating tumour DNA from locally advanced rectal cancer. Sci. Rep. 2018, 8, 1445. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Appelt, A.L.; Andersen, R.F.; Lindebjerg, J.; Jakobsen, A. Prognostic Value of Serum NPY Hypermethylation in Neoadjuvant Chemoradiotherapy for Rectal Cancer: Secondary Analysis of a Randomized Trial. Am. J. Clin. Oncol. 2020, 43, 9–13. [Google Scholar] [CrossRef] [PubMed]
  115. Tie, J.; Cohen, J.D.; Wang, Y.; Li, L.; Christie, M.; Simons, K.; Elsaleh, H.; Kosmider, S.; Wong, R.; Yip, D.; et al. Serial circulating tumour DNA analysis during multimodality treatment of locally advanced rectal cancer: A prospective biomarker study. Gut 2019, 68, 663–671. [Google Scholar] [CrossRef]
  116. Ying, W.; Gao, H.; Dos Reis, F.C.G.; Bandyopadhyay, G.; Ofrecio, J.M.; Luo, Z.; Ji, Y.; Jin, Z.; Ly, C.; Olefsky, J.M. MiR-690, an exosomal-derived miRNA from M2-polarized macrophages, improves insulin sensitivity in obese mice. Cell Metab. 2021, 33, 781–790.e5. [Google Scholar] [CrossRef] [PubMed]
  117. Fabbri, M.; Paone, A.; Calore, F.; Galli, R.; Gaudio, E.; Santhanam, R.; Lovat, F.; Fadda, P.; Mao, C.; Nuovo, G.J.; et al. MicroRNAs bind to Toll-like receptors to induce prometastatic inflammatory response. Proc. Natl. Acad. Sci. USA 2012, 109, E2110–E2116. [Google Scholar] [CrossRef] [Green Version]
  118. Wu, Y.; Li, Q.; Zhang, R.; Dai, X.; Chen, W.; Xing, D. Circulating microRNAs: Biomarkers of disease. Clin. Chim. Acta 2021, 516, 46–54. [Google Scholar] [CrossRef]
  119. Peng, Y.; Croce, C.M. The role of MicroRNAs in human cancer. Signal Transduct. Target. Ther. 2016, 1, 15004. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  120. Dilsiz, N. Role of exosomes and exosomal microRNAs in cancer. Future Sci. OA 2020, 6, FSO465. [Google Scholar] [CrossRef] [Green Version]
  121. Bhome, R.; Del Vecchio, F.; Lee, G.-H.; Bullock, M.D.; Primrose, J.N.; Sayan, A.E.; Mirnezami, A.H. Exosomal microRNAs (exomiRs): Small molecules with a big role in cancer. Cancer Lett. 2018, 420, 228–235. [Google Scholar] [CrossRef]
  122. Svoboda, M.; Holla, L.I.; Sefr, R.; Vrtkova, I.; Kocakova, I.; Tichy, B.; Dvorak, J. Micro-RNAs miR125b and miR137 are frequently upregulated in response to capecitabine chemoradiotherapy of rectal cancer. Int. J. Oncol. 2008, 33, 541–547. [Google Scholar] [CrossRef] [Green Version]
  123. Della Vittoria Scarpati, G.; Falcetta, F.; Carlomagno, C.; Ubezio, P.; Marchini, S.; De Stefano, A.; Singh, V.K.; D’Incalci, M.; De Placido, S.; 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. 2012, 83, 1113–1119. [Google Scholar] [CrossRef] [PubMed]
  124. Kheirelseid, E.A.H.; Miller, N.; Chang, K.H.; Curran, C.; Hennessey, E.; Sheehan, M.; Newell, J.; Lemetre, C.; Balls, G.; Kerin, M.J. miRNA expressions in rectal cancer as predictors of response to neoadjuvant chemoradiation therapy. Int. J. Color. Dis. 2013, 28, 247–260. [Google Scholar] [CrossRef] [PubMed]
  125. Dreussi, E.; Pucciarelli, S.; De Paoli, A.; Polesel, J.; Canzonieri, V.; Agostini, M.; Friso, M.L.; 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 2016, 7, 19781–19793. [Google Scholar] [CrossRef] [PubMed]
  126. D’Angelo, E.; Fassan, M.; Maretto, I.; Pucciarelli, S.; Zanon, C.; Digito, M.; Rugge, M.; Nitti, D.; Agostini, M. Serum miR-125b is a non-invasive predictive biomarker of the pre-operative chemoradiotherapy responsiveness in patients with rectal adenocarcinoma. Oncotarget 2016, 7, 28647–28657. [Google Scholar] [CrossRef] [PubMed]
  127. 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 chemoradiotherapy in locally advanced rectal cancer. Oncotarget 2016, 7, 64233–64243. [Google Scholar] [CrossRef] [Green Version]
  128. Azizian, A.; Kramer, F.; Jo, P.; Wolff, H.A.; Beißbarth, T.; Skarupke, R.; Bernhardt, M.; Grade, M.; Ghadimi, B.M.; Gaedcke, J. Preoperative Prediction of Lymph Node Status by Circulating Mir-18b and Mir-20a During Chemoradiotherapy in Patients with Rectal Cancer. World J. Surg. 2015, 39, 2329–2335. [Google Scholar] [CrossRef]
  129. Meltzer, S.; Bjørnetrø, T.; Lyckander, L.G.; Flatmark, K.; Dueland, S.; Samiappan, R.; Johansen, C.; Kalanxhi, E.; Ree, A.H.; Redalen, K.R. Circulating Exosomal miR-141-3p and miR-375 in Metastatic Progression of Rectal Cancer. Transl. Oncol. 2019, 12, 1038–1044. [Google Scholar] [CrossRef]
  130. Baek, D.W.; Kim, G.; Kang, B.W.; Kim, H.J.; Park, S.Y.; Park, J.S.; Choi, G.-S.; Kang, M.K.; Hur, K.; Kim, J.G. High expression of microRNA-199a-5p is associated with superior clinical outcomes in patients with locally advanced rectal cancer. J. Cancer Res. Clin. Oncol. 2020, 146, 105–115. [Google Scholar] [CrossRef]
  131. Hiyoshi, Y.; Akiyoshi, T.; Inoue, R.; Murofushi, K.; Yamamoto, N.; Fukunaga, Y.; Ueno, M.; Baba, H.; Mori, S.; Yamaguchi, T. Serum miR-143 levels predict the pathological response to neoadjuvant chemoradiotherapy in patients with locally advanced rectal cancer. Oncotarget 2017, 8, 79201–79211. [Google Scholar] [CrossRef] [Green Version]
  132. Bartkowiak, K.; Koch, C.; Gärtner, S.; Andreas, A.; Gorges, T.M.; Pantel, K. In Vitro Modeling of Reoxygenation Effects on mRNA and Protein Levels in Hypoxic Tumor Cells upon Entry into the Bloodstream. Cells 2020, 9, 1316. [Google Scholar] [CrossRef]
  133. Diehl, F.; Li, M.; Dressman, D.; He, Y.; Shen, D.; Szabo, S.; Diaz, L.A.; Goodman, S.N.; David, K.A.; Juhl, H.; et al. Detection and quantification of mutations in the plasma of patients with colorectal tumors. Proc. Natl. Acad. Sci. USA 2005, 102, 16368–16373. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. Chaudhuri, A.A.; Binkley, M.S.; Osmundson, E.C.; Alizadeh, A.A.; Diehn, M. Predicting Radiotherapy Responses and Treatment Outcomes Through Analysis of Circulating Tumor DNA. Semin. Radiat. Oncol. 2015, 25, 305–312. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. Glimelius, B. On a prolonged interval between rectal cancer (chemo)radiotherapy and surgery. Upsala J. Med. Sci. 2017, 122, 1–10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  136. Glimelius, B. What is most relevant in preoperative rectal cancer chemoradiotherapy—the chemotherapy, the radiation dose or the timing to surgery? Acta Oncol. 2016, 55, 1381–1385. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  137. Nagtegaal, I.D.; Glynne-Jones, R. How to measure tumour response in rectal cancer? An explanation of discrepancies and suggestions for improvement. Cancer Treat. Rev. 2020, 84, 101964. [Google Scholar] [CrossRef] [Green Version]
  138. McMillan, D.C. The systemic inflammation-based Glasgow Prognostic Score: A decade of experience in patients with cancer. Cancer Treat. Rev. 2013, 39, 534–540. [Google Scholar] [CrossRef]
  139. Galamb, O.; Barták, B.K.; Kalmár, A.; Nagy, Z.B.; Szigeti, K.A.; Tulassay, Z.; Igaz, P.; Molnár, B. Diagnostic and prognostic potential of tissue and circulating long non-coding RNAs in colorectal tumors. World J. Gastroenterol. 2019, 25, 5026–5048. [Google Scholar] [CrossRef]
  140. Li, A.; Wang, W.C.; McAlister, V.; Zhou, Q.; Zheng, X. Circular RNA in colorectal cancer. J. Cell. Mol. Med. 2021, 25, 3667–3679. [Google Scholar] [CrossRef]
  141. Pulverer, W.; Kruusmaa, K.; Schönthaler, S.; Huber, J.; Bitenc, M.; Bachleitner-Hofmann, T.; Bhangu, J.S.; Oehler, R.; Egger, G.; Weinhäusel, A. Multiplexed DNA Methylation Analysis in Colorectal Cancer Using Liquid Biopsy and Its Diagnostic and Predictive Value. Curr. Issues Mol. Biol. 2021, 43, 1419–1435. [Google Scholar] [CrossRef]
  142. Biomarkers Definitions Working Group; Atkinson, A.J., Jr.; Colburn, W.A.; DeGruttola, V.G.; DeMets, D.L.; Downing, G.J.; Hoth, D.F.; Oates, J.A.; Peck, C.C.; Spilker, B.A.; et al. Biomarkers and surrogate endpoints: Preferred definitions and conceptual framework. Clin. Pharmacol. Ther. 2001, 69, 89–95. [Google Scholar] [CrossRef]
  143. Fratte, C.D.; Mezzalira, S.; Polesel, J.; De Mattia, E.; Palumbo, A.; Buonadonna, A.; Palazzari, E.; De Paoli, A.; Belluco, C.; Canzonieri, V.; et al. A Panel of Tumor Biomarkers to Predict Complete Pathological Response to Neoadjuvant Treatment in Locally Advanced Rectal Cancer. Oncol. Res. 2022, 28, 847–855. [Google Scholar] [CrossRef] [PubMed]
  144. Ryan, J.; Creavin, B.; Sheahan, K. Delivery of Personalized Care for Locally Advanced Rectal Cancer: Incorporating Pathological, Molecular Genetic, and Immunological Biomarkers Into the Multimodal Paradigm. Front. Oncol. 2020, 10, 1369. [Google Scholar] [CrossRef] [PubMed]
  145. Kang, J.; Kim, S.; Cho, M.; Hur, H.; Min, B.; Lee, K.; Lim, J.; Kim, N. The Accuracy of Mr Trg for Prediction of Tumor Response and Oncologic Outcomes in Rectal Cancer after Preop.Crt: Correlation with Pathologic Trg. Dis. Colon Rectum 2019, 62, E212. [Google Scholar] [CrossRef]
  146. Yi, Y.; Shen, L.; Shi, W.; Xia, F.; Zhang, H.; Wang, Y.; Zhang, J.; Wang, Y.; Sun, X.; Zhang, Z.; et al. Gut Microbiome Components Predict Response to Neoadjuvant Chemoradiotherapy in Patients with Locally Advanced Rectal Cancer: A Prospective, Longitudinal Study. Clin. Cancer Res. 2021, 27, 1329–1340. [Google Scholar] [CrossRef]
  147. Galon, J.; Mlecnik, B.; Bindea, G.; Angell, H.K.; Berger, A.; Lagorce, C.; Lugli, A.; Zlobec, I.; Hartmann, A.; Bifulco, C.; et al. Towards the introduction of the ‘Immunoscore’ in the classification of malignant tumours. J. Pathol. 2014, 232, 199–209. [Google Scholar] [CrossRef] [Green Version]
  148. El Sissy, C.; Kirilovsky, A.; Van den Eynde, M.; Muşină, A.-M.; Anitei, M.-G.; Romero, A.M.S.; Marliot, F.; Junca, A.; Doyen, J.; Mlecnik, B.; et al. A Diagnostic Biopsy-Adapted Immunoscore Predicts Response to Neoadjuvant Treatment and Selects Patients with Rectal Cancer Eligible for a Watch-and-Wait Strategy. Clin. Cancer Res. 2020, 26, 5198–5207. [Google Scholar] [CrossRef]
  149. Chatila, W.K.; Kim, J.K.; Walch, H.; Marco, M.R.; Chen, C.-T.; Wu, F.; Omer, D.M.; Khalil, D.N.; Ganesh, K.; Qu, X.; et al. Genomic and transcriptomic determinants of response to neoadjuvant therapy in rectal cancer. Nat. Med. 2022, 28, 1646–1655. [Google Scholar] [CrossRef]
Figure 1. Biomarker production under physiological and pathological conditions. 1. Under physiological conditions, normal epithelia and stroma shed proteins, lipids, and nucleic acids into the blood circulation. 2. Tumors modify the circulating biomarker composition by shedding tumor markers and by modulating the normal circulating biomarker production. 3. Circulating biomarkers levels can be measured in peripheral blood, and it may be predictive of tumor response. Legend: CA 19-9: cancer antigen 19-9, CEA: carcinoembryonic antigen, cfDNA: cell free DNA, ctDNA: circulating tumor DNA, CRP: c-reactive protein, mRNA: messenger RNA, miRNA: micro-RNA, NK: natural killer cells.
Figure 1. Biomarker production under physiological and pathological conditions. 1. Under physiological conditions, normal epithelia and stroma shed proteins, lipids, and nucleic acids into the blood circulation. 2. Tumors modify the circulating biomarker composition by shedding tumor markers and by modulating the normal circulating biomarker production. 3. Circulating biomarkers levels can be measured in peripheral blood, and it may be predictive of tumor response. Legend: CA 19-9: cancer antigen 19-9, CEA: carcinoembryonic antigen, cfDNA: cell free DNA, ctDNA: circulating tumor DNA, CRP: c-reactive protein, mRNA: messenger RNA, miRNA: micro-RNA, NK: natural killer cells.
Cells 12 00413 g001
Figure 2. Flow chart illustrating the screening and selection process.
Figure 2. Flow chart illustrating the screening and selection process.
Cells 12 00413 g002
Table 1. Studies exploring CEA as a biomarker predictive of tumor regression following CRT.
Table 1. Studies exploring CEA as a biomarker predictive of tumor regression following CRT.
StudyCut-OffNMeasured OutcomeSnSpVPPVPNp Value
(Univariate Analysis)
pre-CRT
Engel et al. [21]2.5 ng/mL209pCR59.5%65.3%30.1%86.5%0.004
Aires et al. [22]2.7 ng/mL171TRG 0–1 (Rayan)43.6%75.4%n/an/a0.0213
Song et al. [23]2.85 ng/mL674pCR52.2%66.5%28.3%84.6%<0.0001
Jang et al. [24]3.5 ng/mL109TRG 3–4 (Dworak)66.6%53.1%50%69.4%ns
Heo et al. [25]4.4 ng/mL52pCR14.3%36.8%7.7%53.8%<0.01 *
Yeo et al. [26]5 ng/mL260Yp Stage 0–180.8%45.8%45.8%80.8%<0.01 *
Kitayama et al. [27]5 ng/mL73pCR70%56.5%20.6%92.1%ns
Kim et al. [28]5 ng/mL314TRG 3–4 (Dworak)81.1%41.8%16.4%94%0.007 *
Choi et al. [29]5 ng/mL53pCR63.6%45.3%23.3%86.4%ns
Yang J. et al. [30]5 ng/mL531pCR66%50%23.5%86.4%0.021 *
Cheong et al. [31]5 ng/mL145pCR92.6%63%38.5%97.1%<0.001*
Huang et al. [32]5 ng/mL236pCR71.4%42.2%27.8%82.6%ns
Gago et al. [33]5 ng/mL89pCR63.2%43.5%25.5%79.4%ns
Guo et al. [34]5 ng/mL751TRG 1–2 (Mandard)61.7%51.8%54.1%59.4%0.009 *
Zhang et al. [35]5 ng/mL432pCRn/an/an/an/a0.001 *
Wada et al. [36]5 ng/mL106pCR72.2%30.1%18.3%83.3%ns
Murahashi et al. [37]5 ng/mL85pCRn/an/an/an/ans
Sawada et al. [38]5 ng/mL267TRG 3–4 (Dworak)n/an/an/an/ans
Cai et al. [39]5 ng/mL284pCRn/an/an/an/ans
Yang K.L. et al. [40]6 ng/mL138pCRn/an/an/an/ans
Tawfik et al. [41]n/a98pCRn/an/an/an/a0.002 *
post-CRT pre-surgery
Huang et al. [32]2 ng/mL236pCR16.1 %93.9%45%78.2%0.0285 *
Song et al. [23]2.45 ng/mL674pCR66.9%54.5%27.1%86.7%<0.0001 *
Yang K.L. et al. [40]2.61 ng/mL138pCR76%58.4%n/an/a0.026 *
Jang et al. [24]2.7 ng/mL109TRG 3–4 (Dworak)88.9%42.2%51.9%84.4%<0.001 *
Choi et al. [29]5 ng/mL53pCR90.9%14.3%22.2%100%ns
Cheong et al. [31]5 ng/mL135pCR100%25%25%100%0.008 *
Wada et al. [36]5 ng/mL106pCR94.7%4.9%18.8%80%ns
Cai et al. [39]5 ng/mL284TRG 0–1 (NCCN)n/an/an/an/a<0.001 *
Restivo et al. [42]5 ng/mL260pCR95.3%32.3%21.8%97.2%<0.0001 *
Hu et al. [43]5 ng/mL71pCR81.8%36.7%19.1%91.7%ns
post-CRT/pre-CRT ratio
Cai et al. [39]0.23284TRG 0–1 (NCCN)n/an/an/an/a<0.001 *
pre-CRT/post-CRT ratio
Song et al. [23]1.07674pCR38.2%73.8%26.9%82.5%0.006 *
clearance pattern (R2)
Hu et al. [43]0.971pCR81.8%63.3%29%95%0.008 *
pre-CRT/tumor size ratio
Gago et al. [33]2.4 ng/mL per cm89pCR82.4%19.6%23.7%78.6%0.04
*: also significant in multivariate analysis. Legend: CRT: chemoradiotherapy, NCCN: national comprehensive cancer network, NPV: negative predictive value, ns: non-significant, n/a: not available, N: number of patients, pCR: pathological complete response, yp Stage: pathological stage after neo-adjuvant treatment, PPV: positive predictive value, Sn: sensitivity, Sp: specificity, TRG: tumor regression grade.
Table 2. Studies exploring thrombocytes level as a biomarker predictive of tumor regression following CRT.
Table 2. Studies exploring thrombocytes level as a biomarker predictive of tumor regression following CRT.
StudyCut-OffNMeasure OutcomeSnSpVPPVPNp. Value
(Univariate)
pre-CRT
Kim et al. [28]370 G/L314pCR94.7%22.8%14.6%96.9%0.01 *
Krauthamer et al. [52]350 G/L140pCR68.2%42.9%34.9%75%ns
Aires et al. [22]253.5 G/L171TRG 0–1 (Rayan)75.5%47.8%n/an/a0.0018
Tawfik et al. [41]n/a98pCRn/an/an/an/ans
post-CRT pre-Surgery
Tawfik et al. [41]n/a98pCRn/an/an/an/ans
*: also significant in multivariate analysis. Legend: CRT: chemoradiotherapy, NPV: negative predictive value, ns: non-significant, n/a: not available, N: number of patients, pCR: pathological complete response, PPV: positive predictive value, Sn: sensitivity, Sp: specificity, TRG: tumor regression grade.
Table 3. Studies exploring cfDNA as a biomarker predictive of tumor regression following CRT.
Table 3. Studies exploring cfDNA as a biomarker predictive of tumor regression following CRT.
StudyMethodNMeasure OutcomeMeasured
Marker
Time
Point
Significant Markers
(p < 0.05)
Zitt et al. [100]qPCR26ypDownstagingcfDNA levelspre-CRT
post-CRT
post-Surgery
post-Surgery cfDNA levels
Agostini et al. [101]qPCR67TRG (Mandard)cfDNA levels
Integrity index
pre-CRT
post-CRT
post-Integrity index
Sun et al. [102]qPCR34TRG
(Dworak)
cfDNA levels
cfDNA integrity
MGMT promoter methylation
KRAS mutation
pre-CRT
post-CRT
pre-CRT 400 bp cfDNA concentration
cfDNA integrity
pre-CRT MGMT promoter methylation
Schou et al. [103]Direct Fluorescence123pCRcfDNA levelspre-CRT
post-CRT
ns
Shalaby et al. [104]qPCR93TRG
(Dworak)
MGMT and ERCC-1 promoter methylationpre-CRTpre-CRT methylation of MGMT and ERCC-1 promoters
Legend: cfDNA: cell free DNA, CRT: chemoradiotherapy, ns: non-significant, N: number of patients, pCR: pathological complete response, qPCR: quantitative polymerase chain reaction, TRG: tumor regression grade.
Table 4. Studies exploring ctDNA as a biomarker predictive of tumor regression following CRT.
Table 4. Studies exploring ctDNA as a biomarker predictive of tumor regression following CRT.
StudyMethodNMeasure OutcomeMeasured
Marker
Time
Point
(Detection Rate)
Significant Markers
(p < 0.05)
Murahashi et al. [37]Amplicon-based deep sequencing85TRG
(Dworak)
ctDNA levelpre-CRT (57.6%)
post-CRT (22.3%)
post-Surgery
ctDNA reduction
(post-CRT/pre-CRT ctDNA levels)
Pazdirek et al. [108]singleplex PCR36TRG
(Dworak)
ctDNA levelpre-CRT (21.2%)
during-CRT
ns
Carpinetti et al. [109]WGS4TRG
(Dworak)
ctDNA levelpre-CRT
during-CRT
post-CRT
ns
Zhou et al. [110]NGS104pCR
TRG
(CAP)
ctDNA levelpre-CRT (75%)
during-CRT (15.6%)
post-CRT (10.5%)
post-Surgery (6.7%)
post-CRT ctDNA level
McDuff et al. [111]NGS
ddPCR
29pCRctDNA levelpre-CRT
post-CRT
ns
Khakoo et al. [112]ddPCR47mrTRGctDNA levelpre-CRT (74%)
during CRT (21%)
post-CRT (21%)
post-Surgery (13%)
post-CRT ctDNA
Level
Sclafani et al. [113]ddPCR97CR
(RECIST 1.1)
ctDNA level
KRAS/BRAF mutation
pre-CRT (50–66%)ns
Appelt et al. [114]ddPCR146TRG
(Mandard)
Meth-ctDNA (NPY)pre-CRT (20.5%)ns
Tie et al. [115]NGS159pCRctDNA level pre-CRT (77%)
post-CRT (8.3%)
post-Surgery (12%)
ns
Legend: CRT: chemoradiotherapy, ctDNA: circulating tumor DNA, CR: complete response, CAP: college of american pathologists, ddPCR: dropplet digital polymerase chain reaction, mrTRG: magnetic resonance tumor regression grade, meth-ctDNA: methylated circulating tumor DNA, NGS: next-generation sequencing, N: number of patients, pCR: pathological complete response, PCR: polymerase chain reaction, TRG: tumor regression grade, WGS: whole-genome sequencing.
Table 5. Studies exploring miRNA as a biomarker predictive of tumor regression following CRT.
Table 5. Studies exploring miRNA as a biomarker predictive of tumor regression following CRT.
StudyMethodNVMeasure OutcomeMeasured
Marker
Time
Point
Significant Markers
(p < 0.05)
Wada et al. [36]qRT-PCR4165TRG
(Mandard)
miRNA
(8-panel)
pre-CRTAll panel:
miR-30e-5p
miR-33a-5p
miR-130a-5p
miR-210-3p
miR-214-3p
miR-320a
miR-338-3p
miR-1260a
Dreussi et al. [125]DNA sequencing265/pCRmiRNA-related SNP
(114-panel)
n/aDROSHA-rs10719
SMAD3-rs17228212
SMAD3 rs744910
SMAD3-rs745103
TRBP-rs6088619
D’Angelo et al. [126]qRT-PCR34/TRG
(Mandard)
miRNA
(11-panel)
pre-CRTmiR-125b
Yu et al. [127]qRT-PCR8742TRG
(Mandard)
miRNA
(16-panel)
pre-CRTmiR-345
Azizian et al. [128]Real time PCR42/Lymph Node NegativitymiRNA
(5-panel)
pre-CRT
during CRT
post-CRT
miR-20a
miR-18b
Meltzer et al. [129]n/a29 64TRG
(CAP)
Exosomal miRNA
(372 panel)
pre-CRTns
Baek et al. [130]qRT-PCR89/TRG
(Rödel)
Exosomal miRNA
(16-panel)
pre-CRTExosomal miR-199b-5p
Hiyoshi et al. [131]RT-PCR94/TRG
(Dworak)
miRNA
(18-panel)
pre-CRTmiR-43
Legend: CRT: chemoradiotherapy, CAP: college of american pathologists, miRNA: micro-RNA, n/a: not available, N: number of patients, V: number of patients in a validation cohort, qRT-PCR: quantitative real time polymerase chain reaction, pCR: pathological complete response, RT-PCR: real time polymerase chain reaction, SNP: single nucleotide polymorphisme, TRG: tumor regression grade.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Machado Carvalho, J.V.; Dutoit, V.; Corrò, C.; Koessler, T. Promises and Challenges of Predictive Blood Biomarkers for Locally Advanced Rectal Cancer Treated with Neoadjuvant Chemoradiotherapy. Cells 2023, 12, 413. https://doi.org/10.3390/cells12030413

AMA Style

Machado Carvalho JV, Dutoit V, Corrò C, Koessler T. Promises and Challenges of Predictive Blood Biomarkers for Locally Advanced Rectal Cancer Treated with Neoadjuvant Chemoradiotherapy. Cells. 2023; 12(3):413. https://doi.org/10.3390/cells12030413

Chicago/Turabian Style

Machado Carvalho, Joao Victor, Valérie Dutoit, Claudia Corrò, and Thibaud Koessler. 2023. "Promises and Challenges of Predictive Blood Biomarkers for Locally Advanced Rectal Cancer Treated with Neoadjuvant Chemoradiotherapy" Cells 12, no. 3: 413. https://doi.org/10.3390/cells12030413

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop