51
Views
1
CrossRef citations to date
0
Altmetric
Review

Future directions in combined modality therapy for rectal cancer: reevaluating the role of total mesorectal excision after chemoradiotherapy

, &
Pages 1097-1110 | Published online: 14 Aug 2013

Abstract

Most patients who develop rectal cancer present with locoregionally advanced (T3 or node-positive) disease. The standard management of locoregionally advanced rectal cancer is neoadjuvant concurrent chemoradiotherapy (nCRT), followed by radical resection (low-anterior resection or abdominoperineal resection with total mesorectal excision). Approximately 15% of patients can have a pathologic complete response (pCR) at the time of surgery, indicating that some patients can have no detectable residual disease after nCRT. The actual benefit of surgery in this group of patients is unclear. It is possible that omission of surgery in these patients, termed selective nonoperative management, can limit the toxicities associated with standard, multimodal combined modality therapy without compromising disease control. In this review, we discuss the clinical experiences to date using selective nonoperative management and various attempts at escalation of nCRT to improve the number of patients who have a pCR. We also explore several clinical, laboratory, imaging, histopathologic, and genetic biomarkers that have been tested as tools to predict which patients are most likely to have a pCR after nCRT.

Introduction

Up to 70% of patients with nonmetastatic rectal cancer present with locoregionally advanced disease.Citation1 Locally advanced rectal cancer (LARC) is generally defined as T3 or node-positive rectal cancer. The standard of care for LARC in the United States is neoadjuvant concurrent chemoradiotherapy (nCRT) given with conventional fractionation over 6 weeks, followed by radical resection with total mesorectal excision (TME) and adjuvant chemotherapy.

With TME, there can be up to a 2% mortality rate. Length of hospital admissions average 15 days.Citation2,Citation3 The incidence of morbidity ranges from 6% to 35%, which includes anastomotic leak, pain, and blood loss from the procedure.Citation4 Between 20% and 40% of patients have severe sexual dysfunction after surgery.Citation5,Citation6 There are also compelling data regarding the effect of resection on quality of life. Patients undergoing abdominoperineal resection (APR), in particular, have reported decreased quality of life.Citation7 According to Pachler and Wille-Jørgensen, it is unclear whether low-anterior resection minimizes this decline in quality of life compared with APR.Citation8

Because many reports indicate that approximately 15% of patients with LARC have a pathologic complete response (pCR) to neoadjuvant therapy at the time of surgery,Citation9 it is reasonable to wonder whether the risks of TME outweigh the benefits in this select group of patients. Select series indeed show excellent long-term outcomes in patients who have a pCR to nCRT without subsequent TME, suggesting that it is possible to avoid planned, radical resection in some patients.Citation9,Citation10 A review by O’Neill et al discussed some of the key issues that must be addressed in pursuing such an approach in patients with LARC.Citation11 In this review, we describe the rationale and clinical experiences using selective nonoperative management (SNOM), highlight avenues to improve the efficacy of nCRT to render more patients with a pCR, and explore potential biomarkers associated with response to nCRT that could help identify optimal candidates for SNOM.

Methods

We performed a review of the literature concerning selective nonoperative management in LARC by searching MEDLINE for English-language articles to identify studies with the following subject matter: “rectal cancer”, “neoadjuvant chemoradiotherapy”, “pathologic complete response”, “selective nonoperative management after neoadjuvant chemoradiotherapy”, “radiotherapy dose escalation”, “chemotherapy escalation and novel combinations”, and “clinical, imaging, and biologic biomarkers predicting for response to treatment”. Retrospective cohort and case-control series, as well as prospective cohort and Phase I, II, and III clinical trials, were included in our review.

pCR as an indicator of long-term outcomes

The ideal candidates for SNOM are patients with the best responses to nCRT. pCR, or the complete eradication of macroscopic and microscopic tumor as judged by histopathologic evaluation of the specimen from the radical resection, is the most desirable outcome after nCRT. In the German CAO/ARO/AIO-94 randomized Phase III trial of preoperative versus postoperative CRT on which the current standard of therapy of preoperative chemoradiotherapy is based, 8% of patients had a pCR after preoperative therapy.Citation12 Of these patients, only 2.9% developed a local recurrence at 10 years after TME. Several large series demonstrate that patients with a pCR at the time of surgery have substantially improved long-term local control, distant control, and disease-free survival (DFS; ).Citation9,Citation10,Citation13 It is unclear how much radical resection contributes to the low rate of local recurrence in patients with a pCR. Unfortunately, there are no randomized data that directly compare outcomes with or without surgical excision in this group of patients, and proper identification of patients with a pCR without performing radical resection and detailed histopathologic analysis is difficult. Despite these issues, using the pCR as a surrogate for the ultimate response may help identify the patients who are most appropriate for SNOM.

Table 1 5-year outcomes in a large series evaluating disease outcomes in patients with rectal cancer obtaining a pathologic complete response to neoadjuvant chemoradiotherapy

Clinical experiences exploring SNOM

The most extensive clinical experience using SNOM is from Sao Paulo, Brazil. There have been multiple publications on the outcomes of patients treated at two institutions in Brazil: the University of Sao Paulo Medical School, Sao Paulo and the Angelita and Joaquim Gama Institute, Sao Paulo.Citation14Citation18 Eligibility included T3–4 disease, node-positive or T2 distal rectal tumors that would otherwise require an APR. In the initial reports, staging was performed via physical exam, digital rectal examination (DRE), rigid proctoscopy, chest X-ray, computed tomography (CT) abdomen/pelvis, and serum carcinoembryonic antigen (CEA).Citation14,Citation15 Patients were treated with 50.4 Gy and concurrent 5-fluorouracil (5-FU) and leucovorin.Citation14,Citation15 Response assessment was performed by a colorectal surgeon 8 weeks after nCRT, using the same staging modalities. Patients who were felt to have a complete clinical response (cCR), which was defined as no significant residual ulcer and no positive biopsy, did not undergo surgery. Instead, they had monthly physical exam, DRE, proctoscopy, biopsy if feasible, and serum CEA assessment for the first year, which was increased to every 2 months for the second year and then 6 months for the third year. CT abdomen/pelvis and chest X-ray were repeated every 6 months for the first year.

In their largest report comparing operative with nonoperative management, Habr-Gama et al demonstrated favorable results of SNOM.Citation14 Of 265 patients treated with nCRT, the outcomes of 22 patients with a pCR after surgery were compared with the outcomes of 71 patients who had a cCR for at least 12 months. There was no difference in 5-year overall survival (OS) or DFS between patients. Importantly, there were no cancer-related mortalities in the nonoperative group. Two (3%) patients had endorectal recurrences, both of whom were salvaged (one with brachytherapy and one with transanal excision). A subsequent analysis of 361 patients using the same protocol provided more insight into the patterns of failure of patients treated with SNOM ().Citation15 Of 122 patients who had a cCR to nCRT (34% of the entire cohort), 99 (81%) maintained a cCR for 1 year. Patients with endorectal-only failures (5%) were successfully salvaged without further recurrence, and none died of rectal cancer.

Table 2 Clinical experiences of selective nonoperative management in locally advanced rectal cancer after neoadjuvant chemoradiotherapy

Over time, the Brazilian protocol has evolved.Citation17,Citation18 Patients are currently additionally staged using pelvic magnetic resonance imaging (MRI) or endorectal ultrasound (ERUS).Citation19 Patients are treated with 54 Gy rather than 50.4 Gy and receive three cycles of adjuvant chemotherapy and reassessment at 10 weeks. Patients with any suspicious areas undergo full-thickness local excision for diagnostic purposes. Although the definition likely evolved over the course of the published studies, a strict definition of cCR has been published by Habr-Gama et al.Citation20 Patients with any residual deep ulceration, any superficial ulcer or irregularity (even if only mucosal), or any palpable nodule on DRE are considered to have an incomplete clinical response, whereas those with no palpable abnormality, white discoloration of mucosal surface, telangiectasia, or subtle loss of pliability of the rectal wall could be considered as having a cCR.

There are some limitations to the Brazilian experience. The outcomes of patients who initially achieved a cCR but had a recurrence within the first year (who were excluded from analysis) have not clearly been discussed. Poor outcomes in these patients could sway one away from SNOM. In addition, stage I patients are not typically treated with nCRT and have more favorable outcomes than those with LARC, yet 11% of patients in the latter paper had stage I (T2N0) disease, potentially favorably biasing the results.Citation15

Several other institutions have reported the outcomes of their patients who were treated with SNOM. describes these outcomes.Citation21Citation24 Maas et al reported excellent outcomes in their patients treated with SNOM.Citation21 In this study, patients with a cCR (n = 21) were compared with 20 patients with a pCR at surgery. Nearly half (10/21) of the patients treated nonoperatively would have required an APR, and all had sphincter preservation, whereas nearly half (9/20) of those in the surgical group required permanent colostomy. One of the patients developed a local recurrence in the nonoperative group, whereas none did in the operative group. This patient with recurrence had a transanal excision with complete resection. The cumulative 2-year DFS was 89% versus 93%, and 2-year OS was 100% versus 91%, for the nonoperative and surgical groups, respectively, with differences that were not statistically significant. A series from Memorial Sloan-Kettering Cancer Center found that when comparing 32 patients treated with nonoperative management with 57 patients with pCR at surgery, patients treated without surgery had a higher risk of local recurrence (19%; n = 6).Citation22 However, all recurrences were successfully salvaged with surgery, and there was no difference in DFS, distant metastasis, or survival between the two groups. Two series from the United Kingdom have also evaluated SNOM as a treatment approach, but neither of these studies compared the outcomes of these patients with those of patients treated with planned surgery.Citation23,Citation24

There are several limitations to the published experiences of SNOM. The most important issue is that the bulk of the data come from small, retrospective, single-institution series with relatively short follow-up. In addition, most studies included patients with earlier-stage disease (some cT2N0 patients). The favorable outcome of these patients may have made the outcomes of the nonoperative group appear more favorable than would be expected for LARC. An important point to highlight is that the patients in these relatively small cohorts may have been carefully selected for consideration of nonoperative management based on location and size. For example, in the series by Habr-Gama et al, approximately 72% of patients had N0 disease, and mean tumor size was 3.8 cm.Citation15 It is difficult to translate these results to patients with higher and more advanced tumors with a clinical complete response. In addition, the proportion of patients who achieved a cCR after nCRT in each of these studies was relatively low (10%–34%).Citation15,Citation21,Citation23 The definition of cCR, methods of determining response, follow-up frequency, and clinical/imaging modalities to assess for recurrence differ from study to study. In the series that compares the outcomes of nonoperative with operative patients, the patients selected as controls are those with a pCR after radical surgery. As discussed later, cCR does not necessarily correlate with pCR, and thus these control groups may not be the optimal comparison group for the nonoperative cCR patients.

Before SNOM can be implemented in routine practice, more robust data are necessary. Ultimately, a randomized trial comparing the two approaches would provide the strongest evidence, but a large cohort study with structured and longterm follow-up could also be useful to better define the outcomes with SNOM. A more ideal comparison would involve patients with cCR (rather than pCR), using prospectively defined criteria, who are then either observed or resected.

Efforts to improve the efficacy of nCRT

It is likely that to be a candidate for SNOM, a patient needs to have at least a cCR to treatment. The 10–34% cCR rates in the above studies, as well as the 15% pCR rate, suggest that only a minority of patients are potential candidates for SNOM. Advances in chemotherapy and radiation therapy could improve these rates and increase the proportion of patients eligible for SNOM.

Radiation dose escalation and altered fractionation

One way to improve the pCR rate after nCRT is to intensify radiotherapy (RT). Conventional RT is typically given as 1.8–2 Gy per fraction once a day until the total dose is reached. Altered RT fractionation, which refers to changes in the daily dose and total treatment time, has been attempted to improve the response to RT. Short-course RT is unlikely to be the preferred approach for SNOM because it results in a low pCR rate. Two randomized studies compared long-course RT (50.4 Gy) with short-course RT (25 Gy) followed by radical resection.Citation25,Citation26 Disease control outcomes and reported toxicity between the two regimens were similar, but the pCR rate was higher in the 50.4 Gy group than in the 25 Gy group in both studies (15–16% compared with 1%). A caveat to this is that the short time after completion of RT to resection (1 week) could have limited the number of pCRs in the short-course RT group. If they had undergone surgery later, more pCRs may have been detected.Citation27 Radiation Therapy Oncology Group (RTOG) 0012 evaluated a hyperfractionated schema of 55.2–60 Gy in 1.2 Gy fractions twice daily with concurrent 5-FU compared with 50.4–54 Gy with conventional fractionation and 5-FU and irinotecan.Citation28 The rate of pCR was high for both groups (26%); there was no statistically significant difference.

Several attempts have been made to improve pCR and disease control outcomes using RT dose escalation (). Among patients treated with long-course RT, there appears to be a benefit in dose escalation from 40 to 50 Gy. A study of patients treated in three consecutive RT dose schedules found that with increasing dose from 40 to 50 Gy, the pCR rates, local control, cause-specific survival, and progression-free survival improved.Citation29 Mohiuddin et al found that patients treated with more than 55 Gy and continuous infusion 5-FU had a substantially higher pCR rate compared with those receiving less than 50 Gy (44% versus 13%; P = 0.05).Citation30 In a series by Wiltshire et al, patients treated with 46 Gy or more had improved local recurrence-free survival, DFS, and OS compared with patients treated with 40 Gy, but escalation to 50 Gy did not improve outcomes.Citation31 Unfortunately, most of the above series indicate that with elevated dose, there is also increased toxicity, potentially limiting the applicability of high RT doses.

Table 3 Radiotherapy dose escalation studies in locally advanced rectal cancer

Increasing the RT dose with modalities other than External Beam RT (EBRT) has also been assessed. Unlike EBRT, which is the use of X-rays to deliver radiation to tumors, brachytherapy refers to the placement of radioisotopes within or near tumor tissues, and contact X-ray RT (CXR) is the use of a narrow beam of low-energy photon radiation placed in close proximity to tumor tissues. Both of these specialized modalities have the advantage of limiting dose to surrounding normal tissues in comparison with a multifield, EBRT approach. The role of using these modalities as a boost to escalate RT dose from EBRT has not been established. A randomized trial comparing 50.4 Gy versus 50.4 Gy with 10 Gy endorectal high-dose-rate brachytherapy boost found no difference in pCR rate (18% in both groups).Citation32 In contrast, a randomized trial of EBRT compared with EBRT with CXR boost revealed improved pathologic response (defined as no, or few residual cells at the time of surgery) in the high-dose group.Citation33 Of note, CXR therapy for rectal cancer has been primarily studied for low-lying and less advanced tumors; the applicability of this treatment approach to more advanced or more proximal tumors is unclear.

Newer technologies, such as intensity-modulated radiotherapy (IMRT), may allow for safer RT dose escalation. IMRT uses advanced planning and delivery techniques that can reduce the amount of high-dose radiation to normal tissues by varying the intensity of radiation delivered in each field. This normal tissue sparing has the potential to minimize toxicity because most adverse effects of radiation are a result of high doses to normal tissues. Several series show that IMRT decreases doses to critical normal organs and may thus safely allow for further dose escalation.Citation34,Citation35 De Ridder et al and Engels et al performed a Phase II study of 108 patients with cT3–4 tumors treated with neoadjuvant RT alone, using an IMRT and simultaneous integrated boost (SIB) approach with helical tomotherapy.Citation36,Citation37 Patients with anticipated close circumferential resection margins received 55.2 Gy, whereas the others received 46 Gy. Both groups were treated to a smaller treatment volume to allow for small bowel sparing. The pCR rate was 8%, but a favorable acute toxicity profile was noted in comparison with that seen for previously reported, standard RT techniques. A small Phase II study of eight patients treated using a simultaneous integrated boost approach to 55 Gy in 2.2 Gy/fraction to gross tumor found three of eight patients had a pCR.Citation38 However, a Phase I study of eight patients at Fox Chase Cancer Center (Philadelphia, PA, USA) found unacceptable toxicity (38% had grade 3 toxicity) with a similar treatment approach, albeit with larger treatment volumes.Citation39 The small sample sizes of these studies limit the ability to make conclusions regarding the risk–benefit ratio of dose escalation with IMRT.

The effect of regional hyperthermia to nCRT on response has also been assessed in several series. In a retrospective review of 106 patients, Schroeder et al found that patients treated with the addition of regional hyperthermia had improved pCR rates (16.4% versus 6.7%).Citation40 However, two single-group Phase II studies combining hyperthermia with nCRT found similar pCR compared with nCRT alone.Citation41,Citation42 Therefore, the role of hyperthermia is unclear.

Chemotherapy agent, dose, and timing modifications

The backbone of concurrent chemotherapy in the nCRT regimen has traditionally been 5-FU. Importantly, the schedule in which the drug is administered has been shown to affect its efficacy. Continuous infusional 5-FU has been shown to be more effective than bolus 5-FU in the metastatic setting and adjuvant concurrent CRT settingCitation43,Citation44 and has therefore been adopted as the standard chemotherapy schedule when given as nCRT. An attractive alternative to infusional 5-FU is the oral fluoropyrimidine, capecitabine, which likely is at least equivalent to 5-FU, if not better. In a randomized Phase III trial, capecitabine was associated with a trend toward improved pCR rate (14% versus 5%; P = 0.09), 3-year DFS (71% versus 63%; P = not reported), and 5-year survival (66% versus 61%; P = not reported) in patients treated with nCRT for LARC.Citation45

Despite encouraging Phase II studies, Phase III studies combining other agents with capecitabine or 5-FU have mostly not improved pCR rates (). Although several trials showed no benefit for the addition of oxaliplatin,Citation46Citation48 the German CAO/ARO/AIO-04 randomized Phase 3 trial showed a modest improvement in pCR.Citation49 RTOG 0012 revealed no benefit in pCR with the addition of irinotecan.Citation28 A single-group Phase II study of 25 patients demonstrated an encouraging pCR rate of 32% with the addition of bevacizumab to capecitabine and 50.4 Gy but must be confirmed in a larger randomized trial.Citation50 Adding cetuximab to nCRT does not appear to improve pCR.Citation51,Citation52

Table 4 Addition of combination chemotherapy agents to 5-FU- or CAP-based nCRT

Additional chemotherapy after nCRT and before surgical resection has also been investigated. A follow-up study by Habr-Gama et al included 29 patients treated with three extra cycles of bolus 5-FU and leucovorin after nCRT.Citation17 cCR at 10 weeks was 76% (n = 22), and of these, 63% (n = 14) had a cCR for 1 year. It is important to note that in this study, 17% of patients had stage I disease. A nonrandomized multicenter Phase II trial treated patients with nCRT, followed by 2 cycles of modified FOLFOX-6 (5-fluorouracil, leucovorin, and oxaliplatin) if a cCR by imaging and proctoscopy was achieved after nCRT.Citation53 Patients treated with post-nCRT chemotherapy had a higher rate of pCR at the time of TME compared with patients who had standard nCRT followed by TME (25% versus 18%), but this difference was not statistically significant.

Identifying the best responders

For SNOM to be successful, it is imperative to identify patients who have a pCR after nCRT without radical resection. Various methods are used to assess those who are most likely to have an excellent response, including clinical factors, physical exam findings, and laboratory and genetic factors. Here, we discuss various modalities of assessment and their strengths and weaknesses.

Clinical parameters

Several clinical factors have been associated with improved response to nCRT. One series found higher pCR rates for less rectal wall circumference involvement and lower pCR rates for tumors involving more than 60% of the circumference and being more than 5 cm from anal verge.Citation54 Although some studies suggest that initial clinical T-stage and N-stage may be associated with response,Citation55,Citation56 others suggest that these characteristics may not be as important as genetic factors.Citation57,Citation58

It is possible that treatment response (and factors that may influence response) are dependent on time between nCRT and surgery. A longer interval between nCRT and radical surgery may improve pCR rates, and this delay of resection may not compromise local control.Citation16,Citation27,Citation59,Citation60 A randomized study from Lyon found that pathologic downstaging was increased when the interval between RT and surgery was increased from 2 weeks to 6–8 weeks (10% versus 26%; P = 0.005), although the primary endpoint of sphincter-preserving surgery was not significantly different.Citation61 Even patients treated with short-course RT can have a pCR rate of 8% if surgery occurs after 4 weeks (compared with 1% at 1 week).Citation62 However, in the Brazilian experience, using conventional nCRT followed by surgery, patients who had more than12 weeks delay had no difference in pCR, DFS, or survival compared with those who had surgery at 12 weeks or less.Citation16 The current National Comprehensive Cancer Network guideline is an interval between nCRT and radical resection of 5–10 weeks.Citation63

Clinical assessment is the most commonly used form of post-treatment response assessment. A review from Memorial Sloan-Kettering of patients treated with nCRT followed by resection evaluated post-treatment DRE and proctoscopy to evaluate clinical response to CRT and found that only 25% of patients with cCR actually had a pCR, which was generally 6 weeks after nCRT (with response assessment 1 week before surgery).Citation64 Another study found that complete response by DRE only correctly identified pCR in 21% of patients with a pCR.Citation65 In addition, many patients with pCR may have residual mucosal abnormality at the time of response assessment.Citation66 For patients who have what appears to be an incomplete clinical response, a series from Perez et al found that 21% of patients with a negative endoscopic biopsy actually had a pCR.Citation67 These findings make post-treatment physical exam findings even more difficult to use in decision making. For these reasons, pretreatment clinical parameters, physical exam findings after treatment, and post-treatment biopsy are not sufficient to predict pCR.

Imaging modalities

Several imaging modalities are commonly used in the initial staging and response assessment of patients with rectal cancer, including CT and ERUS. The ability of ERUS and CT to individually predict for pCR both at the primary and nodes is relatively modest.Citation68Citation70 An Italian series of 46 patients compared ERUS, CT, and MRI at 4 weeks after nCRT. In this study, radiographic staging by ERUS or CT that indicated no residual abnormality in the rectal wall was considered T0, tumor felt to be confined to the rectal wall was called T1 or T2, full-thickness involvement of the rectal wall with infiltration of the perirectal fat was T3, and invasion of surrounding organs/structures was T4. Lymph nodes 5 mm or larger were considered to be positive. The accuracy was 64% for ERUS and 74% for CT in predicting pT0 status, and 61% for ERUS and 62% for CT for predicting pN0 status.Citation69

MRI is a promising imaging modality to evaluate anatomic changes after nCRT. Still, RT fibrosis can make interpretation difficult, and although MRI is effective for identifying disease that only involves the rectal wall, it cannot reliably identify patients with pCR. In the Italian series, the accuracy of MRI at 4 weeks after nCRT for pT0 was 77%, and for pN0 it was 65%.Citation69 The Magnetic Resonance Imaging and Rectal Cancer European Equivalence (MERCURY) study was a prospective cohort study of patients treated with nCRT followed by surgical resection.Citation71 In this study, an MRI tumor regression grade (mrTRG) was created based on the TRG for histopathologic response.Citation72 Although the mrTRG was useful for detecting unfavorable pathologic response (ypT3–4 or TRG 0–2), it is unclear whether it can predict pCR at surgery.Citation73 In addition, only 44% of patients had a favorable response (ypT0–3a) in this study at the time of histopathologic evaluation, and only 11% had pCR. This highlights the minority of patients who would be applicable for SNOM with the current treatment platform and MRI response assessment.

Positron emission tomography (PET) offers insight into the metabolic activity of tissues, potentially complementing anatomic imaging. A Brazilian study found a 73% negative predictive value and 85% accuracy of PET/CT for detecting incomplete response.Citation74 Other studies have suggested PET may not be as accurate.Citation75,Citation76 A prospective study of 80 patients treated with nCRT followed by radical resection found that PET/CT did not predict pathologic response, but a decrease in SUV (standardized uptake value) was associated with lower disease recurrence.Citation77 These clinical data, as well as the issue of interobserver variation and no clear standard criteria for response assessment, suggest that the use of PET or PET/CT alone to predict pCR has questionable utility.

Individually, each of these imaging modalities does not appear to be suitable to identify patients with a pCR. However, when combined, they have greater prognostic value. In the Brazilian series, the accuracy of PET/CT combined with clinical exam was 96% in predicting pCR.Citation74 In a Taiwanese series of 166 patients, using MRI, colonoscopy, and rebiopsy to reassess patients at 4 weeks after nCRT, the accuracy was 78% for pCR.Citation78 However, it is important to note that the majority of data using multimodality reassessment are retrospective in nature, are subject to selection bias, and may be influenced by expertise.

CEA and other laboratory findings

Several studies have evaluated whether clinical response assessment can be augmented by pre-and post-treatment CEA levels. One study found that pretreatment CEA levels higher than 5 ng/mL were associated with a poor response to nCRT.Citation57 Another study found that patients with a pCR had a lower pretreatment and post-treatment CEA level, and that no patients with an elevated post-treatment CEA (>5 ng/mL) level had a pCR.Citation79 In this series, post-treatment CEA levels had a stronger association with outcome than pretreatment levels. Another series found that pre-nCRT versus post-nCRT CEA changes were not associated with pCR rate, but those patients with high pretreatment CEA and poor reduction (<70%) after nCRT had a higher risk for local and distant recurrence.Citation80 A Brazilian series found that patients with post-treatment CEA levels lower than 5 ng/mL had higher rates of cCR, pCR, DFS, and OS than those who did not.Citation81 Ultimately, a standard definition of “low” CEA or “good” response must be identified for optimal use of CEA in treatment decision making.

Other laboratory tests have also been evaluated. Kawai et al found that pre-nCRT thrombocytosis was associated with worse response by barium enema, or pathologic response.Citation82 Kitayama et al found that patients who had a complete response had higher lymphocyte ratios and lower neutrophil ratios, suggesting that part of the response to nCRT may be immune-mediated.Citation83 These series hint that other lab parameters may be of use but must be validated in other series before they may be used clinically.

Tumor histopathologic markers

Numerous histopathologic markers have been evaluated to identify patients who will have a good response to nCRT and/or good prognosis ().Citation84Citation105 None have been identified as unequivocal biomarkers of response across all studies. Most biomarkers have been found to be associated with both improved and inferior response in different studies. Most of the studies are small, single-institution series; few are validated externally; and varying nCRT regimens were used. The most extensively studied marker has involved p53 mutations.Citation106Citation108 A meta-analysis found that wild-type p53 was associated with improved rates of “good response.”Citation109 Despite these positive findings, neither p53 nor any of the other studied biomarkers are likely to singularly predict for those destined to have a pCR. The response to treatment is likely too complex to be modeled adequately with one biomarker.

Table 5 Investigated histopathologic factors possibly associated with response to neoadjuvant chemoradiotherapy

Gene and microRNA expression profiles

Several groups have identified tumor gene expression profiles that may be associated with response to nCRT.Citation110Citation112 Interestingly, the gene expression profiles in these studies do not necessarily highlight genes known to be important. Kim et al specifically identified a gene expression profile that identified pCR with 84% accuracy in a testing sample and 87% accuracy in a validation cohort.Citation113 This profile was composed of 95 genes involved in multiple cellular pathways, including MMP14, MSX2, RAD23B, Thymidylate synthase, FGFR4, and ENO1. Similarly, micro (mi)RNA expression profiles may be associated with response to therapy.Citation114,Citation115 An Italian study found a miRNA expression profile that predicted pCR with a sensitivity and specificity of 100%.Citation116 This study included 38 patients with LARC treated with capecitabine and oxaliplatin with 45 Gy RT. Biopsy specimens were analyzed to identify a 13-miRNA expression profile composed of eleven miRNAs that were upregulated and two miRNAs that were downregulated in pCR patients. All of the nine patients with a pCR had upregulation of miR-630 and miR-622, whereas all patients without a pCR had downregulation of these two miRNAs. However, this profile has not been confirmed externally and may not be practical because detailed knowledge regarding individual miRNA levels of expression is required. These findings are encouraging, but further research is necessary to identify and validate the optimal profile to translate it into clinical use.

Circulating/disseminated tumor cells

Circulating tumor cells (CTCs), or tumor cells identified in the peripheral blood, and disseminated tumor cells, tumor cells identified in the bone marrow, have also been evaluated for their role in response prediction. There are no series evaluating whether the presence, concentration, or changes in concentration before or after nCRT of CTCs can identify patients who are likely to have a pCR. One of the challenges with using CTCs as a biomarker for response is that only 30–60% of patients with rectal cancer have detectable CTCs.Citation117,Citation118 An Austrian study found that CTCs were more likely to be detected in patients with a good response to nCRT (ypT0–2) than in nonresponders at the time of surgery (63% versus 18%), and responders had decreased detection over the course of treatment.Citation117 A Norwegian study found that disseminated tumor cell detection was not associated with radiologic TNM stage or TRG score at the time of surgery after nCRT.Citation118

Circulating cell-free DNA

Circulating cell-free DNA (cfDNA) is another promising biomarker. Agostini et al studied 67 patients undergoing nCRT and found that the post-treatment cfDNA integrity index and levels of Alu 247 fragments were independently associated with improved response rate.Citation119 Similarly, Zitt et al found that responders had lower levels of cfDNA after treatment compared to nonresponders.Citation120 However, these findings must be validated, and the ability of cfDNA parameters to predict pCR is unknown.

Toward the development of a SNOM protocol

A standardized approach toward SNOM of rectal cancer has yet to be defined. All of the SNOM series discussed earlier differ in the frequency of follow-up and modalities of initial staging and response assessment. It is likely that even if the merits of a SNOM approach are confirmed in further studies, the optimal protocol would be the subject of much debate. It is clear that the ideal protocol would have comprehensive staging with clinical examination, colonoscopy, and imaging such as ERUS, MRI, and PET-CT both before starting nCRT and for tumor response assessment. The optimal chemoRT regimen is unknown, but 50.4 Gy with FU is not sufficient to achieve meaningful rates of pCR. Patients must be followed frequently. On the basis of the Habr-Gama and Dutch series, it appears that the highest risk for recurrence is within the first year after treatment, when follow-up would need to be closest. In the second year, the frequency of follow-up could be decreased, with gradual relaxation up to 5 years. Long-term follow-up would be essential to establish safety, given the possibility for late recurrencesCitation12 and potential for late toxicity in the intact rectum. Further protocols should also include biomarkers, such as CTCs or cfDNA, which may better predict for pCR and improved outcome. In addition, newer imaging modalities, such as novel PET tracers, should be incorporated, as traditional imaging modalities have limitations in predicting pCR. The ideal timing of surgery in those without a cCR is also unknown. It appears that anywhere from 8 to 12 weeks or more may be safe, but the incremental delay in surgery to potentially allow for increased appreciation of pCR must be weighed against the increased risk of recurrence. However, patients who do reach this point with a cCR could be considered appropriate for omission of surgery.

Conclusion

Several series suggest that SNOM is safe and feasible in a small proportion of patients with locally advanced rectal cancer. However, the literature describing this approach is mostly retrospective and single-institution in nature, with small numbers and varied methodology. In addition, many of these studies lack long-term follow-up. These issues limit the ability to consider SNOM a safe alternative to the standard treatment paradigm. Larger prospective studies with long-term follow-up, ideally randomized prospective trials comparing standard combined modality therapy with SNOM, are necessary to further evaluate the safety and efficacy of this approach. Until more robust studies confirm its appropriateness, SNOM cannot be incorporated into routine practice. Even if further study supports SNOM of rectal cancer, perhaps only a minority of patients will obtain the cCR necessary to pursue SNOM using current nCRT regimens and response assessment tools. Further research is necessary to improve the efficacy of nCRT to induce a complete response. Simultaneously, oncologists must better identify patients who have had a pCR, as current assessment and detection approaches have demonstrated insufficient accuracy to alter treatment. There are biomarkers with encouraging preliminary results, however, which must be validated and confirmed in larger, multicenter cohorts. With progress in these areas, perhaps SNOM will not only be feasible and safe but also be the approach of choice in a subset of patients with LARC.

Disclosure

The authors report no conflicts of interest in this work.

References

  • GundersonLLJessupJMSargentDJGreeneFLStewartARevised tumor and node categorization for rectal cancer based on surveillance, epidemiology, and end results and rectal pooled analysis outcomesJ Clin Oncol201028225626319949015
  • HartleyJEMehiganBJQureshiAEDuthieGSLeePWMonsonJRTotal mesorectal excision: assessment of the laparoscopic approachDis Colon Rectum200144331532111289275
  • LujanJValeroGHernandezQSanchezAFrutosMDParrillaPRandomized clinical trial comparing laparoscopic and open surgery in patients with rectal cancerBr J Surg200996998298919644973
  • BreukinkSPierieJWiggersTLaparoscopic versus open total mesorectal excision for rectal cancerCochrane Database Syst Rev2006184CD00520017054246
  • CornishJATilneyHSHeriotAGLaveryICFazioVWTekkisPPA meta-analysis of quality of life for abdominoperineal excision of rectum versus anterior resection for rectal cancerAnn Surg Oncol20071472056206817431723
  • JayneDGBrownJMThorpeHWalkerJQuirkePGuillouPJBladder and sexual function following resection for rectal cancer in a randomized clinical trial of laparoscopic versus open techniqueBr J Surg20059291124113215997446
  • KasparekMSHassanICimaRRLarsonDRGullerudREWolffBGLong-term quality of life and sexual and urinary function after abdominoperineal resection for distal rectal cancerDis Colon Rectum201255214715422228157
  • PachlerJWille-JørgensenPQuality of life after rectal resection for cancer, with or without permanent colostomyCochrane Database Syst Rev201212CD00432323235607
  • MaasMNelemansPJValentiniVLong-term outcome in patients with a pathological complete response after chemoradiation for rectal cancer: a pooled analysis of individual patient dataLancet Oncol201011983584420692872
  • MartinSTHeneghanHMWinterDCSystematic review and meta-analysis of outcomes following pathological complete response to neoadjuvant chemoradiotherapy for rectal cancerBr J Surg201299791892822362002
  • O’NeillBDBrownGHealdRJCunninghamDTaitDMNonoperative treatment after neoadjuvant chemoradiotherapy for rectal cancerLancet Oncol20078762563317613424
  • SauerRLierschTMerkelSPreoperative versus postoperative chemoradiotherapy for locally advanced rectal cancer: results of the German CAO/ARO/AIO-94 randomized Phase III trial after a median follow-up of 11 yearsJ Clin Oncol201230161926193322529255
  • ParkIJYouYNAgarwalANeoadjuvant treatment response as an early response indicator for patients with rectal cancerJ Clin Oncol201230151770177622493423
  • Habr-GamaAPerezRONadalinWOperative versus nonoperative treatment for stage 0 distal rectal cancer following chemoradiation therapy: long-term resultsAnn Surg2004240471171715383798
  • Habr-GamaAPerezROProscurshimIPatterns of failure and survival for nonoperative treatment of stage c0 distal rectal cancer following neoadjuvant chemoradiation therapyJ Gastrointest Surg200610101319132817175450
  • Habr-GamaAPerezROProscurshimIInterval between surgery and neoadjuvant chemoradiation therapy for distal rectal cancer: does delayed surgery have an impact on outcome?Int J Radiat Oncol Biol Phys20087141181118818234443
  • Habr-GamaAPerezROSabbagaJNadalinWSão JuliãoGPGama-RodriguesJIncreasing the rates of complete response to neoadjuvant chemoradiotherapy for distal rectal cancer: results of a prospective study using additional chemotherapy during the resting periodDis Colon Rectum200952121927193419934911
  • Habr-GamaAPerezROSão JuliãoGPProscurshimIGama-RodriguesJNonoperative approaches to rectal cancer: a critical evaluationSemin Radiat Oncol201121323423921645869
  • Habr-GamaAPerezROLynnPBSao JuliaoGPGama-RodriguesJSelective non-operative management of distal rectal cancer: the Watch and Wait ProtocolSchiesselRMPIntersphincteric Resection for Low Tumors of the RectumNew YorkSpringer-Verlag/Wien20124353
  • Habr-GamaAPerezROWynnGMarksJKesslerHGama-RodriguesJComplete clinical response after neoadjuvant chemoradiation therapy for distal rectal cancer: characterization of clinical and endoscopic findings for standardizationDis Colon Rectum201053121692169821178866
  • MaasMBeets-TanRGLambregtsDMWait-and-see policy for clinical complete responders after chemoradiation for rectal cancerJ Clin Oncol201129354633464022067400
  • SmithJDRubyJAGoodmanKANonoperative management of rectal cancer with complete clinical response after neoadjuvant therapyAnn Surg2012256696597223154394
  • DaltonRSVelineniROsborneMEA single-centre experience of chemoradiotherapy for rectal cancer: is there potential for nonoperative management?Colorectal Dis201214556757121831177
  • YuSKBrownGHealdRJDeferral of rectal surgery following a continued response to preoperative chemoradiotherapy (Watch and Wait) study: a Phase II multicenter study in the United Kingdom [abstract]J Clin Oncol201129suppl 4abstract 489
  • BujkoKNowackiMPNasierowska-GuttmejerAMichalskiWBebenekMKryjMLong-term results of a randomized trial comparing preoperative short-course radiotherapy with preoperative conventionally fractionated chemoradiation for rectal cancerBr J Surg200693101215122316983741
  • NganSYBurmeisterBFisherRJRandomized 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 01.04J Clin Oncol2012130 (31): 3827–3833.
  • KaladyMFde Campos-LobatoLFStocchiLPredictive factors of pathologic complete response after neoadjuvant chemoradiation for rectal cancerAnn Surg2009250458258919710605
  • MohiuddinMWinterKMitchellERadiation Therapy Oncology Group Trial 0012Randomized Phase II study of neoadjuvant combined-modality chemoradiation for distal rectal cancer: Radiation Therapy Oncology Group Trial 0012J Clin Oncol200624465065516446336
  • ChanAKWongAOLangevinJPreoperative chemotherapy and pelvic radiation for tethered or fixed rectal cancer: a Phase II dose escalation studyInt J Radiat Oncol Biol Phys200048384385611020583
  • MohiuddinMRegineWFJohnWJPreoperative chemoradiation in fixed distal rectal cancer: dose time factors for pathological complete responseInt J Radiat Oncol Biol Phys200046488388810705009
  • WiltshireKLWardIGSwallowCPreoperative radiation with concurrent chemotherapy for resectable rectal cancer: effect of dose escalation on pathologic complete response, local recurrence-free survival, disease-free survival, and overall survivalInt J Radiat Oncol Biol Phys200664370971616242252
  • JakobsenAPloenJVuongTAppeltALindebjergJRafaelsenSRDose-effect relationship in chemoradiotherapy for locally advanced rectal cancer: a randomized trial comparing two radiation dosesInt J Radiat Oncol Biol Phys201284494995422592048
  • GerardJPChapetONemozCImproved sphincter preservation in low rectal cancer with high-dose preoperative radiotherapy: the lyon R96-02 randomized trialJ Clin Oncol200422122404240915197202
  • ArbeaLRamosLIMartínez-MongeRMorenoMAristuJIntensity-modulated radiation therapy (IMRT) versus 3D conformal radiotherapy (3DCRT) in locally advanced rectal cancer (LARC): dosimetric comparison and clinical implicationsRadiat Oncol2010265 :17.
  • Guerrero UrbanoMTHenrysAJAdamsEJIntensity-modulated radiotherapy in patients with locally advanced rectal cancer reduces volume of bowel treated to high dose levelsInt J Radiat Oncol Biol Phys200665390791616751073
  • De RidderMTournelKVan NieuwenhoveYPhase II study of preoperative helical tomotherapy for rectal cancerInt J Radiat Oncol Biol Phys200870372873417904302
  • EngelsBTournelKEveraertHPhase II study of preoperative helical tomotherapy with a simultaneous integrated boost for rectal cancerInt J Radiat Oncol Biol Phys201283114214822014952
  • BallonoffAKavanaghBMcCarterMPreoperative capecitabine and accelerated intensity-modulated radiotherapy in locally advanced rectal cancer: a Phase II trialAm J Clin Oncol200831326427018525306
  • FreedmanGMMeropolNJSigurdsonERPhase I trial of preoperative hypofractionated intensity-modulated radiotherapy with incorporated boost and oral capecitabine in locally advanced rectal cancerInt J Radiat Oncol Biol Phys20076751389139317394942
  • SchroederCGaniCLamprechtUPathological complete response and sphincter-sparing surgery after neoadjuvant radiochemotherapy with regional hyperthermia for locally advanced rectal cancer compared with radiochemotherapy aloneInt J Hyperthermia201228870771423006132
  • BarsukovYAGordeyevSSTkachevSIFedyaninMYPerevoshikovAGPhase II study of concomitant chemoradiotherapy with local hyperthermia and metronidazole for locally advanced fixed rectal cancerColorectal Dis Epub5142013
  • RauBWustPHohenbergerPPreoperative hyperthermia combined with radiochemotherapy in locally advanced rectal cancer: a Phase II clinical trialAnn Surg199822733803899527061
  • O’ConnellMJMartensonJAWieandHSImproving adjuvant therapy for rectal cancer by combining protracted-infusion fluorouracil with radiation therapy after curative surgeryN Engl J Med199433185025078041415
  • O’ConnellMJMartensonJAWieandHSEfficacy of intravenous continuous infusion of fluorouracil compared with bolus administration in advanced colorectal cancer. Meta-analysis Group in CancerJ Clin Oncol19981613013089440757
  • HofheinzRDWenzFPostSChemoradiotherapy with capecitabine versus fluorouracil for locally advanced rectal cancer: a randomised, multicentre, non-inferiority, Phase 3 trialLancet Oncol201213657958822503032
  • GérardJPAzriaDGourgou-BourgadeSComparison of two neoadjuvant chemoradiotherapy regimens for locally advanced rectal cancer: results of the Phase III trial ACCORD 12/0405-Prodige 2J Clin Oncol201028101638164420194850
  • AscheleCCioniniLLonardiSPrimary tumor response to preoperative chemoradiation with or without oxaliplatin in locally advanced rectal cancer: pathologic results of the STAR-01 randomized Phase III trialJ Clin Oncol201129202773278021606427
  • RohMSYothersGAO’ConnellMJThe impact of capecitabine and oxaliplatin in the preoperative multimodality treatment in patients with carcinoma of the rectum: NSABP R-04J Clin Oncol201129SupplAbstract 3503
  • RödelCLierschTBeckerHGerman Rectal Cancer Study GroupPreoperative chemoradiotherapy and postoperative chemotherapy with fluorouracil and oxaliplatin versus fluorouracil alone in locally advanced rectal cancer: initial results of the German CAO/ARO/AIO-04 randomised Phase 3 trialLancet Oncol201213767968722627104
  • CraneCHEngCFeigBWPhase II trial of neoadjuvant bevacizumab, capecitabine, and radiotherapy for locally advanced rectal cancerInt J Radiat Oncol Biol Phys201076382483019464823
  • VelenikVOcvirkJOblakIAnderluhFA Phase II study of cetuximab, capecitabine and radiotherapy in neoadjuvant treatment of patients with locally advanced resectable rectal cancerEur J Surg Oncol201036324425020042310
  • DewdneyACunninghamDTaberneroJMulticenter randomized Phase II clinical trial comparing neoadjuvant oxaliplatin, capecitabine, and preoperative radiotherapy with or without cetuximab followed by total mesorectal excision in patients with high-risk rectal cancer (EXPERT-C)J Clin Oncol201230141620162722473163
  • Garcia-AguilarJSmithDDAvilaKBergslandEKChuPKriegRMTiming of Rectal Cancer Response to Chemoradiation ConsortiumOptimal timing of surgery after chemoradiation for advanced rectal cancer: preliminary results of a multicenter, nonrandomized Phase II prospective trialAnn Surg201125419710221494121
  • DasPSkibberJMRodriguez-BigasMAPredictors of tumor response and downstaging in patients who receive preoperative chemoradiation for rectal cancerCancer200710991750175517387743
  • YoonSMKimDYKimTHClinical parameters predicting pathologic tumor response after preoperative chemoradiotherapy for rectal cancerInt J Radiat Oncol Biol Phys20076941167117217967307
  • Moureau-ZabottoLFarnaultBde ChaisemartinCPredictive factors of tumor response after neoadjuvant chemoradiation for locally advanced rectal cancerInt J Radiat Oncol Biol Phys201180248349121093174
  • ParkYASohnSKSeongJSerum CEA as a predictor for the response to preoperative chemoradiation in rectal cancerJ Surg Oncol200693214515016425302
  • ParkCHKimHCChoYBPredicting tumor response after preoperative chemoradiation using clinical parameters in rectal cancerWorld J Gastroenterol201117485310531622219601
  • TulchinskyHShmueliEFigerAKlausnerJMRabauMAn interval >7 weeks between neoadjuvant therapy and surgery improves pathologic complete response and disease-free survival in patients with locally advanced rectal cancerAnn Surg Oncol200815102661266718389322
  • EvansJTaitDSwiftITiming of surgery following preoperative therapy in rectal cancer: the need for a prospective randomized trial?Dis Colon Rectum201154101251125921904139
  • FrancoisYNemozCJBaulieuxJInfluence of the interval between preoperative radiation therapy and surgery on downstaging and on the rate of sphincter-sparing surgery for rectal cancer: the Lyon R90-01 randomized trialJ Clin Oncol1999178239610561302
  • PetterssonDHolmTIversenHBlomqvistLGlimeliusBMartlingAPreoperative short-course radiotherapy with delayed surgery in primary rectal cancerBr J Surg201299457758322241246
  • NCCN Guidelines: Rectal Cancer Version 4.2013 [webpage on the Internet]Fort Washington, PANational Comprehensive Cancer Network2012 Available from: http://www.nccn.org/professionals/physician_gls/pdf/rectal.pdfAccessed March 8, 2013
  • HiotisSPWeberSMCohenAMAssessing the predictive value of clinical complete response to neoadjuvant therapy for rectal cancer: an analysis of 488 patientsJ Am Coll Surg2002194213113511848629
  • GuillemJGChessinDBShiaJClinical examination following preoperative chemoradiation for rectal cancer is not a reliable surrogate end pointJ Clin Oncol200523153475347915908656
  • SmithFMChangKHSheahanKHylandJO’ConnellPRWinterDCThe surgical significance of residual mucosal abnormalities in rectal cancer following neoadjuvant chemoradiotherapyBr J Surg2012997993100122351592
  • PerezROHabr-GamaAPereiraGVRole of biopsies in patients with residual rectal cancer following neoadjuvant chemoradiation after downsizing: can they rule out persisting cancer?Colorectal Dis201214671472022568644
  • HuhJWParkYAJungEJLeeKYSohnSKAccuracy of endorectal ultrasonography and computed tomography for restaging rectal cancer after preoperative chemoradiationJ Am Coll Surg2008207171218589355
  • MarettoIPomerriFPucciarelliSThe potential of restaging in the prediction of pathologic response after preoperative chemoradiotherapy for rectal cancerAnn Surg Oncol200714245546117139456
  • PomerriFPucciarelliSGennaroGMarettoINittiDMuzzioPCComparison between CT volume measurement and histopathological assessment of response to neoadjuvant therapy in rectal cancerEur J Radiol201281123918392422902408
  • PatelUBTaylorFBlomqvistLMagnetic resonance imaging-detected tumor response for locally advanced rectal cancer predicts survival outcomes: MERCURY experienceJ Clin Oncol201129283753376021876084
  • DworakOKeilholzLHoffmannAPathological features of rectal cancer after preoperative radiochemotherapyInt J Colorectal Dis199712119239112145
  • PatelUBBrownGRuttenHComparison of magnetic resonance imaging and histopathological response to chemoradiotherapy in locally advanced rectal cancerAnn Surg Oncol20121992842285222526897
  • PerezROHabr-GamaAGama-RodriguesJAccuracy of positron emission tomography/computed tomography and clinical assessment in the detection of complete rectal tumor regression after neoadjuvant chemoradiation: long-term results of a prospective trial (National Clinical Trial 00254683)Cancer2012118143501351122086847
  • GuillemJGRubyJALeiboldTNeither FDG-PET nor CT is able to distinguish between a pathological complete response and an incomplete response after neoadjuvant chemoradiation in locally advanced rectal cancer: a prospective studyAnn Surg Epub11262012
  • RubyJALeiboldTAkhurstTJFDG-PET assessment of rectal cancer response to neoadjuvant chemoradiotherapy is not associated with long-term prognosis: a prospective evaluationDis Colon Rectum201255437838622426260
  • MartoniAADi FabioFPintoCProspective study on the FDG-PET/CT predictive and prognostic values in patients treated with neoadjuvant chemoradiation therapy and radical surgery for locally advanced rectal cancerAnn Oncol201122365065620847032
  • KuoLJChiouJFTaiCJCan we predict pathologic complete response before surgery for locally advanced rectal cancer treated with preoperative chemoradiation therapy?Int J Colorectal Dis201227561362122080392
  • SongSHongJCMcDonnellSECombined modality therapy for rectal cancer: the relative value of posttreatment versus pretreatment CEA as a prognostic marker for disease recurrenceAnn Surg Oncol20121982471247622327251
  • KimCWYuCSYangSSClinical significance of pre- to post-chemoradiotherapy s-CEA reduction ratio in rectal cancer patients treated with preoperative chemoradiotherapy and curative resectionAnn Surg Oncol201118123271327721537868
  • PerezROSão JuliãoGPHabr-GamaAThe role of carcinoembriogenic antigen in predicting response and survival to neoadjuvant chemoradiotherapy for distal rectal cancerDis Colon Rectum20095261137114319581858
  • KawaiKKitayamaJTsunoNHSunamiEWatanabeTThrombocytosis before pre-operative chemoradiotherapy predicts poor response and shorter local recurrence-free survival in rectal cancerInt J Colorectal Dis201328452753523080345
  • KitayamaJYasudaKKawaiKSunamiENagawaHCirculating lymphocyte number has a positive association with tumor response in neoadjuvant chemoradiotherapy for advanced rectal cancerRadiat Oncol201054720525293
  • KikuchiMMikamiTSatoTHigh Ki67, Bax, and thymidylate synthase expression well correlates with response to chemoradiation therapy in locally advanced rectal cancers: proposal of a logistic model for predictionBr J Cancer2009101111612319491899
  • ArrazubiVSuárezJGuerreroDPrognostic significance of thymidylate synthase polymorphisms in rectal cancer patients treated with neoadjuvant chemoradiotherapyColorectal Dis201315442843522958523
  • SpindlerKLNielsenJNLindebjergJBrandslundIJakobsenAPrediction of response to chemoradiation in rectal cancer by a gene polymorphism in the epidermal growth factor receptor promoter regionInt J Radiat Oncol Biol Phys200666250050416757132
  • SpindlerKLNielsenJNLindebjergJJakobsenAGermline polymorphisms may act as predictors of response to preoperative chemoradiation in locally advanced T3 rectal tumorsDis Colon Rectum20075091363136917661145
  • GiraltJde las HerasMCerezoLGrupo Español de Investigacion Clinica en Oncologia Radioterápica (GICOR)The expression of epidermal growth factor receptor results in a worse prognosis for patients with rectal cancer treated with preoperative radiotherapy: a multicenter, retrospective analysisRadiother Oncol200574210110815816107
  • KimJSKimJMLiSEpidermal growth factor receptor as a predictor of tumor downstaging in locally advanced rectal cancer patients treated with preoperative chemoradiotherapyInt J Radiat Oncol Biol Phys200666119520016839708
  • DvorakJSitorovaVRyskaA[Prognostic significance of changes of tumor epidermal growth factor receptor expression after neoadjuvant chemoradiation in patients with rectal adenocarcinoma.]Strahlenther Onkol20121889833838 German22847519
  • ZlobecIVuongTComptonCCLugliACombined analysis of VEGF and EGFR predicts complete tumour response in rectal cancer treated with preoperative radiotherapyBr J Cancer200898245045618182986
  • ChangHJJungKHKimDYBax, a predictive marker for therapeutic response to preoperative chemoradiotherapy in patients with rectal carcinomaHum Pathol200536436437115891997
  • HuertaSHromJGaoXTissue microarray constructs to predict a response to chemoradiation in rectal cancerDig Liver Dis2010421067968420227932
  • FlugeØGravdalKCarlsenENorwegian Gastrointestinal Cancer GroupExpression of EZH2 and Ki-67 in colorectal cancer and associations with treatment response and prognosisBr J Cancer200910181282128919773751
  • TerziCCandaAESagolOSurvivin, p53, and Ki-67 as predictors of histopathologic response in locally advanced rectal cancer treated with preoperative chemoradiotherapyInt J Colorectal Dis2008231374517805549
  • SturmIRauBSchlagPMGenetic dissection of apoptosis and cell cycle control in response of colorectal cancer treated with preoperative radiochemotherapyBMC Cancer2006612416686938
  • SuzukiTSadahiroSTanakaABiopsy specimens obtained 7 days after starting chemoradiotherapy (CRT) provide reliable predictors of response to CRT for rectal cancerInt J Radiat Oncol Biol Phys20138551232123823158058
  • YanHWangRYuJPredictive value of Smac, VEGF and Ki-67 in rectal cancer treated with neoadjuvant therapyOncol Lett20101464164722966357
  • LinCYTianYFWuLCRsf-1 expression in rectal cancer: with special emphasis on the independent prognostic value after neoadjuvant chemoradiationJ Clin Pathol201265868769222569540
  • Unsal KilicDUnerAAkyurekNErpolatPDursunAPakYMatrix metalloproteinase-9 expression correlated with tumor response in patients with locally advanced rectal cancer undergoing preoperative chemoradiotherapyInt J Radiat Oncol Biol Phys200767119620317071013
  • LochheadPImamuraYMorikawaTInsulin-like growth factor 2 messenger RNA binding protein 3 (IGF2BP3) is a marker of unfavourable prognosis in colorectal cancerEur J Cancer201248183405341322840368
  • YeoSGKimDYKimKHHydroxymethylglutaryl-coenzyme a synthase 2 expression is associated with chemoradiotherapy responses in colorectal cancerDis Colon Rectum201255668669422595849
  • HeYVan’t VeerLJMikolajewska-HanclichIPIK3CA mutations predict local recurrences in rectal cancer patientsClin Cancer Res200915226956696219903786
  • GrimmingerPPBrabenderJWarnecke-EberzUXRCC1 gene polymorphism for prediction of response and prognosis in the multimodality therapy of patients with locally advanced rectal cancerJ Surg Res20101641e61e6620863523
  • SoumaoroLTUetakeHHiguchiTTakagiYEnomotoMSugiharaKCyclooxygenase-2 expression: a significant prognostic indicator for patients with colorectal cancerClin Cancer Res200410248465847115623626
  • KelleySTCoppolaDYeatmanTMarcetJTumor response to neoadjuvant chemoradiation therapy for rectal adenocarcinoma is mediated by p53-dependent and caspase 8-dependent apoptotic pathwaysClin Colorectal Cancer20055211411816098252
  • ChararaMEdmonstonTBBurkholderSMicrosatellite status and cell cycle associated markers in rectal cancer patients undergoing a combined regimen of 5-FU and CPT-11 chemotherapy and radiotherapyAnticancer Res2004245B3161316715510606
  • BrophySSheehanKMMcNamaraDADeasyJBouchier-HayesDJKayEWGLUT-1 expression and response to chemoradiotherapy in rectal cancerInt J Cancer2009125122778278219569052
  • ChenMBWuXYYuRP53 status as a predictive biomarker for patients receiving neoadjuvant radiation-based treatment: a meta-analysis in rectal cancerPLoS One20179e45388
  • GhadimiBMGradeMDifilippantonioMJEffectiveness of gene expression profiling for response prediction of rectal adenocarcinomas to preoperative chemoradiotherapyJ Clin Oncol20052391826183815774776
  • AkiyoshiTKobunaiTWatanabeTPredicting the response to preoperative radiation or chemoradiation by a microarray analysis of the gene expression profiles in rectal cancerSurg Today201242871371922706722
  • WatanabeTKomuroYKiyomatsuTPrediction of sensitivity of rectal cancer cells in response to preoperative radiotherapy by DNA microarray analysis of gene expression profilesCancer Res20066673370337416585155
  • KimIJLimSBKangHCMicroarray gene expression profiling for predicting complete response to preoperative chemoradiotherapy in patients with advanced rectal cancerDis Colon Rectum20075091342135317665260
  • SvobodaMSanaJFabianPMicroRNA expression profile associated with response to neoadjuvant chemoradiotherapy in locally advanced rectal cancer patientsRadiat Oncol2012719523167930
  • KheirelseidEAMillerNChangKHmiRNA expressions in rectal cancer as predictors of response to neoadjuvant chemoradiation therapyInt J Colorectal Dis201328224726022903298
  • Della Vittoria ScarpatiGFalcettaFCarlomagnoCA specific miRNA signature correlates with complete pathological response to neoadjuvant chemoradiotherapy in locally advanced rectal cancerInt J Radiat Oncol Biol Phys20128341113111922172905
  • ZittMZittMMüllerHMDisseminated tumor cells in peripheral blood: a novel marker for therapy response in locally advanced rectal cancer patients undergoing preoperative chemoradiationDis Colon Rectum200649101484149117019657
  • SaelenMGFlatmarkKFolkvordSTumor kinase activity in locally advanced rectal cancer: angiogenic signaling and early systemic disseminationAngiogenesis201114448148921833622
  • AgostiniMPucciarelliSEnzoMVCirculating cell-free DNA: a promising marker of pathologic tumor response in rectal cancer patients receiving preoperative chemoradiotherapyAnn Surg Oncol20111892461246821416156
  • ZittMMüllerHMRochelMCirculating cell-free DNA in plasma of locally advanced rectal cancer patients undergoing preoperative chemoradiation: a potential diagnostic tool for therapy monitoringDis Markers200825315916519096128
  • National Institute for Health and Care Excellence: Colorectal Cancer PathwayLondonNational Institute for Health and Care Excellence2013 Available from: http://pathways.nice.org.uk/pathways/colorectal-cancer#path=view%3A/pathways/colorectal-cancer/managing-local-colorectal-tumours.xml&content=view-node%3Anodes-deciding-whether-to-offer-further-treatmentAccessed April 2, 2013