Your activity: 80 p.v.
your limit has been reached. plz Donate us to allow your ip full access, Email: sshnevis@outlook.com

Neoadjuvant chemoradiotherapy, radiotherapy, and chemotherapy for rectal adenocarcinoma

Neoadjuvant chemoradiotherapy, radiotherapy, and chemotherapy for rectal adenocarcinoma
Authors:
Christopher G Willett, MD
David P Ryan, MD
Section Editor:
Richard M Goldberg, MD
Deputy Editor:
Diane MF Savarese, MD
Literature review current through: Dec 2022. | This topic last updated: Nov 16, 2022.

INTRODUCTION — Surgical resection is the cornerstone of curative treatment for rectal adenocarcinomas. Invasive small rectal adenocarcinomas may be effectively managed with limited surgical procedures, such as local excision. However, the majority of patients have more deeply invasive tumors that require more extensive transabdominal surgery, such as low anterior resection (LAR) or abdominoperineal resection (APR). Others present with locally advanced tumors that are adherent or fixed to adjoining structures, such as the sacrum, pelvic sidewalls, prostate, or bladder. The surgical and oncologic management of these patients varies greatly depending on the stage and location within the rectum. (See "Pretreatment local staging evaluation for rectal cancer" and "Surgical treatment of rectal cancer".)

Tumors in the upper and middle rectum can usually be managed with LAR, coloanal anastomosis, and preservation of the anal sphincter. Although the resulting anorectal function may be imperfect, the risk of postoperative sexual and urinary dysfunction is lessened by preservation of the pelvic autonomic nerves, which is possible in most cases, particularly with total mesorectal excision (TME) (figure 1). (See "Radical resection of rectal cancer", section on 'Total mesorectal excision' and "Surgical treatment of rectal cancer", section on 'Techniques of low anterior resection'.)

Management of adenocarcinomas within 5 cm of the anal verge (figure 2)) poses major challenges. APR has long been considered the standard operation but it entails a permanent colostomy and a high incidence of sexual and urinary dysfunction. (See "Surgical treatment of rectal cancer", section on 'Abdominoperineal resection'.)

Sphincter-sparing approaches for lower rectal adenocarcinomas have evolved along two pathways:

For small superficial tumors that are confined to the rectal wall, local excision may offer local control and survival rates that are comparable with APR, while preserving sphincter function. (See "Surgical treatment of rectal cancer", section on 'Local excision'.)

For larger or more invasive tumors, neoadjuvant therapies have been utilized to promote tumor regression; one potential benefit is increased likelihood of sphincter-sparing surgery, as long as cancer control will not be compromised.

This topic review will discuss neoadjuvant approaches to therapy in patients with rectal adenocarcinoma. Clinical manifestations, diagnosis, and pretreatment staging, surgical management, postoperative adjuvant therapy, and post-treatment follow-up surveillance, as well as management of primary rectal squamous cell cancers, are discussed elsewhere. (See "Clinical presentation, diagnosis, and staging of colorectal cancer" and "Pretreatment local staging evaluation for rectal cancer" and "Radical resection of rectal cancer" and "Surgical treatment of rectal cancer" and "Adjuvant therapy after neoadjuvant therapy for rectal cancer", section on 'Benefit of postoperative chemotherapy' and "Adjuvant therapy for resected rectal adenocarcinoma in patients not receiving neoadjuvant therapy" and "Post-treatment surveillance after colorectal cancer treatment" and "Clinical features and staging of anal cancer".)

INDICATIONS FOR NEOADJUVANT TREATMENT — The only definitive indication for neoadjuvant therapy, supported by the results of randomized trials, is the presence of a clinical (c) T3 or cT4 tumor, as defined by the pretreatment staging evaluation. For these patients, we recommend preoperative (neoadjuvant) CRT or short-course RT rather than initial resection followed by adjuvant therapy. (See 'T3/4 tumors' below and "Pretreatment local staging evaluation for rectal cancer", section on 'Principles of rectal cancer staging by imaging'.)

High-quality evidence to support a clear benefit from neoadjuvant CRT, as compared with initial surgery, for other subgroups of patients with rectal cancer is lacking. However, we also suggest neoadjuvant therapy for other "relative" indications, including:

The presence of clinically node-positive disease in a patient with magnetic resonance imaging (MRI)- or transrectal endoscopic ultrasound (EUS)-staged cT1/2 rectal cancer (see 'T1/2 and clinically node positive' below)

Tumor that appears to invade or "threaten" the mesorectal fascia on preoperative imaging, given the decreased likelihood of achieving a tumor-free circumferential resection margin (CRM) with upfront surgery (see 'Mesorectal fascia involvement' below)

On the other hand, the use of preoperative therapy for a distal clinical T1N0 or T2N0 rectal cancer in an attempt to convert the operation from a needed abdominoperineal resection (APR) into a low anterior resection (LAR) or proctectomy with coloanal anastomosis is controversial and not yet an accepted standard of care. However, if the patient is a poor surgical candidate or declines APR, initial RT or CRT may be chosen, followed by a restaging evaluation with MRI or EUS. (See 'T1-2N0 tumors' below.)

Definitive indications

T3/4 tumors — The only definitive indication for neoadjuvant therapy, supported by the results of randomized trials, is the presence of cT3 or cT4 rectal adenocarcinoma (table 1). These are patients who, if resected initially, would likely require postoperative RT. Data from randomized trials and a meta-analysis [1] suggest that preoperative therapy is associated with a more favorable long-term toxicity profile and fewer local recurrences than postoperative (adjuvant) therapy; overall survival appears similar. (See 'German Rectal Cancer Study Group trial' below and 'Versus surgery alone' below.)

Clinical T3N0 tumors — Given the limitations of current imaging, we consider all patients with cT3N0 rectal adenocarcinoma by EUS or MRI to be appropriate candidates for neoadjuvant therapy, even proximal tumors. However, there is disagreement on this point, and guidelines from both NCCN and ESMO both support upfront surgery for selected patients, such as those with proximal tumors [2,3].

The optimal management of cT3N0 rectal adenocarcinoma (cT3N0 based on preoperative EUS and/or MRI) is debated. Some of these patients have a sufficiently favorable prognosis that questions have been raised as to the necessity of postoperative adjuvant therapy after upfront total mesorectal excision (TME). (See "Adjuvant therapy for resected rectal adenocarcinoma in patients not receiving neoadjuvant therapy", section on 'Can adjuvant therapy be omitted in any patient?'.)

Others have questioned the utility of upfront therapy for patients with cT3N0 tumors that do not threaten the mesorectal fascia, particularly those involving the upper rectum, given the favorable low rates of local recurrence after TME alone in the Dutch TME trial, several retrospective analyses, and one prospective observational study [4-8].

On the other hand, as many as one-fifth of these patients may be understaged by preoperative imaging. In a review of 188 patients with EUS/MRI-staged cT3N0 tumors who received preoperative CRT, 41 (22 percent) were found to have pathologically positive mesorectal lymph nodes at the time of surgery [9]. Given the downstaging effect of CRT, it is likely that an even larger number of these patients would have been found to have node-positive disease (and would have been recommended for postoperative adjuvant therapy) had surgery been undertaken initially.

Issues related to the accuracy of locoregional staging in patients with newly diagnosed rectal cancer are discussed in detail elsewhere. (See "Pretreatment local staging evaluation for rectal cancer".)

Contribution of depth of extramural penetration — The approach of selecting patients with cT3N0 rectal tumors for preoperative therapy based on depth of extramural tumor invasion is not yet standard, at least in the United States, although this approach is supported in guidelines from ESMO [2].

A number of studies have shown that T3 tumors with >5 mm of extramural invasion have a higher rate of nodal involvement [10] and distant metastases as well as a lower cancer-specific survival compared with tumors that have a depth of penetration of 5 mm or less [11-14]. These findings have led some to suggest that it is not necessarily the distinction between T2 and T3 tumors that may potentially govern treatment decisions, but the identification of high-risk T3 tumors with a >5 mm depth of extramural tumor invasion [14,15]. In fact, updated guidelines for treatment of rectal cancer from the European Society for Medical Oncology (ESMO) suggest that patients with a depth of invasion beyond the muscularis propria that is 5 mm or less are appropriate candidates for upfront surgery rather than neoadjuvant therapy, even if they are node positive, as long as the levators are not threatened, the mesorectal fascia is clear, and there is no extranodal extension [2].

While preoperative high-resolution MRI can identify those patients with cT3 tumors and <5 mm of extramural tumor invasion, who may have a good outcome with surgery alone [16], T3 stage subclassification using depth of extramural penetration is not incorporated into tumor, node, metastasis (TNM) staging (table 1), and it is not yet validated as a prognostic factor in rectal cancer. (See "Pretreatment local staging evaluation for rectal cancer", section on 'T3 disease and the depth of extramural invasion'.)

Relative indications

T1/2 and clinically node positive — For patients who have T1 or T2 tumors (table 1) and a suspicion of node-positive disease by MRI or EUS, the determination of "node positivity" can be particularly difficult. Most lymph nodes involved by rectal adenocarcinoma are less than 1 cm, but not all lymph nodes seen with MRI or EUS represent metastatic disease. MRI can assist in the determination of whether small visualized nodes are likely to be malignant by detecting mixed intranodal signal and/or irregularity of the border. (See "Pretreatment local staging evaluation for rectal cancer", section on 'Principles of rectal cancer staging by imaging'.)

Neoadjuvant therapy might be considered an appropriate option for these patients if they had involved lymph nodes. Thus, careful consideration for ultrasound-guided fine needle aspiration biopsy of the lymph nodes should be given to any patient who has a cT1 or cT2 tumor and questionable perirectal lymph nodes.

Mesorectal fascia involvement — Neoadjuvant therapy may also be considered if the preoperative staging evaluation suggests that the tumor invades or "threatens" (ie, is within 1 to 2 mm of) the mesorectal fascia, which is the CRM when TME is performed. Data from several trials indicate that this finding is highly predictive of residual tumor at the CRM, which places the patient at high risk of local recurrence and inferior survival. For anterior tumors, the status of the CRM can be predicted with either EUS or MRI, while for posterior or posterolateral tumors, MRI is the preferred strategy. (See "Pretreatment local staging evaluation for rectal cancer".)

Although there are fewer data to validate this approach, these patients are appropriate candidates for preoperative therapy in an attempt to downstage the tumor and permit a CRM-negative resection. One of the main benefits for preoperative, as compared with postoperative, CRT in the German Rectal Cancer Study Group trial discussed below (which did not focus on patients with evidence of mesorectal fascia involvement) was a reduction in the rate of local recurrence. (See 'German Rectal Cancer Study Group trial' below.)

Distal tumors and sphincter preservation

T3 or node-positive — For patients with distal T3 or node-positive rectal adenocarcinomas, a major goal of preoperative therapy is to convert the surgical procedure from an APR to a sphincter-preserving operation, such as an LAR with coloanal anastomosis [17-21]. Whether or not an APR is required is somewhat subjective, and this makes determining this particular benefit of neoadjuvant therapy difficult to quantify, at least in terms of sphincter preservation. Sphincter preservation rates are highly dependent on the skill and experience of the surgeon, factors that are difficult to document.

The most common method to determine if preoperative therapy has increased the rate of sphincter preservation is through clinical assessment, in which the operating surgeon examines the patient prior to the start of therapy and declares the type of operation required [22]. In the population under study, this rate is then compared with the actual operation that was carried out.

Unfortunately, few reported series provide results from patients who underwent such a prospective assessment by their surgeon and were declared to need an APR before the start of preoperative therapy. The incidence of sphincter preservation in such reports ranges from 39 to 94 percent, averaging 67 percent (table 2) [23-31]. Analyses of local control, survival, and bowel function are encouraging but not definitive.

High-quality definitive evidence is lacking that preoperative therapy can consistently convert patients who need an APR to where a LAR is feasible. The German trial of preoperative versus postoperative CRT demonstrated that patients undergoing preoperative CRT were twice as likely to undergo a sphincter-sparing operation (39 versus 19 percent) [32]. However, the absolute rates of APR in the two cohorts were not significantly different. (See 'German Rectal Cancer Study Group trial' below.)

T1-2N0 tumors — Preoperative CRT or chemotherapy alone followed by local transanal excision might be feasible as an alternative to total mesorectal excision in good responders with cT2N0 distal rectal cancer [31,33-40].

Local excision for high-risk T1-3 rectal cancer remains an area of intense controversy where opinions are based on small experiences in patient cohorts with variable risks of nodal and thus local and systemic recurrence. We continue to advise caution in adopting these approaches as well as the need to have lengthy and complex discussion with patients regarding the potential risks of local recurrence and death. We believe that these risks are often poorly understood by patients and their caregivers. This approach is consistent with guidelines from the National Comprehensive Cancer Network (NCCN), and ESMO, which both consider that transabdominal surgery is the preferred approach in this setting unless the patient is at high surgical risk because of age, frailty, extensive comorbidity, or refuses transabdominal surgery [2,3]. (See "Transanal endoscopic surgery (TES)", section on 'Early rectal cancer'.)

The use of preoperative therapy for a distal clinical T1N0 or T2N0 rectal cancer in an attempt to convert the operation from a needed APR into a LAR or proctectomy with coloanal anastomosis is also controversial and not yet an accepted standard of care. The major difficulty in this situation is that many of these patients, if operated on initially, would be found to have node-negative pathologic T1 to T2 tumors for which neither RT nor chemotherapy would be recommended. At many institutions, the decision to use neoadjuvant therapy commits the treating team to both the RT and a four-month course of postoperative adjuvant chemotherapy. This is because of the significant downstaging that often occurs with neoadjuvant therapy. Patients who are found to have negative nodes following neoadjuvant therapy may have been positive at presentation, and the presence of positive nodes is a marker for an increased risk of systemic recurrence. (See "Adjuvant therapy after neoadjuvant therapy for rectal cancer", section on 'Benefit of postoperative chemotherapy'.)

However, if the patient is a poor surgical candidate or declines APR, initial RT or CRT may be chosen, followed by a restaging evaluation with MRI or EUS. A sphincter-sparing resection should only be undertaken at that time if a negative distal margin can be achieved and if presurgical anorectal sphincter function is adequate. (See "Surgical treatment of rectal cancer", section on 'Local excision' and "Overview of the management of rectal adenocarcinoma", section on 'Stage I disease'.)

PRETREATMENT STAGING EVALUATION — It is vitally important that patients with newly diagnosed rectal cancer are accurately staged prior to treatment. Staging of distant disease extent as well as locoregional staging modalities are discussed in detail elsewhere; the most important issues are summarized below. (See "Clinical presentation, diagnosis, and staging of colorectal cancer" and "Pretreatment local staging evaluation for rectal cancer", section on 'History, physical examination, and endoscopy' and "Endoscopic ultrasound for evaluating patients with rectal cancer".)

A computed tomography (CT) scan to evaluate the liver, retroperitoneal nodes, and chest is indicated in all patients. Serum levels of the tumor marker carcinoembryonic antigen (CEA) should be assayed prior to treatment. Elevated pretreatment CEA levels that do not normalize post-treatment imply the presence of persistent disease and the need for further evaluation. (See "Clinical presentation, diagnosis, and staging of colorectal cancer", section on 'Computed tomography scan' and "Clinical presentation, diagnosis, and staging of colorectal cancer", section on 'Tumor markers'.)

Positron emission tomography scans do not appear to add significant information to CT scans for routine preoperative staging of rectal cancer, but they may be useful in specific circumstances (eg, evaluation of patients who are thought to be present or future candidates for resection of isolated hepatic liver metastases). (See "Clinical presentation, diagnosis, and staging of colorectal cancer", section on 'Positron emission tomography scans'.)

Locoregional staging – The selection of appropriate patients with rectal adenocarcinoma for initial RT or CRT, rather than surgery, is heavily dependent on accurate preoperative locoregional staging, which is mainly accomplished through physical examination, endoscopy, and imaging evaluation with MRI and/or transrectal endoscopic ultrasound (EUS). (See "Pretreatment local staging evaluation for rectal cancer", section on 'History, physical examination, and endoscopy'.)

Either EUS or high-resolution MRI is an acceptable radiographic method to determine preoperative local tumor stage. Both are more accurate than axial CT scans for assessing the depth of tumor invasion, nodal involvement, and the status of the circumferential resection margin (CRM). However, if available, optimal thin-section (high-resolution) MRI using a surface pelvic phased-array coil MRI is generally preferred when patients are being evaluated for neoadjuvant therapy approaches because of its greater utility in assessing the CRM. EUS is only able to assess the status of the CRM for anteriorly located tumors. (See "Endoscopic ultrasound for evaluating patients with rectal cancer", section on 'Circumferential resection margin'.)

This subject is discussed in detail elsewhere. (See "Pretreatment local staging evaluation for rectal cancer", section on 'Imaging evaluation'.)

OVERVIEW OF OUR APPROACH TO NEOADJUVANT THERAPY — Therapy for locally advanced rectal cancer is in evolution, particularly with regard to whether clinical complete responders to neoadjuvant therapy can be safely managed nonoperatively. The following represents our suggested approach to therapy with currently available information, which is outlined in the algorithm (algorithm 1):

Long-course CRT versus short-course RT – For patients with nonmetastatic rectal cancer who are appropriate candidates for neoadjuvant therapy, the best regimen has not been established. For most patients, we suggest conventional fractionation RT with concurrent fluoropyrimidine chemotherapy (ie, long-course CRT) rather than the short-course Swedish approach of RT alone, especially for bulky or T4 tumors. Updated National Comprehensive Cancer Network (NCCN) guidelines consider that neoadjuvant short-course RT (or short-course RT followed by chemotherapy) represents an option (although not preferred over long-course CRT with or without chemotherapy) for T3 any N disease with a clear circumferential resection margin, or T1-2, N1-2 disease [41].

However, outside of the United States, short-course RT has been adopted in many institutions as the standard preoperative approach for operable rectal cancer, and this is an acceptable alternative for many patients. We would not pursue this for a bulky or T4 tumor. Many institutions in the United States are using short-course RT in selected patients, such as those with a borderline performance status or substantial comorbidity who may not to tolerate full-course CRT, as well as prior to surgery in the setting of synchronous metastatic disease to minimize delays in initiating chemotherapy. (See 'Long-course chemoradiotherapy' below and 'Short-course radiotherapy' below and 'Local treatment for patients with distant metastases' below.)

Total neoadjuvant therapy – For most patients with locally advanced rectal cancer who are at high risk for a margin-positive resection (ie, T4 disease or an involved mesorectal fascia), as well as for those with clearly node-positive disease and a low-lying rectal tumor, we suggest total neoadjuvant therapy (TNT; ie, oxaliplatin-based chemotherapy combined with long-course CRT or short-course RT) rather than long-course CRT or short-course RT alone. TNT is associated with increased compliance with chemotherapy (because of the greater tolerability in the preoperative as compared with the postoperative setting), improved local control, and the ability to consider nonoperative treatment if the patient declines surgery. (See 'Total neoadjuvant therapy for locally advanced tumors' below and 'Avoidance of radical surgery' below.)

We generally do not utilize this strategy in patients with lower-risk locally advanced cancers (ie, early cT3N0 disease without a threatened mesorectal fascia, especially involving the upper rectum, distal cT1-2N0 tumors) because these patients may not need chemotherapy at all. (See 'Nonoperative management (watch and wait)' below and 'Total neoadjuvant therapy for locally advanced tumors' below.)

For cT3N0 disease that is low lying and would require either an APR or a very low coloanal anastomosis, the decision to pursue TNT for the primary goal of organ preservation is a complex decision that requires careful explanations of the pros and cons with the patient. It cannot be recommended as the standard of care at this time until datasets become more mature.

If TNT is chosen, and chemotherapy is initiated first, we recommend that clinicians proceed cautiously with scans after two months of chemotherapy and move directly to CRT if no response is seen. This is particularly important in the setting of a tumor with deficient mismatch repair/high levels of microsatellite instability, a substantial proportion of which may be resistant to neoadjuvant chemotherapy. During the chemotherapy portion of treatment, we suggest use of an oxaliplatin-based chemotherapy regimen rather than a fluoropyrimidine alone. Options include FOLFOX or FOLFIRINOX, as was used in the PRODIGE 23 study. We would not administer more than four months of preoperative oxaliplatin-based therapy. (See 'Total neoadjuvant therapy for locally advanced tumors' below.)

Nonoperative management for complete clinical responders – Nonoperative management for clinical complete responders to conventional neoadjuvant therapy is a controversial issue. Despite the lack of randomized trials, nonoperative management is becoming an acceptable alternative for those patients who experience a clinical complete response (cCR) to neoadjuvant therapy, especially TNT. It should be emphasized that mature data are not yet available to ensure that survival outcomes are equivalent, and this is particularly important given the natural history of rectal cancer and the rate of late recurrences between 5 and 10 years after resection.

Year 2020 consensus-based guidelines from the NCCN state that for patients who achieve a cCR with no evidence of residual tumor on digital rectal examination (DRE), rectal MRI, and direct endoscopic evaluation, an initial nonoperative approach may be considered with an experienced multidisciplinary team. However, there is not uniform agreement on whether nonoperative approaches should be considered standard at this time, even among the authors and editors associated with this topic review. Given the degree to which the risk of local and distant relapse has not been adequately characterized, any decision for nonoperative management should involve a careful discussion with the patient as to their risk tolerance. (See 'Avoidance of radical surgery' below.)

Deficient mismatch repair and upfront immunotherapy – All patients should be tested for deficient mismatch repair (dMMR; both germline and somatic tumor testing). Upfront immunotherapy is not yet a standard approach for treatment of locally advanced dMMR rectal cancer, but it may be chosen as a non-standard approach by patients who place a high value on potentially avoiding pelvic RT and surgery. (See 'Neoadjuvant immunotherapy for dMMR tumors' below.)

TREATMENT OPTIONS FOR NONMETASTATIC DISEASE

Long-course chemoradiotherapy — Largely based on the German Rectal Cancer Study Group trial, neoadjuvant CRT with conventional fractionation RT has become a standard approach to treatment of rectal adenocarcinoma in the United States. (See 'Indications for neoadjuvant treatment' above and "Pretreatment local staging evaluation for rectal cancer".)

Several randomized trials and at least two meta-analyses have demonstrated that concurrent administration of chemotherapy with conventional fractionation RT is critical to the success of this approach, at least in terms of local failure rates [42-46]. As a result, the addition of chemotherapy to conventional fractionation RT has become a standard approach to neoadjuvant CRT.

Clinical trials of neoadjuvant CRT have focused on two main questions: the comparative benefit of preoperative versus postoperative CRT and the optimal chemotherapy component.

Preoperative versus postoperative therapy

German Rectal Cancer Study Group trial — Adjuvant CRT had been the standard of care for resectable rectal cancer until a seminal trial from Germany firmly established the role of neoadjuvant CRT. (See "Adjuvant therapy for resected rectal adenocarcinoma in patients not receiving neoadjuvant therapy".)

The seminal German Rectal Cancer Study Group trial randomly assigned 823 patients with clinically staged T3/4 or node-positive rectal cancer to the same CRT regimen administered either preoperatively or postoperatively: 50.4 Gy in 28 daily fractions to the tumor and pelvic lymph nodes concurrent with infusional fluorouracil (FU; 1000 mg/m2 daily for five days during the first and fifth weeks of RT) [32]. All patients underwent total mesorectal excision (TME) and four additional cycles of adjuvant single-agent FU (500 mg/m2 bolus daily for five days every four weeks). The patients randomly assigned to postoperative treatment all received an RT boost of 5.4 Gy. Of note, only 5 percent of the patients in either group had T1/2 node-positive tumors.

At a median follow-up of 46 months, preoperative CRT was associated with a significantly lower pelvic relapse rate (6 versus 13 percent with postoperative therapy); the difference persisted with longer follow-up, although it was of a lower magnitude at 10 years (7 versus 10 percent) [47]. The five-year disease-free survival (DFS; 68 versus 65 percent) and overall survival rates (76 versus 74 percent) were similar for preoperative and postoperative therapy, respectively; 10-year rates were also comparable (DFS approximately 68 percent in both groups, overall survival approximately 60 percent in both groups).

Stage distribution at surgery was suggestive of significant downstaging effects. The American Joint Committee on Cancer (AJCC)/Union for International Cancer Control (UICC) stage distribution (I to IV) was 25, 29, 25, and 6 percent for the preoperative group, compared with 18, 29, 40, and 7 percent in the postoperative group. Among the 194 patients with low-lying tumors who were thought preoperatively to require an abdominoperineal resection (APR), those undergoing preoperative CRT were twice as likely to undergo a sphincter-sparing operation (39 versus 19 percent).

Other trials

NSABP R-03 – Preoperative CRT was directly compared with postoperative CRT in National Surgical Adjuvant Breast and Bowel Project (NSABP) protocol R-03, which randomly assigned patients with operable rectal cancer (regardless of stage) to one of two groups [48]:

Preoperative therapy, consisting of one cycle of weekly bolus FU and leucovorin (LV) for six weeks, two courses of FU and LV (daily for five days during the first and fifth course of RT) concomitant with pelvic irradiation (50.4 Gy), surgery, and then four cycles of postoperative weekly bolus FU and LV.

Postoperative therapy, which consisted of surgery, one cycle of weekly bolus FU plus LV, two cycles of FU and LV concomitant with pelvic RT (as described above), and then four cycles of weekly bolus FU and LV.

Accrual did not reach planned levels, and the protocol was closed early. In the final analysis of 267 enrolled patients, preoperative therapy was associated with a significantly higher rate of five-year DFS (65 versus 53 percent), but only a trend toward better overall survival (75 versus 66 percent, p = 0.065), and no difference in locoregional control (five-year cumulative incidence of locoregional recurrence 11 percent in both arms).

Korean trial – A specific benefit for preoperative CRT could not be shown in a Korean trial that compared preoperative and postoperative CRT using capecitabine alone (1650 mg/m2 daily during RT) in 240 patients with cT3 or node-positive rectal cancer [49]. Only four patients total had T2N+ tumors. All patients received four cycles of postoperative capecitabine (2500 mg/m2 daily). The pCR rate was 17 percent after preoperative CRT. At a median follow-up of 52 months, the three- and five-year DFS rates were similar in both groups, as were the cumulative rates of local recurrence (3 versus 2 percent). The sphincter-sparing rates were similar in the two groups, although among patients with low-lying tumors, the preoperative CRT arm had a higher rate of sphincter-sparing surgery (68 versus 42 percent).

Prognosis and extent of tumor regression

Impact of postoperative stage on prognosis – Data from the German Rectal Cancer Study Group trial and others indicate that survival in patients with rectal cancer undergoing neoadjuvant therapy is driven by the post-therapy pathologic (yp) stage and not the pretherapy clinical (c) stage.

Two reports from the German Rectal Cancer Study Group focused on prognostic stratification in patients undergoing neoadjuvant CRT [50,51]. Prognosis was related to the final tumor (T) stage in the surgical specimen as well as the presence of involved lymph nodes in the surgical specimen (table 3). According to the tumor, node, metastasis (TNM) staging system of the combined AJCC/UICC, the "y" prefix added to the pathologic (p) stage designates a TNM stage that is assigned after multimodality therapy (table 1). (See "Pathology and prognostic determinants of colorectal cancer", section on 'Tumor regression after neoadjuvant therapy'.)

The poor outcomes seen in patients with ypN2 disease in this and other reports [52-55] suggest that these patients are good candidates for novel treatment approaches, such as expanded postoperative chemotherapy, although this is not yet a standard approach.

Tumor regression grade – Prognosis has also correlated with tumor regression grade (TRG), which incorporates the degree of fibrosis [56,57] as well as the percentage of viable tumor cells (table 4) [50,51,58]. The better prognosis with higher TRG was maintained with long-term follow-up (table 3) [51].

Of note, this is only one of several available and commonly used systems to assess TRG (table 5). Of the various TRG systems in use, some score only the presence of residual tumor, while others assess residual tumor plus any residual tissue taking the place of tumor (eg, fibrosis, inflammation, pools of acellular mucin, necrosis, and/or calcification). In addition, the tier systems range from three to five. The TRG system used by the combined AJCC/UICC (table 6) [59] for surgical specimens after preoperative RT or CRT for rectal cancer (which was recommended in College of American Pathologists [CAP] guidelines [60]) differs from that used by the German Rectal Cancer Study Group. However, one analysis concluded that the AJCC staging manual system is the most accurate and should be adopted as the standard [61]. The AJCC/CAP TRG system has been validated independently in a series of patients undergoing long-course CRT followed by surgery for mid to low rectal adenocarcinoma [62].

Regardless of which system is used, whether and how TRG could be used to modify the postoperative treatment strategy (eg, whether adjuvant chemotherapy could be avoided in those with a favorable TRG) are not clear. (See "Adjuvant therapy after neoadjuvant therapy for rectal cancer", section on 'Benefit of postoperative chemotherapy'.)

Importantly, TRG scores have not accounted for the possible involvement of lymph nodes. Accumulating data support the view that patients with initially clinically positive perirectal lymph nodes who have pathologically negative lymph nodes after preoperative RT or CRT have an excellent outcome [63,64]. The correlation between the different values of TRG and the incidence of positive nodes is an area of active investigation [65].

The use of imaging-based preoperative TRG stratification systems to select patients for nonoperative management after neoadjuvant therapy is discussed in detail below. (See 'Tumor response assessment and follow-up' below.)

Assessing treatment response and timing of surgery — Most patients undergoing long-course CRT undergo reimaging of the primary site approximately four to six weeks after the end of treatment to assess treatment response.

Responders – The optimal interval between completion of neoadjuvant long-course conventional fractionation CRT and surgery in rectal adenocarcinoma is not established. However, for most patients who have a radiographic response to CRT as determined by repeat post-treatment imaging, we suggest surgical resection within 7 to 10 weeks following the completion of CRT. Waiting a longer period of time may not improve the pCR rate, and the delay may increase the difficulty of surgical resection and the postoperative morbidity.

Traditionally, the interval between completion of neoadjuvant conventional fractionation CRT and surgery in rectal adenocarcinoma has been six weeks (approximately 11 to 12 weeks after the start of RT) as this was the duration used in the seminal German Rectal Cancer Study Group trial [32]. (See 'German Rectal Cancer Study Group trial' above.)

However, the process of tumor regression takes time. In one study examining this issue, a tumor of size 54 cm3 would require an interval of 20 weeks from after the start of treatment to surgery to regress to <0.1 cm3 (10 volume-halving times = 140 days) [66]. These results suggest that there might be a benefit to delaying surgery beyond six weeks.

To date, at least five randomized trials have examined the time interval between CRT and surgery [67-71], only two of which have shown a higher pCR rate with longer as compared with shorter wait times [68,71]. In neither trial did higher pCR rates with delayed surgery translate into better oncologic outcomes (survival, recurrence).

A meta-analysis of data from four of these trials and 22 other nonrandomized series (totaling 25,445 patients) came to the following conclusions [68-72]:

Compared with a standard six- to eight-week interval from completion of neoadjuvant RT to surgery, an interval of eight weeks or more was associated with greater odds of a pCR (odds ratio [OR] 1.41, 95% CI 1.30-1.52) and tumor downstaging (mainly the T stage, OR 1.33, 95% CI 1.04-1.72), but no differences in rates of complete (R0) resection, sphincter preservation, or complication.

The higher rate of a pCR translated into reduced distant metastases (OR 0.71, 95% CI 0.54-0.93) and overall recurrences (OR 0.76, 95% CI 0.58-0.98), but not reduced local recurrences (OR 0.83, 95% CI 0.49-1.42) or overall survival (OR 1.02, 95% CI 0.71-1.45).

Not surprisingly, consensus-based guidelines on this issue are discordant:

Updated guidelines from the European Society for Medical Oncology (ESMO) provide no specific recommendation other than to state that in practice, there is wide variation in the timing of surgery (4 to 12 weeks), and that longer intervals may enhance pCR rates, but this risks repopulation, delays the use of postoperative chemotherapy, and risks subsequent metastases [2].

National Comprehensive Cancer Network (NCCN) [41] guidelines suggest that surgery be performed 5 to 12 weeks following full-dose neoadjuvant CRT.

Nonresponders – Some patients will not achieve meaningful regression in tumor size or stage with neoadjuvant CRT; in this group, outcomes may be poorer if surgery is delayed [73]. Patients who do not respond well to neoadjuvant CRT should be identified early after the end of CRT and undergo surgery without delay.

Complications — As long as waiting times are fewer than 11 weeks, the available data suggest that preoperative CRT does not increase the perioperative complication rate from surgical resection [32,48,74,75]. In the German study described above (a six-week waiting time between the end of CRT and surgery), the incidence of grade 3 or 4 gastrointestinal toxicity was similar in both the preoperative and postoperative CRT groups (28.8 versus 31.7 percent, respectively), and postoperative morbidity rates were not higher with neoadjuvant therapy [32]. Compared with postoperative RT, significantly fewer patients undergoing neoadjuvant therapy had chronic anastomotic strictures (2.7 versus 8.5 percent).

On the other hand, data from the randomized GRECCAR-6 trial (discussed above) suggest that the perioperative complication rate may be higher among those who undergo delayed surgery at 11 as compared with 7 weeks after completing CRT. (See 'Assessing treatment response and timing of surgery' above.)

However, RT is associated with late adverse effects:

Multimodality therapy negatively affects anorectal function after TME. In a meta-analysis of 25 studies examining long-term functional outcomes after rectal cancer resection with preoperative (chemo)radiotherapy, the majority of studies (14 of 18) reported higher rates of anorectal dysfunction after preoperative (chemo)radiotherapy [76]. In particular, fecal incontinence occurred more often in irradiated patients (risk ratio [RR] 1.67, 95% CI 1.36-2.05), and manometric results (mean resting pressures, maximum squeeze pressures) were significantly worse relative to patients undergoing TME alone. The contribution of RT to bowel and anorectal problems after treatment for colorectal cancer is discussed in detail elsewhere. (See "Approach to the long-term survivor of colorectal cancer", section on 'Bowel and anorectal problems'.)

Patients undergoing treatment for rectal cancer are also at risk for sexual dysfunction. The risk is higher in those undergoing APR compared with low anterior resection (LAR), in those undergoing non-TME versus TME surgery, and in patients who undergo combined modality treatment that includes RT. The independent contribution of surgical technique (and of nerve preservation in particular) versus that of RT and whether preoperative as compared with postoperative CRT alters rates of sexual dysfunction after treatment for rectal cancer are unknown. (See "Approach to the long-term survivor of colorectal cancer", section on 'Sexual dysfunction' and "Management of intra-abdominal, pelvic, and genitourinary complications of colorectal surgery", section on 'Sexual dysfunction'.)

Sacral insufficiency fractures are a relatively uncommon late complication of pelvic RT [77-79]:

In one report, the incidence of sacral insufficiency fractures at three years after preoperative CRT was 3 percent overall; however, women appeared to be at higher risk (5.8 versus 1.6 percent in men) [77].

A higher rate of sacral insufficiency fractures (7.1 percent) was reported in a retrospective review of 492 patients treated with CRT for rectal cancer over a nine-year period [79]. Independent risk factors associated with fracture were osteoporosis, female sex, and age greater than 60 years.

Choice of chemotherapy during RT

Fluoropyrimidines — For most patients, we recommend infusional FU (on the days when RT is delivered, five days per week) rather than bolus FU during neoadjuvant RT. Daily capecitabine is an appropriate alternative.

Infusional versus bolus FU – Although the techniques and dose of irradiation are similar in published CRT studies, there is marked variability in the administration of chemotherapy. Some studies (eg, the German trial described above [32]) use bolus fluorouracil (FU) alone, while others use bolus LV-modulated FU [42,43] for five consecutive days during the first and last weeks of RT.

Retrospective data suggest that infusional, rather than bolus, FU during RT increases the likelihood of a pCR in patients with locally advanced rectal cancer [80]. However, the current preference at many institutions, including ours, for concomitant infusional FU during RT is based mainly on an intergroup study that demonstrated superiority of adjuvant concurrent infusional FU compared with bolus FU during RT in patients with resected rectal cancer. (See "Adjuvant therapy for resected rectal adenocarcinoma in patients not receiving neoadjuvant therapy".)

CapecitabineCapecitabine has been shown to be therapeutically equivalent to infusional FU when used during concomitant CRT, albeit with a different toxicity profile [81-84]. As an example, a phase III German trial directly compared CRT (50.4 Gy) with concomitant capecitabine (825 mg/m2 twice daily on days 1 to 38) versus infusional FU (1000 mg/m2 by continuous infusion on days 1 to 5 and 29 to 33) in 401 patients with locally advanced rectal cancer who were treated in the neoadjuvant setting [81]. At a median follow-up of 52 months, the local recurrence rate was similar (6 versus 7 percent with infusional FU), but the distant metastasis rate was lower with capecitabine (19 versus 28 percent). Capecitabine was not inferior to FU for five-year overall survival (the primary endpoint). Patients in the capecitabine group had more hand-foot skin reactions, fatigue, and proctitis than did those in the FU group, whereas leucopenia was more frequent with FU.

One concern is that capecitabine metabolism is variable, and systemic exposure to capecitabine correlates poorly with efficacy and toxicity [85]. The variable bioavailability of oral fluoropyrimidines in individual patients raises concerns as to adequate dosing of these agents. However, if capecitabine is chosen, it is reasonable to use 825 mg/m2 twice daily, five days per week, during RT. Similarly, based on the administration schedule adopted in the NSABP R-04 trial [82], if infusional FU is chosen, we limit the chemotherapy treatment to the days that RT is administered (ie, Monday through Friday), rather than continuous infusion, because of better tolerability.

Oxaliplatin — Outside of the context of a clinical trial, we suggest not adding oxaliplatin to fluoropyrimidine-based CRT. Toxicity is clearly worse compared with CRT using a fluoropyrimidine alone and efficacy is not yet proven.

Oxaliplatin has become an important component of treatment for advanced colorectal cancer; in addition, oxaliplatin plus FU and LV outperforms FU/LV alone in adjuvant treatment of stage III colon cancer and has been adopted as a standard regimen. (See "Systemic therapy for nonoperable metastatic colorectal cancer: Selecting the initial therapeutic approach" and "Adjuvant therapy for resected stage III (node-positive) colon cancer".)

Data from at least 10 randomized trials of the addition of a platinum drug (nine oxaliplatin, one cisplatin) to fluoropyrimidine-based CRT in a total of 5599 patients with stage II or III rectal cancer [82,83,86-97] were the subject of a systematic review and meta-analysis [98], which came to the following conclusions:

The addition of a platinum derivative significantly increased the likelihood of a pCR at the time of surgery (OR 1.31, 95% CI 1.10-1.55) and reduced the likelihood of a distant recurrence (OR 0.78, 95% CI 0.66-0.92).

These benefits did not translate into improvements in overall survival (hazard ratio [HR] for death 0.93, 95% CI 0.82-1.05), DFS (HR 0.91, 95% CI 0.83-1.01), or local recurrence (OR 0.83, 95% CI 0.66-1.05).

The addition of a platinum agent increased rates of grade 3 or 4 toxicities, including diarrhea, nausea, neurosensory toxicity, and fatigue.

Agents that are not used — Outside of the context of a clinical trial, we recommend not adding irinotecan, or inhibitors of angiogenesis, or the epidermal growth factor receptor (EGFR) to fluoropyrimidine-based CRT.

Nonrandomized trials suggested benefit from adding irinotecan to the CRT regimen [99-101], but benefit could not be shown in a RTOG trial in which 106 patients with T3/4 distal rectal cancer were randomly assigned to continuous infusional FU (225 mg/m2 daily) concurrent with hyperfractionated RT (55.2 to 60 Gy at 1.2 Gy twice daily) or to infusional FU (225 mg/m2 daily, five days per week) plus irinotecan (50 mg/m2 once weekly for four weeks) and concurrent conventional fractionation RT (50.4 to 54 Gy in daily 1.8 Gy fractions) [102]. For surgically-treated patients, the pCR rate was similar in both arms (30 versus 26 percent with irinotecan), as were rates of acute and late toxicity.

Most (but not all) early reports suggest that the addition of bevacizumab (a humanized monoclonal antibody targeting the vascular endothelial growth factor [VEGF]) or aflibercept (a recombinant fusion protein that functions as a decoy receptor, preventing VEGFA, VEGFB, and placenta growth factor [PlGF] from binding to their receptors) to conventional FU-based CRT provides encouraging pCR rates, although the data on postoperative complications have been mixed [103-107]. However, the impact of this strategy on long-term outcomes, especially post-treatment complications, awaits the development and completion of phase III trials.

Early reports are also mixed regarding the benefit of adding cetuximab or panitumumab, two humanized monoclonal antibodies targeting the epidermal growth factor receptor (EGFR), to conventional FU-based CRT [108-113]. The impact of this strategy on long-term outcomes, especially post-treatment complications, awaits the completion of phase III studies.

Radiation technique — In the treatment of abdominopelvic tumors, careful attention must be paid to RT technique. It is generally not appropriate to treat rectal tumors with anteroposterior-posteroanterior (AP-PA) fields alone. With this field arrangement, the anterior structures receive a high dose; since these structures are not substantially at risk for local failure, the two-field technique results in increased treatment-related toxicity without a significant improvement in local control. At our institutions, we generally treat patients with a four-field box technique using three-dimensional conformal RT (3D-CRT), although a three-field technique with right and left lateral fields and a posterior field is also reasonable. The use of lateral fields permits sparing of a portion of the bladder and some anteriorly placed small bowel.

Dose and schedule – Patients are treated in the prone position. For those with disease limited to the submucosa and muscularis propria, pelvic irradiation to 45 Gy in 25 fractions is given using a four-field technique, followed by the first field reduction, with lateral fields to 50.4 Gy, and a second field reduction (if appropriate), with lateral fields to 54 Gy. Patients are treated at 1.8 Gy per day in five fractions per week. For the initial fields (45 Gy), the superior border should be 1.5 cm above the level of the sacral promontory, and the lower border of the field should be 4 to 5 cm below the defined tumor bed. Laterally, the AP-PA fields extend 1 to 1.5 cm beyond the true bony pelvis. To treat the entire presacral space with adequate margins and the full dose, the lateral fields are designed so that the posterior border encompasses the entire sacrum with a 1 cm margin posterior to the sacrum. Anteriorly, the fields are designed to encompass the previous tumor bed, including the posterior wall of the vagina for females and a large portion of the prostate for males.

After 45 Gy, lateral fields with an approximately 3 cm margin around the marked tumor bed are typically used for three fractions to bring the dose to 50.4 Gy. This is followed by a further field reduction to a 2 cm margin around the marked tumor bed to bring the dose to 54 Gy. A small bowel series must be performed to ensure that no small bowel is within these lateral boost fields.

Based on patterns of failure in contemporary series analyzing patients who have undergone TME and sphincter-conservation surgery, some authors have suggested a modification in the clinical target volume to reduce the amount of normal tissue in the treated field [114].

Role of intensity-modulated RT – Intensity-modulated RT (IMRT) is an advanced form of 3D-CRT that changes the intensity of radiation in different parts of a single radiation beam while the treatment is delivered. IMRT relies on computer control capabilities to maximize the delivery of radiation to the planned treatment volume while minimizing radiation to normal tissue outside the target. Early data from retrospective series and phase II trials suggest good compliance, low rates of acute bowel toxicity, and high pCR rates after preoperative IMRT with concurrent fluoropyrimidine therapy [115-122].

However, there are no randomized trials comparing IMRT with conventional 3D-CRT, and its routine use cannot yet be recommended. Where available, we would consider use of IMRT in unique clinical situations that require extreme conformality of radiation to the target (rectal cancer, the mesorectum, and nodes) with avoidance of surrounding normal tissues (small bowel, bladder, pelvic bone) because of prior pelvic RT or underlying comorbidities (eg, inflammatory bowel disease).

Short-course radiotherapy — In many countries, short-course RT has been adopted as the standard preoperative approach for operable rectal adenocarcinomas based on two randomized trials demonstrating that this approach is associated with long-term outcomes comparable to those with long-course CRT. However, in many institutions, particularly in the United States, long-course CRT is still the preferred approach for most patients, particularly those with T4 and/or bulky tumors or if the circumferential resection margin (CRM) or complete resection status are predicted to be at risk. Updated NCCN guidelines consider that neoadjuvant short-course RT (or short-course RT followed by chemotherapy) represents an option (although not preferred over long-course CRT with or without chemotherapy) for T3 any N disease with a clear CRM, or T1-2, N1-2 disease [41].

Many institutions in the United States are using short-course RT prior to rectal surgery in selected patients, such as those thought not to be able to tolerate long-course CRT, and in the setting of metastatic disease to minimize delays in initiation of systemic therapy. (See 'Synchronous unresectable metastases' below.)

Short-course RT has been compared with surgery alone, and with conventional long-course CRT.

Versus surgery alone — At least three phase III trials have demonstrated improved local control in patients with rectal cancer receiving short-course preoperative RT followed by surgery versus surgery alone but more long-term side effects [4,5,21,123]. As examples:

The early Swedish study randomly assigned 1168 patients to receive 25 Gy delivered in five fractions in one week followed by surgery versus surgery alone [21,123]. After five years, RT was associated with significant improvements in both local control (89 versus 73 percent) and overall survival (58 versus 48 percent). With long-term follow-up, the number of hospital admissions for gastrointestinal disorders (bowel obstruction, abdominal pain) was two- to fourfold higher in irradiated patients [123].

In a later Dutch trial in which 1861 patients with resectable rectal cancer were randomized to TME alone or high-dose-rate RT (5 x 5 Gy) followed by TME, preoperative RT was associated with an improvement in the five-year local recurrence rate compared with TME alone (5.6 versus 10.9 percent) but similar five-year survival (64 percent in both groups) [4,5]. Despite comparable rates of intraoperative and postoperative complications overall, the irradiated group had significantly more perineal wound problems following APR than those undergoing surgery alone (29 versus 18 percent) [124]. They also had more sexual dysfunction and slower recovery of bowel function [125], and with long-term follow-up, they had more fecal incontinence (62 versus 38 percent), anal blood loss (11 versus 3 percent), dissatisfaction with bowel function [126], and problems with erections [127].

Versus conventional chemoradiotherapy — At least three randomized trials comparing short-course Swedish-style RT with conventional fractionation long-course CRT have concluded that the rates of local recurrence (at least for non-distal tumors), DFS, distal recurrence, overall survival, and toxicity are comparable [128-130]. The two largest trials are described below:

A Polish trial involving 316 patients with T3/4 rectal cancer compared conventional fractionation RT (50.4 Gy) in conjunction with bolus FU and LV during weeks 1 and 5 versus short-course RT (5 x 5 Gy fractions with surgery within seven days of the last RT dose) [128]. The pCR rate was significantly higher in the conventional CRT group (16 versus 1 percent), and there were fewer cases of radial margin positivity (4 versus 13 percent), but the rate of sphincter preservation in both groups was comparable (58 and 61 percent, respectively). Early radiation toxicity was higher in the CRT group (18 versus 3 percent), but rates of local recurrence (9 versus 14 percent), DFS (58 versus 56 percent), and severe late toxicity (10 versus 7 percent) were not significantly different.

The Trans-Tasman Radiation Oncology Group (TROG) 01.04 trial, randomly assigned 326 patients to short-course RT (5 x 5 Gy fractions) or conventional fractionation CRT (50.4 Gy with concomitant infusional FU) [129]. Patients in the short-course RT arm also received six courses of postsurgical adjuvant chemotherapy, while those in the conventional fractionation CRT group received four courses. As with the prior trial, the pCR rate was significantly higher with long-course CRT (15 versus 1 percent), but no difference in rates of margin positivity or sphincter preservation. At a median follow-up of nearly six years, patients receiving long-course CRT had a small but statistically insignificant lower cumulative local recurrence rate at both three years (4.4 versus 7.5 percent) and five years (5.7 versus 7.5 percent). There were also no significant differences in the rates of distant recurrence, relapse-free survival, overall survival, or late grade 3 or 4 toxicity.

A subset analysis of the 79 patients with distal tumors treated on the TROG 01.04 trial revealed a cumulative incidence of local recurrence of 12.5 percent for short-course RT and 0 percent for long-course CRT. However, a later meta-analysis of this and three other trials [128,131,132] concluded that the difference in local failure rate for low-lying tumors (<5 cm from the anal verge) with short-course RT versus long-course CRT was not significantly different (pooled OR for local failure 0.87, 95% CI 0.53-1.44) [133].

Additional data on the benefits of short-course RT used as a component of total neoadjuvant therapy are discussed below. (See 'Total neoadjuvant therapy for locally advanced tumors' below.)

Timing of surgery — There is no widely accepted "standard" approach to timing of surgery after short-course RT and clinical practice is variable. In our view, if neoadjuvant chemotherapy is not used, either immediate surgery (<10 days after the first RT fraction) or delayed surgery (four to eight weeks after the end of RT) is acceptable. Specific recommendations for patients undergoing short-course RT as a component of total neoadjuvant therapy are discussed below. (See 'Sequencing of RT and chemotherapy' below.)

The optimal timing of surgery after short-course RT is debated, and there is no consensus as to the best approach. Most of the major trials described above, which compared short-course RT followed by surgery versus surgery alone, or short-course RT versus long-course CRT prior to surgery, used an interval to surgery of seven days after completion of short-course RT [21,128,129,134]. However, in the Dutch TME trial, the median interval between the end of short-course RT and surgery was 21 days [4]. An alternative approach is to delay surgery for four to eight weeks, with the possibility of enhanced tumor downstaging and at least some pCRs [135-138]. On the other hand, other studies have suggested that an even shorter interval between the end of short-course RT and surgery (ie, zero to three days) might be beneficial, hypothesizing that a poor leukocyte response or treatment-related leukopenia might be contributory to worse outcomes when surgery is delayed, even by one week [139-141].

The Stockholm III trial, a noninferiority trial that directly compared short-course RT with immediate surgery, short-course RT with delayed (four to eight weeks) surgery, and long-course RT with delayed (four to eight weeks) surgery in 843 patients with resectable primary rectal cancer [142]. It appeared to be oncologically safe to delay surgery for four to eight weeks after the end of short-course RT, with fewer postoperative complications than with short-course RT with surgery within a week. However, the trial took 15 years to accrue and was amended after one year to allow participating hospitals to accrue only to the two short-course RT arms, resulting in imbalances among the groups. Furthermore, there was no concurrent chemotherapy in the long-course RT arm, and very few patients in any of the arms received adjuvant chemotherapy (15, 13, and 19 percent of those in the short-course RT with immediate surgery, short-course RT with delayed surgery, and long-course RT arms, respectively). The use of neoadjuvant chemotherapy was not reported. These deficiencies render the results from the intention-to-treat analysis uninterpretable.

The optimal interval between the end of short-course RT and surgery was later addressed in a secondary analysis of 810 patients who were randomized in the Stockholm III trial and analyzed as treated (not intended); patients were stratified according to the overall treatment time (OTT; defined as the number of days between the start of RT and surgery) [143]. Among the patients receiving short-course RT, there were significantly more complications in those with an OTT of 8 to 13 days than in those with an OTT of 7 days, 5 to 7 weeks, or 8 to 13 weeks. When compared with patients with an OTT of 8 to 13 days, the risks of overall and any infectious complications were significantly lower in those with an OTT of 5 to 7 or 8 to 13 weeks. The risk of any surgical complications was reduced in those with an OTT of 8 to 12 weeks compared with those with an OTT of 8 to 13 days. There were no differences in 30- or 90-day mortality rates among the groups. Oncologic outcomes were not reported. Based on this analysis, the authors concluded that surgery should optimally be delayed for 4 to 12 weeks after the end of short-course RT.

ESMO guidelines suggest that either approach (immediate surgery <10 days after the first RT fraction or delayed surgery four to eight weeks after the end of RT) is acceptable [2].

Clinical T4 and/or large bulky tumors — Short-course RT has generally not been pursued for patients with cT4 or large bulky tumors. However, encouraging results have been noted in several trials for short-course RT in conjunction with systemic induction chemotherapy. (See 'Total neoadjuvant therapy for locally advanced tumors' below.)

Total neoadjuvant therapy for locally advanced tumors

Indications — For most patients with locally advanced rectal cancer who are at high risk for a margin-positive resection (ie, T4 disease or an involved mesorectal fascia, extramural venous invasion), as well as for those with clearly node-positive disease and a low-lying rectal tumor, we suggest total neoadjuvant therapy (TNT; ie, oxaliplatin-based chemotherapy combined with long-course CRT or short-course RT) rather than long-course CRT or short-course RT alone. TNT is associated with increased compliance with chemotherapy (because of the greater tolerability in the preoperative as compared with the postoperative setting), improved local control, and the ability to consider nonoperative treatment if the patient declines surgery. (See 'Avoidance of radical surgery' below.)

We generally do not utilize this strategy in patients with lower-risk locally advanced cancers (ie, early cT3N0 disease without a threatened mesorectal fascia, especially involving the upper rectum, distal cT1-2N0 tumors) because these patients may not need chemotherapy at all. (See 'Choice of strategy' below and 'Nonoperative management (watch and wait)' below.)

For cT3N0 disease that is low lying and would require either an APR or a very low coloanal anastomosis, the decision to pursue TNT for the primary goal of organ preservation is a complex decision that requires careful explanations of the pros and cons with the patient. It cannot be recommended as the standard of care at this time until datasets become more mature.

If TNT is chosen, and chemotherapy is initiated first, we recommend that clinicians proceed cautiously with scans after two months of chemotherapy and move directly to CRT if no response is seen. This is particularly important in the setting of a tumor with deficient mismatch repair/high levels of microsatellite instability, a substantial proportion of which may be resistant to neoadjuvant chemotherapy [144]. During the chemotherapy portion of treatment, we suggest use of an oxaliplatin-based chemotherapy regimen rather than a fluoropyrimidine alone. Options include FOLFOX of FOLFIRINOX, as was used in the PRODIGE 23 study. We would not administer more than four months of preoperative oxaliplatin-based therapy.

Rationale and benefit — The rationale for total neoadjuvant therapy is to intensify the neoadjuvant therapy through the addition of chemotherapy to CRT, leading to higher rates of resectability and pCR [145-147]. However, whether the higher pCR rates are attributed to the chemotherapy itself or to increasing the interval from RT to surgery (which achieves greater pathologic downstaging) is uncertain. Data from the OPRA trial suggest that increasing the interval from RT to surgery may be the more important mechanism of benefit for TNT, at least when long-course CRT is utilized [148]. (See 'Nonoperative management (watch and wait)' below.)

Other benefits include addressing potential systemic disease early and more effectively compared with postoperative administration of chemotherapy, improving compliance with systemic therapy improving the fraction of patients who actually complete chemotherapy, and increasing the likelihood of organ preservation. One of the potential hazards of premature incorporation of TNT into treatment paradigms before randomized trials have fully characterized the benefits and harms is the risk of overtreatment [149].

The use of TNT rests on the assumption that its theoretical benefits translate into improved outcomes. However, an important point is that no significant improvement in overall survival has been shown for TNT relative to standard preoperative RT or CRT, followed by surgery and adjuvant chemotherapy in randomized trials. The results of meta-analyses are discordant:

A meta-analysis of seven trials comparing TNT using long-course CRT plus induction chemotherapy versus neoadjuvant CRT followed by adjuvant chemotherapy (2416 individual patients, including two randomized phase III [150,151], one randomized phase II trial [145], one subset analysis from a single arm phase II trial [152], and the remainder retrospective analyses) concluded that TNT approximately doubled the pathologic CR rate (30 versus 15 percent), and in the three studies with available data, DFS was two-fold higher [153]. There was no significant difference in the likelihood of sphincter preserving surgery (OR 1.06, 95% CI 0.73-1.54).

In a second meta-analysis of eight phase II/III trials involving 2196 patients comparing TNT versus standard neoadjuvant CRT for locally advanced rectal cancer, including an early analysis of the STELLAR trial (see 'Choice of strategy' below), TNT using either short-course RT or long-course CRT was associated with higher pCR rates (OR 1.77, 95% CI 1.28-2.45), longer median DFS (HR 0.83, 95% CI 0.72-0.96), and a reduced risk of distant metastases (HR 0.81, 95% CI 0.68-0.95), but the difference in overall survival was not statistically significant (HR 0.88, 95% CI 0.74-1.05) [154].

On the other hand, a third analysis of 15 studies (including seven randomized trials, only two phase III [95,155]) concluded that the benefits of TNT included higher complete pathologic response rates (22.3 versus 14.2 percent, pooled OR 1.51, 95% CI 1.29-1.78), significantly lower rates of distant metastases (OR 0.78, 95% CI 0.63-0.96), and better rates of three-years DFS (70.6 versus 65.3 percent, pooled RR 0.86, 95% CI 0.77-0.96) and overall survival (84.9 versus 82.3, pooled RR 0.83, 95% CI 0.71-0.97) [156].

Despite these issues, updated guidelines from the NCCN [41] include TNT among the several options for patients with T3 tumors with an involved CRM, T4 tumors, or N1/2 disease, or for locally unresectable or medically inoperable patients with rectal cancer.

Choice of strategy — TNT can be accomplished using either long-course CRT (our preferred strategy) or short-course RT, both in conjunction with additional systemic oxaliplatin-based chemotherapy.

Long-course CRT – There has been increasing interest in TNT using long-course CRT for patients with locally advanced rectal cancer, but few randomized trials are available [110,146,150,157-166]. The following studies support this approach:

The benefit of TNT relative to initial CRT followed by surgery and then postoperative chemotherapy was addressed in the randomized phase III PRODIGE 23 trial suggests that a more intensive approach to induction chemotherapy prior to long-course CRT may provide better results [167]. In this trial, 461 patients with clinical T3 or T4 rectal cancer <15 cm from the anal verge were randomly assigned to TNT using three months of modified FOLFIRINOX (oxaliplatin 85 mg/m2, leucovorin 400 mg/m2, irinotecan 180 mg/m2 day 1, and FU 2400 mg/m2 over 46 hours every 14 days) followed by long-course CRT (50 Gy in 25 fractions plus concurrent capecitabine), TME, and three additional months of chemotherapy (FOLFOX or capecitabine, choice of the treating center), or the control arm, which consisted of the same long-course CRT followed by surgery and six months of adjuvant chemotherapy, either FOLFOX or capecitabine. Neoadjuvant FOLFIRINOX was well tolerated, and 92 percent completed all courses. By contrast, of the patients assigned to preoperative CRT, only 79 percent received any adjuvant chemotherapy, and only 81 percent of these completed all six months of therapy.

Neoadjuvant chemotherapy did not hamper compliance with CRT or postoperative chemotherapy, or increase surgical morbidity, and it more than doubled the complete pathologic response rate (28 versus 12 percent). At a median follow-up of 46.5 months, three-year DFS, the primary endpoint, was significantly better with TNT (76 versus 69 percent, HR 0.69, 95% CI 0.49-0.97). Three-year overall survival rates were not significantly better with neoadjuvant FOLFIRINOX (91 versus 88 percent, stratified HR 0.65, 95% CI 0.40-1.05). Patients who received the bulk of their oxaliplatin preoperatively also seemed to fare better from the standpoint of neurotoxicity.

During the three months of neoadjuvant FOLFIRINOX, grade 1 or 2 peripheral sensory neuropathy developed in 83 percent, but only 2 percent had grade 3 symptoms; during the adjuvant portion of therapy, a similar fraction of patients had grade 1 or 2 peripheral sensory neuropathy (64 percent of each group) but grade 3 peripheral sensory neuropathy rates were lower in the neoadjuvant FOLFIRINOX group (12 versus 21 percent), despite their similar cumulative exposure to oxaliplatin.

Further support for TNT using long-course CRT is provided by a retrospective cohort analysis using records from Memorial Sloan Kettering Cancer Center (MSKCC); of the 811 patients who presented with T3/4 or node-positive rectal cancer between 2009 and 2015, 320 received CRT with planned adjuvant chemotherapy, while 308 had TNT using induction oxaliplatin-based treatment followed by CRT [146]. Overall, patients receiving TNT received a greater percentage of the planned chemotherapy than did those who received postoperative therapy. For the patients undergoing surgery, the pCR rate with TNT was not higher than with neoadjuvant CRT and planned adjuvant chemotherapy (18 versus 17 percent). However, the "complete response rate" (which included both the pCRs in those who underwent surgery and the sustained clinical complete response [cCR] rate, for at least 12 months post-treatment, in those who did not undergo surgery) was higher with TNT (36 versus 21 percent). Nonoperative treatment increased during the study period, and rates were higher in patients who received TNT (73 of 308 [27 percent] versus 24 of 320 [7.5 percent]). Long-term outcomes were not reported.

A key issue raised by this study is whether TNT should be used to enhance the likelihood of organ preservation in disease settings in which there is an ambiguous or absent survival benefit for systemic chemotherapy (eg, distal T1-3N0 tumors without a threatened mesorectal fascia). Although TNT enhanced rates of organ preservation at one year, chemotherapy alone is associated with a mortality risk, and it is possible that the addition of an oxaliplatin-based chemotherapy regimen to upfront CRT may incur long-lasting neuropathy without impacting cancer control. This issue is addressed in more detail below. (See 'Nonoperative management (watch and wait)' below.)

Short-course RT – At least three trials have addressed the benefit of TNT using short-course RT versus long-course CRT plus postoperative chemotherapy; no trial has directly compared the two approaches with TNT (ie, short-course RT versus long-course CRT, both in conjunction with neoadjuvant chemotherapy):

The Polish II trial compared short-course RT followed by chemotherapy (three months of FOLFOX) prior to surgery versus long-course CRT utilizing oxaliplatin plus bolus FU and LV in 541 patients with cT4 or fixed cT3 tumors [157]. Preoperative toxicity rates were lower with the short-course treatment, while complete resection rates (77 versus 71 percent) and pCR rates (16 versus 12 percent) were not significantly better. In the latest analysis, at a median follow-up of seven years, eight-year overall survival was identical (49 percent), and there were no differences in the cumulative incidence of local or distant failure or rates of late complications [131].

The control arm in this study, oxaliplatin-containing CRT, is generally not considered a standard approach for long-course CRT due to the greater treatment-related toxicity and no better outcomes when directly compared with standard fluoropyrimidine-based CRT. (See 'Oxaliplatin' above.)

Preoperative short-course RT followed by 18 weeks of oxaliplatin-containing chemotherapy was directly compared with conventional long-course capecitabine-based CRT followed by postoperative adjuvant oxaliplatin-based chemotherapy in the international randomized RAPIDO trial, which enrolled 920 patients with either cT4a/b or cN2 stage, extramural vascular invasion, involved mesorectal fascia, or enlarged lateral lymph nodes [151,155]. pCR rates were higher in the short-course TNT arm (28 versus 14 percent), and there were significant benefits in terms of three-year cumulative probability of disease-related treatment failure (24 versus 30 percent, HR 0.76, 95% CI 0.60-0.96), and three-year rates of distant metastases (20 versus 27 percent, HR 0.69, 95% CI 0.54-0.90). Locoregional failure rates at three years were comparable (8 versus 6 percent), and serious treatment-related adverse events occurred in a similar proportion (38 versus 34 percent), both at early and late time points [168]. A major flaw of this study is that postoperative chemotherapy was not mandated, and fewer than 50 percent of patients initiated it.

In the STELLAR trial, 599 patients with cT3-4 or node-positive rectal cancer were randomly assigned to short-course RT (25 Gy in five fractions over one week) followed by four cycles of capecitabine plus oxaliplatin or conventional fractionation CRT (50 Gy in 25 fractions over five weeks) concurrent with capecitabine, followed by surgery and additional chemotherapy (two cycles of CAPOX for the short-course group, and six cycles on the CRT group) [169]. Patients treated with TNT had a two-fold higher rate of acute grade 3 or 4 toxicities during preoperative treatment (26.5 versus 12.6 percent). At a median follow-up of 35 months, three-year DFS was not superior with TNT using short-course RT (64.5 versus 62.3 percent, HR 0.883) and there were no differences in metastasis-free survival or locoregional recurrence, but the TNT group had an inexplicably higher three-year overall survival (86.5 versus 75.12 percent, p = 0.033).

Sequencing of RT and chemotherapy — The optimal way to sequence chemotherapy and RT when using a TNT approach is not established. For most patients, we suggest initial long-course fluoropyrimidine-based CRT or short-course RT, followed by 12 to 16 weeks of systemic oxaliplatin-based chemotherapy prior to surgery, especially if organ preservation is an important goal. However, for patients in whom metastatic disease is strongly suspected, but cannot be confirmed, we prefer to start with 12 to 16 weeks of initial systemic chemotherapy followed by radiologic reevaluation; if metastatic disease is deemed unlikely, either long-course CRT or short-course RT can then be administered prior to surgery. Although there are no prospective data directly addressing the optimal duration of induction chemotherapy, if the TNT approach is used, we suggest limiting neoadjuvant chemotherapy to four months and eliminating postoperative chemotherapy.

Few data are available addressing the optimal sequence of neoadjuvant RT and chemotherapy when TNT is chosen. The following studies inform this issue:

Initial CRT followed by consolidation chemotherapy and then surgery was directly compared with initial (induction) chemotherapy followed by CRT and then surgery in the randomized phase II German CAO/ARO/AIO-12 trial of 311 patients withT3 to T4 and/or node-positive rectal cancer [166]. Upfront CRT followed by chemotherapy resulted in better compliance with CRT but worse compliance with chemotherapy. This approach did not significantly increase the fraction of patients who were able to undergo a complete resection (90 versus 92 percent), the pCR rate (25 versus 17 percent), the fraction of patients who underwent sphincter-preserving surgery (72 versus 68 percent), or the fraction with a positive circumferential resection margin (7 versus 10 percent) [166]. Notably, the group receiving CRT first had a significantly longer median time between the end of CRT and surgery (90 versus 45 days), which might have influenced the pCR rate. In the latest analysis, with median follow-up 43 months, despite the higher pCR rate in the CRT first group, this did not translate into a benefit in terms of three-year DFS (73 percent in both groups), three-year cumulative incidence of locoregional recurrence (6 versus 5 percent) or rate of distant metastases (18 versus 16 percent) [170].

This issue was also addressed in the Organ Preservation of Rectal Adenocarcinoma (OPRA) trial, in which 324 patients with MRI stage II or III rectal cancer were randomly assigned to four months of oxaliplatin-based chemotherapy before (induction) or after (consolidation) long-course fluoropyrimidine-based CRT [148]. Patients were restaged 8 to 12 weeks after TNT, and those with a complete or near-complete response were offered watchful waiting; those with an incomplete response had TME. At a median follow-up of three years, compliance with systemic chemotherapy was similar in both arms (81 and 82 percent) as was three-year DFS (76 and 75 percent). However, patients treated with consolidation rather than induction chemotherapy had significant higher rates of organ preservation (TME-free survival, 53 versus 41 percent). Patients who underwent TME after restaging had similar three-year DFS as those who underwent TME after regrowth. (See 'Nonoperative management (watch and wait)' below.)

An important point is that this trial utilized the FOLFOX and CAPOX chemotherapy regimens. As more institutions are adopting FOLFIRINOX as the neoadjuvant chemotherapy regimen for TNT (based on the PRODIGE 23 trial (see 'Choice of strategy' above)), it is not clear that FOLFIRINOX will be well-tolerated if administered after rather than before RT, as was done in the PRODIGE 23 trial. The safety of this approach has not been tested.

The use of initial chemotherapy as a strategy to permit more selective use of RT prior to surgery is discussed below. (See 'Selective use of RT' below.)

Alternative strategies

Avoidance of radical surgery

Nonoperative management (watch and wait) — Despite the lack of randomized trials, nonoperative management is becoming an acceptable alternative for those patients who experience a clinical complete response (cCR) to neoadjuvant therapy, especially to TNT. It should be emphasized that mature data are not yet available to ensure that survival outcomes are equivalent to conventional operative management, and this is particularly important given the natural history of rectal cancer and the rate of late recurrences between 5 and 10 years after resection. there is not uniform agreement on whether nonoperative approaches should be considered standard at this time, even among the authors and editors associated with this topic review [171].

As examples:

Year 2020 consensus-based guidelines from the NCCN state that for patients who achieve a cCR with no evidence of residual tumor on digital rectal examination (DRE), rectal MRI, and direct endoscopic evaluation, an initial nonoperative approach may be considered with an experienced multidisciplinary team [41].

Year 2020 guidelines from the American Society of Colon and Rectal Surgeons state that patients with an apparent clinical complete response to neoadjuvant therapy should typically be offered radical resection [172], although a "watch and wait" management approach could be considered for highly selected patients in the context of a protocolized setting.

Modern conventional fractionation concomitant CRT protocols with and without neoadjuvant chemotherapy, and short-course RT approaches combined with induction chemotherapy can achieve a pCR (ie, no tumor in the surgical specimen) in 16 to 27 percent of rectal cancers [50,157,162,173-175].

Most retrospective series registry reports and systematic reviews report excellent outcomes in patients who achieve a pCR after preoperative therapy [174,176]. These favorable results have led some to question whether selected patients with radiologic and clinical evidence of a complete response after neoadjuvant therapy might be able to avoid surgery altogether

No randomized trials are available in which patients with a cCR to neoadjuvant therapy were assigned to surgery or no surgery. However, data on the long-term outcomes of nonsurgically treated patients are available from accumulating reports [148,173,177-189], the most important of which are described in detail below:

The strategy of using TNT for organ preservation was addressed in the OPRA trial, described above, in which 324 patients with MRI stage II or III rectal cancer were randomly assigned to four months of oxaliplatin-based chemotherapy before (induction) or after (consolidation) long-course fluoropyrimidine-based CRT [148]. (See 'Sequencing of RT and chemotherapy' above.)

Patients were restaged 8 to 12 weeks after TNT with DRE, flexible sigmoidoscopy and MRI, and those with a complete or near-complete response were offered watchful waiting; those with an incomplete response had TME. At a median follow-up of three years, patients treated with consolidation rather than induction chemotherapy had significant higher rates of organ preservation (TME-free survival, 53 versus 41 percent).

The safety of a watch and wait approach for locally advanced rectal cancer after a cCR following neoadjuvant CRT was addressed in a systematic review and meta-analysis of 23 studies (all retrospective or prospective cohorts, no randomized trials) totaling 867 patients [186]. Overall, 10 of the 23 studies provided sufficient data to calculate a pooled two-year local regrowth rate with the watch and wait approach (15.7 percent, 95% CI 11.8-20.1). Following local regrowth, the pooled proportion of patients who had salvage therapy was 95.4 percent; sphincter preservation was achieved in 49.8 percent of patients undergoing salvage surgery. Longer intervals between neoadjuvant CRT and assessment of clinical response were associated with lower rates of local regrowth.

In the eight studies comparing patients managed with a watch and wait strategy with patients who underwent radical surgery with a cCR or pCR, there were no significant differences in distant recurrences, cancer-specific mortality, or overall survival. However, the patients managed with a watch and wait approach did have poorer DFS than those who underwent radical surgery, driven by the intraluminal local regrowth. In the five studies comparing patients with a cCR managed with a watch and wait approach with those who had radical surgery and were found to have a pCR, the two-year local regrowth rates ranged from 4.8 to 21 percent with watch and wait (versus a 0 to 7.7 percent two-year rate of local recurrence after radical surgery). In the three studies comparing patients with a cCR managed with a watch and wait approach versus radical surgery, the two-year local regrowth rate ranged from 3.3 to 30.4 percent with watch and wait (versus a 0 to 2.2 percent local recurrence rate in patients with a cCR who had radical surgery).

Data on nonoperative management in patients with a cCR following neoadjuvant therapy for rectal cancer are also available from the International Watch and Wait Database (IWWD), an international multicenter registry [187]. Of the 1009 patients submitted to the registry between 2015 and 2017 who received neoadjuvant treatment and were managed with a watch and wait approach rather than radical resection (which included 192 patients treated in Brazil and 149 who were enrolled in the OnCoRe database, described above), there were 880 with a cCR. CRT was the most commonly used neoadjuvant strategy. At a median follow-up of 3.3 years, the two-year cumulative incidence of local regrowth was 25.2 percent, 88 percent of all local regrowth was diagnosed in the first two years, and 97 percent was located in the bowel wall (3 percent in regional nodes only). Details of surgical treatment were only available for 148 of the 213 patients with local regrowth. In total, 115 (78 percent) of the 148 patients with a local recurrence needed a TME resection for local regrowth; the remainder had local excision. Distant metastases developed in 71 patients (8 percent) during follow-up. Among the patients with local regrowth, the proportion of distant metastases was higher (38 of 213, 18 percent). Five-year DFS for the whole group was 94 percent, and five-year overall survival was 85 percent.

Not all reports are favorable, however [188,190]. A retrospective case series compared the outcomes of 113 patients who achieved a cCR after completing neoadjuvant therapy and agreed to a watch and wait strategy versus those of 136 patients who had a pCR after neoadjuvant therapy and mesorectal excision [188]. All 22 regrowths in the watch and wait group were detected on routine surveillance and treated with salvage surgery, and rectal preservation was achieved in 93 of the 113 patients. However, patients in the watch and wait group who had a local recurrence had a higher rate of distant metastases than those whose tumors did not recur (36 versus 1 percent), and survival was inferior in the watch and wait group compared with the surgically treated patients (DFS 90 versus 98 percent).

Taken together, most of the data suggest that a careful endoscopic, clinical, and radiographic evaluation might be able to identify patients who have a complete clinical response to upfront therapy and who have a good likelihood of local tumor control and may not need surgery. However, none of these data are from trials that randomly assigned patients with a cCR to surgery or no surgery, and they all suffer from significant limitations.

Globally, several additional prospective randomized trials of nonoperative therapy in complete responders to induction therapy are underway (including NCT02514278, NCT02794520, NCT01047969, and NCT03426397) that use accurate pelvic MRI staging at baseline and predefined criteria for a cCR, which should resolve lingering concerns about long-term outcomes by comparing the intervention of avoiding surgery in patients identified as having a cCR. Eligible patients should be encouraged to enroll.

Local excision in lieu of total mesorectal excision after chemoradiotherapy — Local excision is an accepted surgical option for cT1 rectal adenocarcinomas. However, local excision as the sole surgical treatment for ≥cT2N0 rectal cancer after CRT is still considered investigational because of concerns for micrometastases in the mesorectum and residual disease in the excision bed, both of which could contribute to higher rates of recurrence and a poorer outcome compared with definitive surgery. (See "Surgical treatment of rectal cancer", section on 'Local excision'.)

There has been increasing interest in the use of transanal endoscopic surgery (TES) as a mechanism for evaluating pathologic response to neoadjuvant CRT in patients with cT1-3N0 rectal cancer, with further surgical treatment dictated by the pathologic findings on the local excision specimen. There are conflicting data from systematic reviews as to whether or not outcomes (local recurrence, survival) are inferior in patients who undergo a subsequent TME after a local excision has been performed; the available data are probably insufficient to answer this question [191,192]. Furthermore, others have shown that as many as one-fifth of patients with ypT1 to ypT2 tumors after CRT are ypN+, and that local excision would result in untreated metastatic disease in these patients [193]. This subject is discussed in detail elsewhere. (See "Transanal endoscopic surgery (TES)", section on 'T1-3N0 rectal cancer following neoadjuvant therapy'.)

For all of these reasons, we do not support TES as the sole surgical treatment in patients with ≥cT2N0 disease who undergo local excision and have any residual tumor after CRT. Furthermore, in patients with ≥cT2N0 tumors who achieved a pCR with preoperative CRT, prospective studies are needed to demonstrate that long-term outcomes with local excision are comparable with those attained with conventional surgery. In our view, local excision should only be used after CRT in patients who are frail or otherwise unfit for abdominal surgery or in the setting of a clinical trial. This subject is discussed in detail elsewhere. (See "Transanal endoscopic surgery (TES)", section on 'T1-3N0 rectal cancer following neoadjuvant therapy'.)

Tumor response assessment and follow-up — Consistent with consensus-based guidelines from the NCCN [41], patients who achieve a cCR after neoadjuvant therapy with no evidence of residual tumor on DRE, rectal MRI, and direct endoscopic evaluation may be considered for an initial nonoperative approach with an experienced multidisciplinary team. There is no standard classification of response based upon DRE, endoscopy, and MRI; one approach, which is based on the schema used at MSKCC, is outlined in the table (table 7) [194]. We would consider that only patients who are classified as a cCR are eligible for nonoperative management. For most patients, we would repeat these studies every three months for two years, and then every six months to complete five years of surveillance.

A major obstacle to selecting patients who may avoid radical surgery is that pCR can most accurately be determined after a full pathologic examination of the resected specimen. There is no single test that is capable of identifying patients with complete tumor regression following neoadjuvant therapy. A combination of clinical and endoscopic findings does not provide sufficient information to allow identification of patients with a sustained cCR who are likely to have a pCR. The limitations of this approach can be illustrated by a study from the MSKCC in which all patients undergoing preoperative CRT were subjected to resection; 75 percent of the complete clinical responders had persistent foci of tumor that were not detectable on preoperative DRE or proctoscopy [195].

In our view, the decision to pursue or not to pursue surgery should not be based solely on the absence of clinically palpable or visible tumor after neoadjuvant treatment. A major problem is the risk of lymph node metastases despite complete primary tumor regression. Even in ypT0 rectal cancers treated with TME, the risk of lymph node metastases or mesorectal deposits is as high as 12 percent [196-198]. (See 'Prognosis and extent of tumor regression' above.)

The role of imaging for restaging for assessment of the primary tumor and regional nodes after neoadjuvant therapy has been the subject of several studies, and all suggest that neither MRI, computed tomography (CT), transrectal endoscopic ultrasound (EUS), or integrated positron emission tomography (PET)/CT alone is sufficiently accurate to identify the true complete responders [199-210]. A major problem with MRI is that it is difficult to differentiate small areas of residual tumor from fibrosis, and readers tend to overestimate the presence of tumor [206,211-213]. PET/CT findings that suggest a cCR are also associated with a low positive predictive value for a pCR (39 percent in one systematic review [214]).

Nevertheless, MRI has been increasingly used in addition to DRE and endoscopy for this assessment. When MRI is used, as part of the evaluation, the TRG is a critical aspect of the evaluation (table 8) [199,215,216]. According to this widely used 5-tiered schema, only patients with a TRG1 or 2 radiographic response on MRI would be eligible for a nonoperative approach. Alternative MRI TRG schema have been proposed [217].

Selective use of RT — In our view, outside of the context of a clinical trial, the use of preoperative chemotherapy alone with selective use of RT for poor responders remains investigational, unless patients refuse or are ineligible for pelvic RT (eg, if they received prior pelvic radiation for a different malignancy). In such a situation, if four months of neoadjuvant chemotherapy was administered prior to surgery, we would omit postoperative chemotherapy.

Given the increasing recognition of the long-term late adverse effects of RT (particularly with regard to intestinal and sexual function), the sensitivity of rectal cancer to modern systemic chemotherapy, the more favorable compliance and toxicity profile with preoperative as compared with postoperative chemotherapy seen in early studies [218], and the lower risk of local recurrence in patients who undergo a good-quality TME, the use of upfront chemotherapy is under active study as a means of permitting more selective use of RT. The use of initial chemotherapy prior to CRT in patients with T4, bulky, or N2 tumors is discussed above. (See 'Total neoadjuvant therapy for locally advanced tumors' above.)

For patients with nonbulky, potentially resectable tumors, the available data suggest that initial chemotherapy might permit more selective use of RT [95,219]. The best data are from the randomized phase III FOWARC trial, in which 495 patients with clinical stage II or III rectal cancer within 12 cm of the anal verge were randomly assigned to preoperative RT with concurrent FU (control), preoperative RT with concurrent modified FOLFOX6, or modified FOLFOX6 alone [95]. The group receiving chemotherapy alone had the lowest pCR rate (7 versus 28 percent for FOLFOX plus RT and 14 percent for FU plus RT). However, local recurrence rates were similar (8 versus 7 and 8 percent, respectively), as were three-year DFS (77 percent with FOLFOX versus 74 percent with FOLFOX plus RT and 73 percent with FU plus RT) and overall survival (91 percent with FOLFOX versus 89 percent with FOLFOX plus RT and 91 percent with FU plus RT). The authors concluded that modified FOLFOX6, with or without RT, did not significantly improve outcomes as compared with RT with FU alone.

These promising results have led to trials of neoadjuvant FOLFOX, or more aggressive chemotherapy regimens [220], with selective use of CRT versus preoperative CRT for locally advanced rectal cancer based on response to initial chemotherapy. One phase III trial (the PROSPECT or N1048 trial) has completed enrollment, and results are expected in 2022.

It should be noted that not all reports on this approach are favorable [221]. In our view, outside of the context of a clinical trial, this approach remains investigational, unless patients refuse or are ineligible for pelvic RT. In such a situation, if four months of neoadjuvant chemotherapy was administered prior to surgery, we would omit postoperative chemotherapy.

Neoadjuvant immunotherapy for dMMR tumors — Upfront immunotherapy followed by nonoperative management for clinical complete responders cannot yet be considered a standard approach for rectal cancers with deficient mismatch repair (dMMR). However, individuals with dMMR tumors who place a high value on avoiding the long-term morbidity associated with surgery and pelvic RT might choose this non-standard approach if they are willing to accept the uncertainty of long-term outcomes, the small risk of fatal side effects from checkpoint inhibitors, and the need for very close follow-up (every four month pelvic MRI, digital rectal examination, sigmoidoscopy with biopsy).

A small subset of rectal tumors (approximately 3 percent [222]) are dMMR, the biologic footprint of which is high levels of microsatellite instability (MSI-H). The presence of dMMR/MSI-H indicates potential responsiveness to immunotherapy using immune checkpoint inhibitors in the setting of metastatic disease. (See "Systemic therapy for nonoperable metastatic colorectal cancer: Selecting the initial therapeutic approach", section on 'Patients with deficient DNA mismatch repair/microsatellite unstable tumors' and "Systemic therapy for nonoperable metastatic colorectal cancer: Approach to later lines of systemic therapy", section on 'Microsatellite unstable/deficient mismatch repair tumors'.)

Colorectal cancer with dMMR/MSI-H has a favorable prognosis overall, but in the setting of locally advanced rectal cancer, some have noted a diminished response to conventional neoadjuvant CRT approaches [144], although this is not a universal finding [223]. On the other hand, high levels of pathologic complete or near complete response have been observed following neoadjuvant immunotherapy [224-229], and several trials are now exploring this option, including in the setting of locally advanced rectal cancer [230,231].

Early data are available from a small prospective trial, in which 12 patients with dMMR stage II or III (94 percent) rectal cancer first received single-agent therapy with the anti PD-1 monoclonal antibody dostarlimab (500 mg every three weeks) for six months, which was to be followed by standard CRT and surgery; patients who had a complete clinical response after dostarlimab (defined as the absence of residual disease on digital and endoscopic rectal examination as well as the absence of residual disease on rectal MRI with no restricted diffusion on T2-weighted imaging), could proceed without CRT and surgery [231]. In an early report, in which all 12 patients had at least six months of follow-up after completion of dostarlimab (median follow-up 12 months after study enrollment, range 6 to 25 months), all 12 had a complete clinical response with no evidence of residual or recurrent tumor on MRI, fluorodeoxyglucose-PET, endoscopic or DRE, or biopsy. No patient has required chemotherapy, radiation, or surgery. Treatment was well-tolerated with no ≥grade 3 adverse effects. Long-term oncologic outcomes were not reported.

Adjuvant therapy — Indications for and choice of adjuvant chemotherapy following resection in patients who have received neoadjuvant CRT or RT are addressed in detail elsewhere. (See "Adjuvant therapy after neoadjuvant therapy for rectal cancer".)

In general, adjuvant chemotherapy is not pursued for patients who received neoadjuvant therapy that included four months of chemotherapy (ie, the TNT approach). (See 'Total neoadjuvant therapy for locally advanced tumors' above.)

LOCAL TREATMENT FOR PATIENTS WITH DISTANT METASTASES

Potentially resectable metastases — There are no established guidelines for pelvic irradiation in patients with synchronous, potentially resectable metastases (predominantly in liver), most of whom will begin treatment with systemic chemotherapy. A few studies have sought to elucidate the role of local treatments, such as RT, in this setting; however, given the small patient cohorts and variable use of chemotherapy, results are not conclusive and treatment must be individualized. For potentially operable patients who still have operable disease after initial systemic chemotherapy, timing of resection of the primary and liver metastases (simultaneous versus staged) is discussed in detail elsewhere. (See "Hepatic resection for colorectal cancer liver metastasis", section on 'Synchronous colorectal liver metastases'.)

The contribution of pelvic RT to outcomes (especially survival) in patients with synchronous, potentially resectable liver metastases is unclear. Consensus-based guidelines from the National Comprehensive Cancer Network (NCCN) [41] suggest that one of the following strategies is acceptable: initial chemotherapy followed by short-course RT or long-course CRT and then followed by resection (synchronous or staged), or initial CRT or short-course RT followed by chemotherapy and then resection. In this setting, we prefer short-course RT rather than long-course CRT; this approach is also supported by European Society for Medical Oncology (ESMO) guidelines [2].

There have been multiple approaches to patients with synchronous, potentially resectable metastases, some of which include RT and some of which do not [232-236]. Information regarding the natural history and patterns of recurrence in patients who undergo complete resection of both synchronous rectal cancer and liver metastases has not been well documented. The following data are available:

In a retrospective evaluation of 89 patients presenting with synchronous liver metastases and rectal cancer, 30 percent of whom received perioperative RT, 25 patients (28 percent) developed a pelvic recurrence; the difference between those who did and those who did not receive pelvic RT was potentially clinically meaningful, although it was not statistically significant (15 versus 34 percent, p = 0.066) [233]. However, there was little influence of pelvic RT on overall survival. In the subgroup of 56 patients who underwent local treatment for the liver metastases, the two-year overall survival rates were 80 and 74 percent in patients who did and who did not receive pelvic RT (p = 0.616).

A second retrospective analysis included 185 patients who underwent complete resection of a rectal primary and synchronous liver metastases at a single institution over an 18-year period [234]. Chemotherapy was administered to 180 patients (97 percent), while pelvic RT was given to 91 patients (49 percent) either before (n = 65) or after (n = 26) rectal resection. At a median follow-up of 44 months, 130 patients (70 percent) had a disease recurrence, and these recurrences usually involved distant sites (liver, lung), rather than locoregional recurrences at the primary tumor site, even in patients treated without pelvic RT. Overall, pelvic recurrence was an uncommon event and was observed as an isolated episode in <5 percent of patients. Patients who did or did not receive RT had similar rates of recurrence (63 versus 67 percent), pelvic recurrence (11 versus 16 percent), and isolated pelvic recurrence (9 versus 8 percent). In addition, use of RT did not improve disease-specific survival.

In this setting, the influence of chemotherapy and RT on overall survival is likely small relative to the survival benefit of surgically resecting all gross disease, and in our view, efforts should focus on achieving margin-negative resections of both the primary site and metastases above all else. Neoadjuvant chemotherapy and RT may enhance the possibility of a complete resection. Even when a complete resection is clearly possible, prevention of recurrence through the addition of chemotherapy and/or RT, delivered either preoperatively or postoperatively, remains a worthy goal, as locoregional recurrence can be extremely morbid. Future studies are needed to sort out the relative benefits of both RT and chemotherapy in resectable stage IV disease.

Synchronous unresectable metastases — For patients who present with synchronous, unresectable stage IV disease and a symptomatic rectal primary tumor, we suggest short-course RT in conjunction with modern combination systemic chemotherapy. This approach may allow patients to avoid surgery, even those with a nearly obstructing lesion. By contrast, a benefit for short-course RT or long-course CRT for asymptomatic patients with unresectable metastatic disease is not established, and we would generally not pursue RT for most of these patients, focusing instead on palliative systemic chemotherapy. This approach is consistent with consensus-based guidelines from the NCCN [41].

For patients with a symptomatic rectal primary tumor and synchronous, unresectable metastatic disease, creation of a diverting stoma or palliative resection is often carried out before initiation of systemic chemotherapy to obtain fast relief from symptoms and to avoid complete bowel obstruction or perforation necessitating emergency surgery. Another nonsurgical option is placement of a stent (if feasible). (See "Locoregional methods for management and palliation in patients who present with stage IV colorectal cancer", section on 'Incurable metastatic disease' and "Locoregional methods for management and palliation in patients who present with stage IV colorectal cancer", section on 'Nonsurgical palliative options'.)

If placement of a stent is not feasible, RT with modern combination systemic chemotherapy may allow selected patients to avoid surgery, even those with a nearly obstructing lesion [237-239]. This issue was addressed in a phase II study in which 40 patients with symptomatic primary rectal cancer and synchronous unresectable metastases received short-course RT (5 x 5 Gy) and oxaliplatin-based systemic chemotherapy; median survival was 11.5 months, and only eight patients (20 percent) required surgery during the course of their disease [238]. Pelvic symptoms were completely resolved in 30 percent, and another 35 percent had significant improvement.

Resource-stratified guidelines — The approach outlined above assumes that health care resources are not a limiting factor. Resource-stratified guidelines for treatment of patients with late-stage colorectal cancer are available from the American Society of Clinical Oncology [240]. They include specific recommendations for initial diagnostic evaluation, systemic therapy, surgical management of patients with potentially resectable metastatic disease, and issues specific to primary site radiation therapy in patients with metastatic disease.

POST-TREATMENT SURVEILLANCE AND SURVIVOR ISSUES — Recommendations for post-treatment cancer surveillance and issues that arise in long-term survivors of rectal cancer (genitourinary problems, bowel and anorectal dysfunction) are discussed in detail elsewhere. (See "Post-treatment surveillance after colorectal cancer treatment" and "Approach to the long-term survivor of colorectal cancer".)

SPECIAL CONSIDERATIONS DURING THE COVID-19 PANDEMIC — The COVID-19 pandemic has increased the complexity of cancer care. Important issues include balancing the risk from delaying cancer treatment versus harm from COVID-19, minimizing the number of clinic and hospital visits to reduce exposure whenever possible, mitigating the negative impacts of social distancing on delivery of care, and appropriately and fairly allocating limited health care resources. Specific considerations for patients with locally advanced rectal cancer include substituting neoadjuvant short-course radiation therapy for long-course CRT to minimize the number of clinic visits, and using total neoadjuvant therapy as a means of delaying definitive surgery until after the peak of the pandemic, if appropriate. (See 'Short-course radiotherapy' above and 'Total neoadjuvant therapy for locally advanced tumors' above.)

These and other recommendations for cancer care during active phases of the COVID-19 pandemic are discussed separately. (See "COVID-19: Considerations in patients with cancer".)

SOCIETY GUIDELINE LINKS — Links to society and government-sponsored guidelines from selected countries and regions around the world are provided separately. (See "Society guideline links: Colorectal cancer".)

INFORMATION FOR PATIENTS — UpToDate offers two types of patient education materials, "The Basics" and "Beyond the Basics." The Basics patient education pieces are written in plain language, at the 5th to 6th grade reading level, and they answer the four or five key questions a patient might have about a given condition. These articles are best for patients who want a general overview and who prefer short, easy-to-read materials. Beyond the Basics patient education pieces are longer, more sophisticated, and more detailed. These articles are written at the 10th to 12th grade reading level and are best for patients who want in-depth information and are comfortable with some medical jargon.

Here are the patient education articles that are relevant to this topic. We encourage you to print or e-mail these topics to your patients. (You can also locate patient education articles on a variety of subjects by searching on "patient info" and the keyword(s) of interest.)

Basics topics (see "Patient education: Colon and rectal cancer (The Basics)")

Beyond the Basics topics (see "Patient education: Colon and rectal cancer (Beyond the Basics)" and "Patient education: Colorectal cancer treatment; metastatic cancer (Beyond the Basics)")

SUMMARY AND RECOMMENDATIONS

Pretreatment considerations – The selection of appropriate candidates for initial radiotherapy (RT) or chemoradiotherapy (CRT), rather than surgery, depends on accurate preoperative locoregional staging, which includes pelvic MRI and/or transrectal endoscopic ultrasound (TEUS). All patients should be tested for deficient mismatch repair (dMMR; germline and somatic tumor testing), given the potential to consider upfront immunotherapy. (See 'Pretreatment staging evaluation' above.)

Our approach to treatment is outlined in the algorithm (algorithm 1), and described below.

Patients with nonmetastatic disease, proficient mismatch repair

Indications for neoadjuvant therapy

-For patients with clinical (c)T3/4 rectal adenocarcinoma, we recommend preoperative (neoadjuvant) CRT or short-course RT rather than initial resection followed by adjuvant therapy (Grade 1B). (See 'T3/4 tumors' above.)

-For patients with clinically node-positive disease regardless of the primary tumor (T) stage, or a tumor that appears to invade or "threaten" the mesorectal fascia on preoperative imaging, we also suggest neoadjuvant therapy (Grade 2C). (See 'Relative indications' above.)

-The use of neoadjuvant therapy for a distal cT1-2N0 rectal cancer in an attempt to convert the operation from a needed abdominoperineal resection (APR) into a sphincter sparing procedure is controversial and not yet an accepted standard of care. However, if the patient is a poor surgical candidate or declines APR, initial RT or CRT may be chosen, followed by a restaging evaluation. (See 'T1-2N0 tumors' above.)

Short-course RT versus long-course CRT

-For most patients with bulky N2 or T4 tumors, we suggest conventional fractionation RT with concurrent fluoropyrimidine chemotherapy (ie, long-course CRT) rather than the short-course Swedish approach to RT alone (Grade 2C). (See 'Long-course chemoradiotherapy' above.)

Short-course RT represents an acceptable alternative to long-course CRT for many patients who do not have a cT4 or N2 tumor. (See 'Short-course radiotherapy' above.)

-For most patients undergoing long-course CRT, we suggest infusional fluorouracil (FU; 225 mg/m2 daily) administered five days per week during RT rather than bolus FU (Grade 2C). Oral capecitabine (825 mg/m2 twice daily, five days per week) is an appropriate alternative. (See 'Choice of chemotherapy during RT' above.)

Role of total neoadjuvant therapy

-For most patients with cT4 disease, an involved or threatened mesorectal fascia, clearly node-positive disease, a low-lying tumor, or extramural venous invasion, we suggest total neoadjuvant therapy (TNT; ie, oxaliplatin-based chemotherapy combined with either long-course CRT or short-course RT) rather than preoperative long-course CRT or short-course RT alone (Grade 2C). (See 'Total neoadjuvant therapy for locally advanced tumors' above.)

-If TNT is chosen, for most patients, we suggest initial RT, followed by 12 to 16 weeks of systemic oxaliplatin-based chemotherapy prior to surgery rather than chemotherapy followed by RT (Grade 2C). However, if metastatic disease is strongly suspected, but cannot be confirmed, we prefer to start with systemic chemotherapy followed by radiologic reevaluation; if metastatic disease is deemed unlikely, either long-course CRT or short-course RT can then be administered prior to surgery. If chemotherapy is initiated first, clinicians should repeat scans after two months of chemotherapy and move directly to RT or CRT if no response is seen. (See 'Sequencing of RT and chemotherapy' above.)

-If the TNT approach is used, we suggest limiting neoadjuvant chemotherapy to four months and eliminating postoperative chemotherapy. (See 'Adjuvant therapy' above.)

Management of complete clinical responders

-Patients who achieve a clinical complete response after conventional neoadjuvant therapy with no evidence of residual tumor on digital rectal examination (DRE), rectal MRI, and direct endoscopic evaluation may be considered for an initial nonoperative approach with an experienced multidisciplinary team. However, there is not uniform agreement on whether nonoperative approaches should be considered standard at this time, even among the authors and editors associated with this topic review. Any decision for nonoperative management should involve a careful discussion with the patient as to their risk tolerance. (See 'Avoidance of radical surgery' above.)

-For patients who choose nonoperative management, we repeat DRE, MRI, and endoscopic examination every three months for two years, and then every six months to complete a full five years of surveillance. (See 'Tumor response assessment and follow-up' above.)

Patients with nonmetastatic disease, deficient mismatch repair – Upfront immunotherapy followed by nonoperative management for clinical complete responders cannot yet be considered a standard approach for rectal cancers with dMMR. However, individuals with dMMR tumors who place a high value on avoiding the long-term morbidity associated with surgery and pelvic RT might choose this non-standard approach if they are willing to accept the uncertainty of long-term outcomes, the small risk of fatal side effects from checkpoint inhibitors, and the need for very close follow-up (every four month pelvic MRI, DRE, sigmoidoscopy with biopsy). (See 'Neoadjuvant immunotherapy for dMMR tumors' above.)

Patients with synchronous metastatic disease

For patients with synchronous, potentially resectable liver metastases, any of the following strategies is acceptable: initial chemotherapy followed by short-course RT or long-course CRT and then followed by resection (synchronous or staged), or initial CRT or short-course RT followed by chemotherapy and then resection. In this setting, we suggest short-course RT rather than long-course CRT (Grade 2C). (See 'Potentially resectable metastases' above.)

For patients presenting with synchronous, categorically unresectable stage IV disease and a symptomatic primary tumor who are not amenable to stenting, we suggest short-course pelvic RT in conjunction with modern combination systemic chemotherapy rather than chemotherapy alone (Grade 2C), This approach may allow selected patients to avoid surgery, even those with a nearly obstructing lesion. We do not generally pursue RT for asymptomatic patients with unresectable metastatic disease. (See 'Synchronous unresectable metastases' above.)

  1. Ma B, Gao P, Wang H, et al. What has preoperative radio(chemo)therapy brought to localized rectal cancer patients in terms of perioperative and long-term outcomes over the past decades? A systematic review and meta-analysis based on 41,121 patients. Int J Cancer 2017; 141:1052.
  2. Glynne-Jones R, Wyrwicz L, Tiret E, et al. Rectal cancer: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol 2017; 28:iv22.
  3. NCCN Clinical Practice Guidelines in Oncology. Available at: https://www.nccn.org/professionals/physician_gls/default.aspx (Accessed on April 12, 2021).
  4. Kapiteijn E, Marijnen CA, Nagtegaal ID, et al. Preoperative radiotherapy combined with total mesorectal excision for resectable rectal cancer. N Engl J Med 2001; 345:638.
  5. Peeters KC, Marijnen CA, Nagtegaal ID, et al. The TME trial after a median follow-up of 6 years: increased local control but no survival benefit in irradiated patients with resectable rectal carcinoma. Ann Surg 2007; 246:693.
  6. Chan E, Wise PE, Chakravarthy AB. Controversies in radiation for upper rectal cancers. J Natl Compr Canc Netw 2012; 10:1567.
  7. Nissan A, Stojadinovic A, Shia J, et al. Predictors of recurrence in patients with T2 and early T3, N0 adenocarcinoma of the rectum treated by surgery alone. J Clin Oncol 2006; 24:4078.
  8. Ruppert R, Kube R, Strassburg J, et al. Avoidance of Overtreatment of Rectal Cancer by Selective Chemoradiotherapy: Results of the Optimized Surgery and MRI-Based Multimodal Therapy Trial. J Am Coll Surg 2020; 231:413.
  9. Guillem JG, Díaz-González JA, Minsky BD, et al. cT3N0 rectal cancer: potential overtreatment with preoperative chemoradiotherapy is warranted. J Clin Oncol 2008; 26:368.
  10. Quirke P, Durdey P, Dixon MF, Williams NS. Local recurrence of rectal adenocarcinoma due to inadequate surgical resection. Histopathological study of lateral tumour spread and surgical excision. Lancet 1986; 2:996.
  11. Merkel S, Mansmann U, Siassi M, et al. The prognostic inhomogeneity in pT3 rectal carcinomas. Int J Colorectal Dis 2001; 16:298.
  12. Tan JJ, Carten RV, Babiker A, et al. Prognostic Importance of MRI-Detected Extramural Venous Invasion in Rectal Cancer: A Literature Review and Systematic Meta-Analysis. Int J Radiat Oncol Biol Phys 2021; 111:385.
  13. Rouleau Fournier F, Motamedi MAK, Brown CJ, et al. Oncologic Outcomes Associated With MRI-detected Extramural Venous Invasion (mrEMVI) in Rectal Cancer: A Systematic Review and Meta-analysis. Ann Surg 2022; 275:303.
  14. Massucco P, Fontana AP, Balbo Mussetto A, et al. MRI-detected extramural vascular invasion (mrEMVI) as the best predictive factor to identify candidates to total neoadjuvant therapy in locally advanced rectal cancer. J Surg Oncol 2022; 125:1024.
  15. Evans J, Patel U, Brown G. Rectal cancer: primary staging and assessment after chemoradiotherapy. Semin Radiat Oncol 2011; 21:169.
  16. Taylor FG, Quirke P, Heald RJ, et al. Preoperative high-resolution magnetic resonance imaging can identify good prognosis stage I, II, and III rectal cancer best managed by surgery alone: a prospective, multicenter, European study. Ann Surg 2011; 253:711.
  17. Allal AS, Bieri S, Pelloni A, et al. Sphincter-sparing surgery after preoperative radiotherapy for low rectal cancers: feasibility, oncologic results and quality of life outcomes. Br J Cancer 2000; 82:1131.
  18. Rider WD, Palmer JA, Mahoney LJ, Robertson CT. Preoperative irradiation in operable cancer of the rectum: report of the Toronto trial. Can J Surg 1977; 20:335.
  19. The evaluation of low dose pre-operative X-ray therapy in the management of operable rectal cancer; results of a randomly controlled trial. Br J Surg 1984; 71:21.
  20. Gérard A, Buyse M, Nordlinger B, et al. Preoperative radiotherapy as adjuvant treatment in rectal cancer. Final results of a randomized study of the European Organization for Research and Treatment of Cancer (EORTC). Ann Surg 1988; 208:606.
  21. Swedish Rectal Cancer Trial, Cedermark B, Dahlberg M, et al. Improved survival with preoperative radiotherapy in resectable rectal cancer. N Engl J Med 1997; 336:980.
  22. Minsky BD. Sphincter preservation for rectal cancer: fact or fiction? J Clin Oncol 2002; 20:1971.
  23. Wagman R, Minsky BD, Cohen AM, et al. Sphincter preservation in rectal cancer with preoperative radiation therapy and coloanal anastomosis: long term follow-up. Int J Radiat Oncol Biol Phys 1998; 42:51.
  24. Rouanet P, Saint-Aubert B, Lemanski C, et al. Restorative and nonrestorative surgery for low rectal cancer after high-dose radiation: long-term oncologic and functional results. Dis Colon Rectum 2002; 45:305.
  25. Hyams DM, Mamounas EP, Petrelli N, et al. A clinical trial to evaluate the worth of preoperative multimodality therapy in patients with operable carcinoma of the rectum: a progress report of National Surgical Breast and Bowel Project Protocol R-03. Dis Colon Rectum 1997; 40:131.
  26. Kuvshinoff B, Maghfoor I, Miedema B, et al. Distal margin requirements after preoperative chemoradiotherapy for distal rectal carcinomas: are < or = 1 cm distal margins sufficient? Ann Surg Oncol 2001; 8:163.
  27. Valentini V, Coco C, Cellini N, et al. Preoperative chemoradiation for extraperitoneal T3 rectal cancer: acute toxicity, tumor response, and sphincter preservation. Int J Radiat Oncol Biol Phys 1998; 40:1067.
  28. Francois Y, Nemoz CJ, Baulieux J, et al. Influence 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 trial. J Clin Oncol 1999; 17:2396.
  29. Grann A, Feng C, Wong D, et al. Preoperative combined modality therapy for clinically resectable uT3 rectal adenocarcinoma. Int J Radiat Oncol Biol Phys 2001; 49:987.
  30. Gambacorta MA, Valentini V, Morganti AG, et al. Chemoradiation with raltitrexed (Tomudex) in preoperative treatment of stage II-III resectable rectal cancer: a phase II study. Int J Radiat Oncol Biol Phys 2004; 60:130.
  31. Rengan R, Paty P, Wong WD, et al. Distal cT2N0 rectal cancer: is there an alternative to abdominoperineal resection? J Clin Oncol 2005; 23:4905.
  32. Sauer R, Becker H, Hohenberger W, et al. Preoperative versus postoperative chemoradiotherapy for rectal cancer. N Engl J Med 2004; 351:1731.
  33. Rullier E, Rouanet P, Tuech JJ, et al. Organ preservation for rectal cancer (GRECCAR 2): a prospective, randomised, open-label, multicentre, phase 3 trial. Lancet 2017; 390:469.
  34. Rullier E, Vendrely V, Asselineau J, et al. Organ preservation with chemoradiotherapy plus local excision for rectal cancer: 5-year results of the GRECCAR 2 randomised trial. Lancet Gastroenterol Hepatol 2020; 5:465.
  35. Verseveld M, de Graaf EJ, Verhoef C, et al. Chemoradiation therapy for rectal cancer in the distal rectum followed by organ-sparing transanal endoscopic microsurgery (CARTS study). Br J Surg 2015; 102:853.
  36. Garcia-Aguilar J, Renfro LA, Chow OS, et al. Organ preservation for clinical T2N0 distal rectal cancer using neoadjuvant chemoradiotherapy and local excision (ACOSOG Z6041): results of an open-label, single-arm, multi-institutional, phase 2 trial. Lancet Oncol 2015; 16:1537.
  37. Rouanet P, Rivoire M, Gourgou S, et al. Sphincter-saving surgery after neoadjuvant therapy for ultra-low rectal cancer where abdominoperineal resection was indicated: 10-year results of the GRECCAR 1 trial. Br J Surg 2021; 108:10.
  38. Weiser MR, Quah HM, Shia J, et al. Sphincter preservation in low rectal cancer is facilitated by preoperative chemoradiation and intersphincteric dissection. Ann Surg 2009; 249:236.
  39. Marchegiani F, Palatucci V, Capelli G, et al. Rectal Sparing Approach After Neoadjuvant Therapy in Patients with Rectal Cancer: The Preliminary Results of the ReSARCh Trial. Ann Surg Oncol 2022; 29:1880.
  40. Kennecke HF, O'Callaghan CJ, Loree JM, et al. Neoadjuvant Chemotherapy, Excision, and Observation for Early Rectal Cancer: The Phase II NEO Trial (CCTG CO.28) Primary End Point Results. J Clin Oncol 2022; :JCO2200184.
  41. National Comprehensive Cancer Network (NCCN). NCCN clinical practice guidelines in oncology. Available at: https://www.nccn.org/professionals/physician_gls (Accessed on May 18, 2022).
  42. Gérard JP, Conroy T, Bonnetain F, et al. Preoperative radiotherapy with or without concurrent fluorouracil and leucovorin in T3-4 rectal cancers: results of FFCD 9203. J Clin Oncol 2006; 24:4620.
  43. Bosset JF, Calais G, Mineur L, et al. Enhanced tumorocidal effect of chemotherapy with preoperative radiotherapy for rectal cancer: preliminary results--EORTC 22921. J Clin Oncol 2005; 23:5620.
  44. Boulis-Wassif S, Gerard A, Loygue J, et al. Final results of a randomized trial on the treatment of rectal cancer with preoperative radiotherapy alone or in combination with 5-fluorouracil, followed by radical surgery. Trial of the European Organization on Research and Treatment of Cancer Gastrointestinal Tract Cancer Cooperative Group. Cancer 1984; 53:1811.
  45. Ceelen WP, Van Nieuwenhove Y, Fierens K. Preoperative chemoradiation versus radiation alone for stage II and III resectable rectal cancer. Cochrane Database Syst Rev 2009; :CD006041.
  46. McCarthy K, Pearson K, Fulton R, Hewitt J. Pre-operative chemoradiation for non-metastatic locally advanced rectal cancer. Cochrane Database Syst Rev 2012; 12:CD008368.
  47. Sauer R, Liersch T, Merkel S, et al. Preoperative 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 years. J Clin Oncol 2012; 30:1926.
  48. Roh MS, Colangelo LH, O'Connell MJ, et al. Preoperative multimodality therapy improves disease-free survival in patients with carcinoma of the rectum: NSABP R-03. J Clin Oncol 2009; 27:5124.
  49. Park JH, Yoon SM, Yu CS, et al. Randomized phase 3 trial comparing preoperative and postoperative chemoradiotherapy with capecitabine for locally advanced rectal cancer. Cancer 2011; 117:3703.
  50. Rödel C, Martus P, Papadoupolos T, et al. Prognostic significance of tumor regression after preoperative chemoradiotherapy for rectal cancer. J Clin Oncol 2005; 23:8688.
  51. Fokas E, Liersch T, Fietkau R, et al. Tumor regression grading after preoperative chemoradiotherapy for locally advanced rectal carcinoma revisited: updated results of the CAO/ARO/AIO-94 trial. J Clin Oncol 2014; 32:1554.
  52. Chang GJ, Rodriguez-Bigas MA, Eng C, Skibber JM. Lymph node status after neoadjuvant radiotherapy for rectal cancer is a biologic predictor of outcome. Cancer 2009; 115:5432.
  53. Yeo SG, Kim DY, Kim TH, et al. Pathologic complete response of primary tumor following preoperative chemoradiotherapy for locally advanced rectal cancer: long-term outcomes and prognostic significance of pathologic nodal status (KROG 09-01). Ann Surg 2010; 252:998.
  54. Klos CL, Shellito PC, Rattner DW, et al. The effect of neoadjuvant chemoradiation therapy on the prognostic value of lymph nodes after rectal cancer surgery. Am J Surg 2010; 200:440.
  55. Park IJ, You YN, Agarwal A, et al. Neoadjuvant treatment response as an early response indicator for patients with rectal cancer. J Clin Oncol 2012; 30:1770.
  56. Wheeler JM, Warren BF, Mortensen NJ, et al. Quantification of histologic regression of rectal cancer after irradiation: a proposal for a modified staging system. Dis Colon Rectum 2002; 45:1051.
  57. Vecchio FM, Valentini V, Minsky BD, et al. The relationship of pathologic tumor regression grade (TRG) and outcomes after preoperative therapy in rectal cancer. Int J Radiat Oncol Biol Phys 2005; 62:752.
  58. Fokas E, Ströbel P, Fietkau R, et al. Tumor Regression Grading After Preoperative Chemoradiotherapy as a Prognostic Factor and Individual-Level Surrogate for Disease-Free Survival in Rectal Cancer. J Natl Cancer Inst 2017; 109.
  59. Jessup JM, Goldberg RM, Asare AE, et al. Colon and Rectum. In: AJCC Cancer Staging Manual, 8th ed, Amin MB (Ed), AJCC, Chicago 2017. p.251.
  60. Washington MK, Berlin J, Branton P, et al. Protocol for the examination of specimens from patients with primary carcinoma of the colon and rectum. Arch Pathol Lab Med 2009; 133:1539.
  61. Minsky BD, Rodel C. Identifying the most predictive post-chemoradiation TRG system for rectal cancer. J Natl Cancer Inst 2014; 106.
  62. Jäger T, Neureiter D, Urbas R, et al. Applicability of American Joint Committee on Cancer and College of American Pathologists Regression Grading System in Rectal Cancer. Dis Colon Rectum 2017; 60:815.
  63. Vychnevskaia K, Dumont F, Agostini J, et al. Prognostic Value of Sterilized Lymph Nodes After Preoperative Chemoradiotherapy for Patients with ypN0 Rectal Cancer. Ann Surg Oncol 2017; 24:1304.
  64. Delitto D, George TJ Jr, Loftus TJ, et al. Prognostic Value of Clinical vs Pathologic Stage in Rectal Cancer Patients Receiving Neoadjuvant Therapy. J Natl Cancer Inst 2018; 110:460.
  65. Huebner M, Wolff BG, Smyrk TC, et al. Partial pathologic response and nodal status as most significant prognostic factors for advanced rectal cancer treated with preoperative chemoradiotherapy. World J Surg 2012; 36:675.
  66. Dhadda AS, Zaitoun AM, Bessell EM. Regression of rectal cancer with radiotherapy with or without concurrent capecitabine--optimising the timing of surgical resection. Clin Oncol (R Coll Radiol) 2009; 21:23.
  67. Cotte E, Passot G, Decullier E, et al. Pathologic Response, When Increased by Longer Interval, Is a Marker but Not the Cause of Good Prognosis in Rectal Cancer: 17-year Follow-up of the Lyon R90-01 Randomized Trial. Int J Radiat Oncol Biol Phys 2016; 94:544.
  68. Lefevre JH, Mineur L, Kotti S, et al. Effect of Interval (7 or 11 weeks) Between Neoadjuvant Radiochemotherapy and Surgery on Complete Pathologic Response in Rectal Cancer: A Multicenter, Randomized, Controlled Trial (GRECCAR-6). J Clin Oncol 2016.
  69. Saglam S, Bugra D, Saglam EK, et al. Fourth versus eighth week surgery after neoadjuvant radiochemotherapy in T3-4/N0+ rectal cancer: Istanbul R-01 study. J Gastrointest Oncol 2014; 5:9.
  70. Foster JD, Ewings P, Falk S, et al. Surgical timing after chemoradiotherapy for rectal cancer, analysis of technique (STARRCAT): results of a feasibility multi-centre randomized controlled trial. Tech Coloproctol 2016; 20:683.
  71. Akgun E, Caliskan C, Bozbiyik O, et al. Randomized clinical trial of short or long interval between neoadjuvant chemoradiotherapy and surgery for rectal cancer. Br J Surg 2018; 105:1417.
  72. Ryan ÉJ, O'Sullivan DP, Kelly ME, et al. Meta-analysis of the effect of extending the interval after long-course chemoradiotherapy before surgery in locally advanced rectal cancer. Br J Surg 2019; 106:1298.
  73. Deidda S, Elmore U, Rosati R, et al. Association of Delayed Surgery With Oncologic Long-term Outcomes in Patients With Locally Advanced Rectal Cancer Not Responding to Preoperative Chemoradiation. JAMA Surg 2021; 156:1141.
  74. Enker WE, Merchant N, Cohen AM, et al. Safety and efficacy of low anterior resection for rectal cancer: 681 consecutive cases from a specialty service. Ann Surg 1999; 230:544.
  75. Ulrich A, Weitz J, Slodczyk M, et al. Neoadjuvant treatment does not influence perioperative outcome in rectal cancer surgery. Int J Radiat Oncol Biol Phys 2009; 75:129.
  76. Loos M, Quentmeier P, Schuster T, et al. Effect of preoperative radio(chemo)therapy on long-term functional outcome in rectal cancer patients: a systematic review and meta-analysis. Ann Surg Oncol 2013; 20:1816.
  77. Herman MP, Kopetz S, Bhosale PR, et al. Sacral insufficiency fractures after preoperative chemoradiation for rectal cancer: incidence, risk factors, and clinical course. Int J Radiat Oncol Biol Phys 2009; 74:818.
  78. Inoue Y, Miki C, Ojima E, et al. Pelvic insufficiency fractures after preoperative radiotherapy for rectal carcinoma. Int J Clin Oncol 2003; 8:336.
  79. Kim HJ, Boland PJ, Meredith DS, et al. Fractures of the sacrum after chemoradiation for rectal carcinoma: incidence, risk factors, and radiographic evaluation. Int J Radiat Oncol Biol Phys 2012; 84:694.
  80. Mohiuddin M, Regine WF, John WJ, et al. Preoperative chemoradiation in fixed distal rectal cancer: dose time factors for pathological complete response. Int J Radiat Oncol Biol Phys 2000; 46:883.
  81. Hofheinz RD, Wenz F, Post S, et al. Chemoradiotherapy with capecitabine versus fluorouracil for locally advanced rectal cancer: a randomised, multicentre, non-inferiority, phase 3 trial. Lancet Oncol 2012; 13:579.
  82. O'Connell MJ, Colangelo LH, Beart RW, et al. Capecitabine and oxaliplatin in the preoperative multimodality treatment of rectal cancer: surgical end points from National Surgical Adjuvant Breast and Bowel Project trial R-04. J Clin Oncol 2014; 32:1927.
  83. Allegra CJ, Yothers G, O'Connell MJ, et al. Neoadjuvant 5-FU or Capecitabine Plus Radiation With or Without Oxaliplatin in Rectal Cancer Patients: A Phase III Randomized Clinical Trial. J Natl Cancer Inst 2015; 107.
  84. Zou XC, Wang QW, Zhang JM. Comparison of 5-FU-based and Capecitabine-based Neoadjuvant Chemoradiotherapy in Patients With Rectal Cancer: A Meta-analysis. Clin Colorectal Cancer 2017; 16:e123.
  85. Gieschke R, Burger HU, Reigner B, et al. Population pharmacokinetics and concentration-effect relationships of capecitabine metabolites in colorectal cancer patients. Br J Clin Pharmacol 2003; 55:252.
  86. Aschele C, Cionini L, Lonardi S, et al. Primary tumor response to preoperative chemoradiation with or without oxaliplatin in locally advanced rectal cancer: pathologic results of the STAR-01 randomized phase III trial. J Clin Oncol 2011; 29:2773.
  87. Gérard JP, Azria D, Gourgou-Bourgade S, et al. Comparison of two neoadjuvant chemoradiotherapy regimens for locally advanced rectal cancer: results of the phase III trial ACCORD 12/0405-Prodige 2. J Clin Oncol 2010; 28:1638.
  88. Gérard JP, Azria D, Gourgou-Bourgade S, et al. Clinical outcome of the ACCORD 12/0405 PRODIGE 2 randomized trial in rectal cancer. J Clin Oncol 2012; 30:4558.
  89. Rödel C, Liersch T, Becker H, et al. Preoperative 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 trial. Lancet Oncol 2012; 13:679.
  90. Rödel C, Graeven U, Fietkau R, et al. Oxaliplatin added to fluorouracil-based preoperative chemoradiotherapy and postoperative chemotherapy of locally advanced rectal cancer (the German CAO/ARO/AIO-04 study): final results of the multicentre, open-label, randomised, phase 3 trial. Lancet Oncol 2015; 16:979.
  91. Jiao D, Zhang R, Gong Z, et al. Fluorouracil-based preoperative chemoradiotherapy with or without oxaliplatin for stage II/III rectal cancer: a 3-year follow-up study. Chin J Cancer Res 2015; 27:588.
  92. Haddad P, Miraie M, Farhan F, et al. Addition of oxaliplatin to neoadjuvant radiochemotherapy in MRI-defined T3, T4 or N+ rectal cancer: a randomized clinical trial. Asia Pac J Clin Oncol 2017; 13:416.
  93. Saha A, Ghosh SK, Roy C, et al. A randomized controlled pilot study to compare capecitabine-oxaliplatin with 5-FU-leucovorin as neoadjuvant concurrent chemoradiation in locally advanced adenocarcinoma of rectum. J Cancer Res Ther 2015; 11:88.
  94. Kayal PK, Saha A, Dastidar AG, et al. A randomized comparative study between neoadjuvant 5-fluorouracil and leukovorin versus 5-fluorouracil and cisplatin along with concurrent radiation in locally advanced carcinoma rectum. Clin Cancer Invest J 2014; 3:32.
  95. Deng Y, Chi P, Lan P, et al. Neoadjuvant Modified FOLFOX6 With or Without Radiation Versus Fluorouracil Plus Radiation for Locally Advanced Rectal Cancer: Final Results of the Chinese FOWARC Trial. J Clin Oncol 2019; 37:3223.
  96. Schmoll HJ, Stein A, Van Cutsem E, et al. Pre- and Postoperative Capecitabine Without or With Oxaliplatin in Locally Advanced Rectal Cancer: PETACC 6 Trial by EORTC GITCG and ROG, AIO, AGITG, BGDO, and FFCD. J Clin Oncol 2021; 39:17.
  97. Aschele C, Lonardi S, Cionini L, et al. Final results of STAR-01: A randomized phase III trial comparing preoperative chemoradiation with or without oxaliplatin in locally advanced rectal cancer. J Clin Oncol 2016; 34S: ASCO #3521.
  98. Hüttner FJ, Probst P, Kalkum E, et al. Addition of Platinum Derivatives to Fluoropyrimidine-Based Neoadjuvant Chemoradiotherapy for Stage II/III Rectal Cancer: Systematic Review and Meta-Analysis. J Natl Cancer Inst 2019; 111:887.
  99. Navarro M, Dotor E, Rivera F, et al. A Phase II study of preoperative radiotherapy and concomitant weekly irinotecan in combination with protracted venous infusion 5-fluorouracil, for resectable locally advanced rectal cancer. Int J Radiat Oncol Biol Phys 2006; 66:201.
  100. Willeke F, Horisberger K, Kraus-Tiefenbacher U, et al. A phase II study of capecitabine and irinotecan in combination with concurrent pelvic radiotherapy (CapIri-RT) as neoadjuvant treatment of locally advanced rectal cancer. Br J Cancer 2007; 96:912.
  101. Gollins S, Sun Myint A, Haylock B, et al. Preoperative chemoradiotherapy using concurrent capecitabine and irinotecan in magnetic resonance imaging-defined locally advanced rectal cancer: impact on long-term clinical outcomes. J Clin Oncol 2011; 29:1042.
  102. Mohiuddin M, Paulus R, Mitchell E, et al. Neoadjuvant chemoradiation for distal rectal cancer: 5-year updated results of a randomized phase 2 study of neoadjuvant combined modality chemoradiation for distal rectal cancer. Int J Radiat Oncol Biol Phys 2013; 86:523.
  103. Willett CG, Duda DG, di Tomaso E, et al. Efficacy, safety, and biomarkers of neoadjuvant bevacizumab, radiation therapy, and fluorouracil in rectal cancer: a multidisciplinary phase II study. J Clin Oncol 2009; 27:3020.
  104. Crane CH, Eng C, Feig BW, et al. Phase II trial of neoadjuvant bevacizumab, capecitabine, and radiotherapy for locally advanced rectal cancer. Int J Radiat Oncol Biol Phys 2010; 76:824.
  105. Uehara K, Hiramatsu K, Maeda A, et al. Neoadjuvant oxaliplatin and capecitabine and bevacizumab without radiotherapy for poor-risk rectal cancer: N-SOG 03 Phase II trial. Jpn J Clin Oncol 2013; 43:964.
  106. Landry JC, Feng Y, Prabhu RS, et al. Phase II Trial of Preoperative Radiation With Concurrent Capecitabine, Oxaliplatin, and Bevacizumab Followed by Surgery and Postoperative 5-Fluorouracil, Leucovorin, Oxaliplatin (FOLFOX), and Bevacizumab in Patients With Locally Advanced Rectal Cancer: 5-Year Clinical Outcomes ECOG-ACRIN Cancer Research Group E3204. Oncologist 2015; 20:615.
  107. Fernández-Martos C, Pericay C, Losa F, et al. Effect of Aflibercept Plus Modified FOLFOX6 Induction Chemotherapy Before Standard Chemoradiotherapy and Surgery in Patients With High-Risk Rectal Adenocarcinoma: The GEMCAD 1402 Randomized Clinical Trial. JAMA Oncol 2019; 5:1566.
  108. Velenik V, Ocvirk J, Oblak I, Anderluh F. A phase II study of cetuximab, capecitabine and radiotherapy in neoadjuvant treatment of patients with locally advanced resectable rectal cancer. Eur J Surg Oncol 2010; 36:244.
  109. Horisberger K, Treschl A, Mai S, et al. Cetuximab in combination with capecitabine, irinotecan, and radiotherapy for patients with locally advanced rectal cancer: results of a Phase II MARGIT trial. Int J Radiat Oncol Biol Phys 2009; 74:1487.
  110. Dewdney A, Cunningham D, Tabernero J, et al. Multicenter 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 Oncol 2012; 30:1620.
  111. Kim SY, Shim EK, Yeo HY, et al. KRAS mutation status and clinical outcome of preoperative chemoradiation with cetuximab in locally advanced rectal cancer: a pooled analysis of 2 phase II trials. Int J Radiat Oncol Biol Phys 2013; 85:201.
  112. Helbling D, Bodoky G, Gautschi O, et al. Neoadjuvant chemoradiotherapy with or without panitumumab in patients with wild-type KRAS, locally advanced rectal cancer (LARC): a randomized, multicenter, phase II trial SAKK 41/07. Ann Oncol 2013; 24:718.
  113. Sclafani F, Gonzalez D, Cunningham D, et al. TP53 mutational status and cetuximab benefit in rectal cancer: 5-year results of the EXPERT-C trial. J Natl Cancer Inst 2014; 106.
  114. Joye I, Haustermans K. Clinical target volume delineation for rectal cancer radiation therapy: time for updated guidelines? Int J Radiat Oncol Biol Phys 2015; 91:690.
  115. Hernando-Requejo O, López M, Cubillo A, et al. Complete pathological responses in locally advanced rectal cancer after preoperative IMRT and integrated-boost chemoradiation. Strahlenther Onkol 2014; 190:515.
  116. Zhu J, Liu F, Gu W, et al. Concomitant boost IMRT-based neoadjuvant chemoradiotherapy for clinical stage II/III rectal adenocarcinoma: results of a phase II study. Radiat Oncol 2014; 9:70.
  117. Parekh A, Truong MT, Pashtan I, et al. Acute gastrointestinal toxicity and tumor response with preoperative intensity modulated radiation therapy for rectal cancer. Gastrointest Cancer Res 2013; 6:137.
  118. Engels B, Platteaux N, Van den Begin R, et al. Preoperative intensity-modulated and image-guided radiotherapy with a simultaneous integrated boost in locally advanced rectal cancer: report on late toxicity and outcome. Radiother Oncol 2014; 110:155.
  119. Jabbour SK, Patel S, Herman JM, et al. Intensity-modulated radiation therapy for rectal carcinoma can reduce treatment breaks and emergency department visits. Int J Surg Oncol 2012; 2012:891067.
  120. Arbea L, Martínez-Monge R, Díaz-González JA, et al. Four-week neoadjuvant intensity-modulated radiation therapy with concurrent capecitabine and oxaliplatin in locally advanced rectal cancer patients: a validation phase II trial. Int J Radiat Oncol Biol Phys 2012; 83:587.
  121. Samuelian JM, Callister MD, Ashman JB, et al. Reduced acute bowel toxicity in patients treated with intensity-modulated radiotherapy for rectal cancer. Int J Radiat Oncol Biol Phys 2012; 82:1981.
  122. Li Y, Wang L, Li Z, et al. Compliance and safety of neoadjuvant intensity modulated radiotherapy (IMRT) with concurrent capecitabine for locally advanced rectal cancer: Updated results from a phase II trial (ChiCTR-TNC-10001094). J Clin Oncol 2014; 32S: ASCO #3598.
  123. Birgisson H, Påhlman L, Gunnarsson U, et al. Adverse effects of preoperative radiation therapy for rectal cancer: long-term follow-up of the Swedish Rectal Cancer Trial. J Clin Oncol 2005; 23:8697.
  124. Marijnen CA, Kapiteijn E, van de Velde CJ, et al. Acute side effects and complications after short-term preoperative radiotherapy combined with total mesorectal excision in primary rectal cancer: report of a multicenter randomized trial. J Clin Oncol 2002; 20:817.
  125. Marijnen CA, van de Velde CJ, Putter H, et al. Impact of short-term preoperative radiotherapy on health-related quality of life and sexual functioning in primary rectal cancer: report of a multicenter randomized trial. J Clin Oncol 2005; 23:1847.
  126. Peeters KC, van de Velde CJ, Leer JW, et al. Late side effects of short-course preoperative radiotherapy combined with total mesorectal excision for rectal cancer: increased bowel dysfunction in irradiated patients--a Dutch colorectal cancer group study. J Clin Oncol 2005; 23:6199.
  127. Wiltink LM, Chen TY, Nout RA, et al. Health-related quality of life 14 years after preoperative short-term radiotherapy and total mesorectal excision for rectal cancer: report of a multicenter randomised trial. Eur J Cancer 2014; 50:2390.
  128. Bujko K, Nowacki MP, Nasierowska-Guttmejer A, et al. Long-term results of a randomized trial comparing preoperative short-course radiotherapy with preoperative conventionally fractionated chemoradiation for rectal cancer. Br J Surg 2006; 93:1215.
  129. Ngan SY, Burmeister B, Fisher RJ, et al. Randomized trial of short-course radiotherapy versus long-course chemoradiation comparing rates of local recurrence in patients with T3 rectal cancer: Trans-Tasman Radiation Oncology Group trial 01.04. J Clin Oncol 2012; 30:3827.
  130. Chakrabarti D, Rajan S, Akhtar N, et al. Short-course radiotherapy with consolidation chemotherapy versus conventionally fractionated long-course chemoradiotherapy for locally advanced rectal cancer: randomized clinical trial. Br J Surg 2021; 108:511.
  131. Ciseł B, Pietrzak L, Michalski W, et al. Long-course preoperative chemoradiation versus 5 × 5 Gy and consolidation chemotherapy for clinical T4 and fixed clinical T3 rectal cancer: long-term results of the randomized Polish II study. Ann Oncol 2019; 30:1298.
  132. Kairevičė L, Latkauskas T, Tamelis A, et al. Preoperative long-course chemoradiotherapy plus adjuvant chemotherapy versus short-course radiotherapy without adjuvant chemotherapy both with delayed surgery for stage II-III resectable rectal cancer: 5-Year survival data of a randomized controlled trial. Medicina (Kaunas) 2017; 53:150.
  133. Socha J, Kairevice L, Kępka L, et al. Should Short-Course Neoadjuvant Radiation Therapy Be Applied for Low-Lying Rectal Cancer? A Systematic Review and Meta-Analysis of the Randomized Trials. Int J Radiat Oncol Biol Phys 2020; 108:1257.
  134. Sebag-Montefiore D, Stephens RJ, Steele R, et al. Preoperative radiotherapy versus selective postoperative chemoradiotherapy in patients with rectal cancer (MRC CR07 and NCIC-CTG C016): a multicentre, randomised trial. Lancet 2009; 373:811.
  135. Radu C, Berglund A, Påhlman L, Glimelius B. Short-course preoperative radiotherapy with delayed surgery in rectal cancer - a retrospective study. Radiother Oncol 2008; 87:343.
  136. Pettersson D, Holm T, Iversen H, et al. Preoperative short-course radiotherapy with delayed surgery in primary rectal cancer. Br J Surg 2012; 99:577.
  137. Hatfield P, Hingorani M, Radhakrishna G, et al. Short-course radiotherapy, with elective delay prior to surgery, in patients with unresectable rectal cancer who have poor performance status or significant co-morbidity. Radiother Oncol 2009; 92:210.
  138. Erlandsson J, Lörinc E, Ahlberg M, et al. Tumour regression after radiotherapy for rectal cancer - Results from the randomised Stockholm III trial. Radiother Oncol 2019; 135:178.
  139. van den Broek CB, Vermeer TA, Bastiaannet E, et al. Impact of the interval between short-course radiotherapy and surgery on outcomes of rectal cancer patients. Eur J Cancer 2013; 49:3131.
  140. Pettersson D, Glimelius B, Iversen H, et al. Impaired postoperative leucocyte counts after preoperative radiotherapy for rectal cancer in the Stockholm III Trial. Br J Surg 2013; 100:969.
  141. Fokstuen T, Holm T, Glimelius B. Postoperative morbidity and mortality in relation to leukocyte counts and time to surgery after short-course preoperative radiotherapy for rectal cancer. Radiother Oncol 2009; 93:293.
  142. Erlandsson J, Holm T, Pettersson D, 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.
  143. Erlandsson J, Pettersson D, Glimelius B, et al. Postoperative complications in relation to overall treatment time in patients with rectal cancer receiving neoadjuvant radiotherapy. Br J Surg 2019; 106:1248.
  144. Cercek A, Dos Santos Fernandes G, Roxburgh CS, et al. Mismatch Repair-Deficient Rectal Cancer and Resistance to Neoadjuvant Chemotherapy. Clin Cancer Res 2020; 26:3271.
  145. Garcia-Aguilar J, Chow OS, Smith DD, et al. Effect of adding mFOLFOX6 after neoadjuvant chemoradiation in locally advanced rectal cancer: a multicentre, phase 2 trial. Lancet Oncol 2015; 16:957.
  146. Cercek A, Roxburgh CSD, Strombom P, et al. Adoption of Total Neoadjuvant Therapy for Locally Advanced Rectal Cancer. JAMA Oncol 2018; 4:e180071.
  147. Petrelli F, Trevisan F, Cabiddu M, et al. Total Neoadjuvant Therapy in Rectal Cancer: A Systematic Review and Meta-analysis of Treatment Outcomes. Ann Surg 2020; 271:440.
  148. Garcia-Aguilar J, Patil S, Gollub MJ, et al. Organ Preservation in Patients With Rectal Adenocarcinoma Treated With Total Neoadjuvant Therapy. J Clin Oncol 2022; 40:2546.
  149. Shi DD, Mamon HJ. Playing With Dynamite? A Cautious Assessment of TNT. J Clin Oncol 2021; 39:103.
  150. Conroy T, Lamfichekh N, Etienne PL, et al. Total neoadjuvant therapy with mFOLFIRINOX versus preoperative chemoradiation in patients with locally advanced rectal cancer: Final results of PRODIGE 23 phase III trial, a UNICANCER GI trial. J Clin Oncol 2020; 38S: ASCO #4007.
  151. van der Valk MJM, Marijnen CAM, van Etten B, et al. Compliance and tolerability of short-course radiotherapy followed by preoperative chemotherapy and surgery for high-risk rectal cancer - Results of the international randomized RAPIDO-trial. Radiother Oncol 2020; 147:75.
  152. Markovina S, Youssef F, Roy A, et al. Improved Metastasis- and Disease-Free Survival With Preoperative Sequential Short-Course Radiation Therapy and FOLFOX Chemotherapy for Rectal Cancer Compared With Neoadjuvant Long-Course Chemoradiotherapy: Results of a Matched Pair Analysis. Int J Radiat Oncol Biol Phys 2017; 99:417.
  153. Kasi A, Abbasi S, Handa S, et al. Total Neoadjuvant Therapy vs Standard Therapy in Locally Advanced Rectal Cancer: A Systematic Review and Meta-analysis. JAMA Netw Open 2020; 3:e2030097.
  154. Liu S, Jiang T, Xiao L, et al. Total Neoadjuvant Therapy (TNT) versus Standard Neoadjuvant Chemoradiotherapy for Locally Advanced Rectal Cancer: A Systematic Review and Meta-Analysis. Oncologist 2021; 26:e1555.
  155. Bahadoer RR, Dijkstra EA, van Etten B, 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.
  156. Kong JC, Soucisse M, Michael M, et al. Total Neoadjuvant Therapy in Locally Advanced Rectal Cancer: A Systematic Review and Metaanalysis of Oncological and Operative Outcomes. Ann Surg Oncol 2021; 28:7476.
  157. Bujko K, Wyrwicz L, Rutkowski A, et al. Long-course oxaliplatin-based preoperative chemoradiation versus 5 × 5 Gy and consolidation chemotherapy for cT4 or fixed cT3 rectal cancer: results of a randomized phase III study. Ann Oncol 2016; 27:834.
  158. Chua YJ, Barbachano Y, Cunningham D, et al. Neoadjuvant capecitabine and oxaliplatin before chemoradiotherapy and total mesorectal excision in MRI-defined poor-risk rectal cancer: a phase 2 trial. Lancet Oncol 2010; 11:241.
  159. Schou JV, Larsen FO, Rasch L, et al. Induction chemotherapy with capecitabine and oxaliplatin followed by chemoradiotherapy before total mesorectal excision in patients with locally advanced rectal cancer. Ann Oncol 2012; 23:2627.
  160. Maréchal R, Vos B, Polus M, et al. Short course chemotherapy followed by concomitant chemoradiotherapy and surgery in locally advanced rectal cancer: a randomized multicentric phase II study. Ann Oncol 2012; 23:1525.
  161. Fernandez-Martos C, Garcia-Albeniz X, Pericay C, et al. Chemoradiation, surgery and adjuvant chemotherapy versus induction chemotherapy followed by chemoradiation and surgery: long-term results of the Spanish GCR-3 phase II randomized trial†. Ann Oncol 2015; 26:1722.
  162. Chau I, Brown G, Cunningham D, et al. Neoadjuvant capecitabine and oxaliplatin followed by synchronous chemoradiation and total mesorectal excision in magnetic resonance imaging-defined poor-risk rectal cancer. J Clin Oncol 2006; 24:668.
  163. Sclafani F, Brown G, Cunningham D, et al. PAN-EX: a pooled analysis of two trials of neoadjuvant chemotherapy followed by chemoradiotherapy in MRI-defined, locally advanced rectal cancer. Ann Oncol 2016; 27:1557.
  164. Perez K, Safran H, Sikov W, et al. Complete Neoadjuvant Treatment for Rectal Cancer: The Brown University Oncology Group CONTRE Study. Am J Clin Oncol 2017; 40:283.
  165. Ludmir EB, Palta M, Willett CG, Czito BG. Total neoadjuvant therapy for rectal cancer: An emerging option. Cancer 2017; 123:1497.
  166. Fokas E, Allgäuer M, Polat B, et al. Randomized Phase II Trial of Chemoradiotherapy Plus Induction or Consolidation Chemotherapy as Total Neoadjuvant Therapy for Locally Advanced Rectal Cancer: CAO/ARO/AIO-12. J Clin Oncol 2019; 37:3212.
  167. Conroy T, Bosset JF, Etienne PL, 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.
  168. Dijkstra EA, Hospers GAP, Kranenbarg EM, et al. Quality of life and late toxicity after short-course radiotherapy followed by chemotherapy or chemoradiotherapy for locally advanced rectal cancer - The RAPIDO trial. Radiother Oncol 2022; 171:69.
  169. Jin J, Tang Y, Hu C, et al. Multicenter, Randomized, Phase III Trial of Short-Term Radiotherapy Plus Chemotherapy Versus Long-Term Chemoradiotherapy in Locally Advanced Rectal Cancer (STELLAR). J Clin Oncol 2022; 40:1681.
  170. Fokas E, Schlenska-Lange A, Polat B, et al. Chemoradiotherapy Plus Induction or Consolidation Chemotherapy as Total Neoadjuvant Therapy for Patients With Locally Advanced Rectal Cancer: Long-term Results of the CAO/ARO/AIO-12 Randomized Clinical Trial. JAMA Oncol 2022; 8:e215445.
  171. Romesser PB, Smith G, Crane CH. Nonoperative Management of Rectal Cancer Shows Cost-Effectiveness, but Can Comparative Effectiveness Be Established? J Natl Cancer Inst 2020; 112:771.
  172. You YN, Hardiman KM, Bafford A, et al. The American Society of Colon and Rectal Surgeons Clinical Practice Guidelines for the Management of Rectal Cancer. Dis Colon Rectum 2020; 63:1191.
  173. Habr-Gama A, Perez RO, Nadalin W, et al. Operative versus nonoperative treatment for stage 0 distal rectal cancer following chemoradiation therapy: long-term results. Ann Surg 2004; 240:711.
  174. Martin ST, Heneghan HM, Winter DC. Systematic review and meta-analysis of outcomes following pathological complete response to neoadjuvant chemoradiotherapy for rectal cancer. Br J Surg 2012; 99:918.
  175. Hospers G, Bahadoer RR, Dijkstra EA, et al. Short-course radiotherapy followed by chemotherapy before TME in locally advanced rectal cancer: The randomized RAPIDO trial. J Clin Oncol 2020; 38S: ASCO #4006.
  176. Maas M, Nelemans PJ, Valentini V, et al. Long-term outcome in patients with a pathological complete response after chemoradiation for rectal cancer: a pooled analysis of individual patient data. Lancet Oncol 2010; 11:835.
  177. Habr-Gama A, Perez RO, Proscurshim I, et al. Patterns of failure and survival for nonoperative treatment of stage c0 distal rectal cancer following neoadjuvant chemoradiation therapy. J Gastrointest Surg 2006; 10:1319.
  178. Maas M, Beets-Tan RG, Lambregts DM, et al. Wait-and-see policy for clinical complete responders after chemoradiation for rectal cancer. J Clin Oncol 2011; 29:4633.
  179. Glynne-Jones R, Hughes R. Critical appraisal of the 'wait and see' approach in rectal cancer for clinical complete responders after chemoradiation. Br J Surg 2012; 99:897.
  180. Renehan AG, Malcomson L, Emsley R, et al. Watch-and-wait approach versus surgical resection after chemoradiotherapy for patients with rectal cancer (the OnCoRe project): a propensity-score matched cohort analysis. Lancet Oncol 2016; 17:174.
  181. Appelt AL, Pløen J, Harling H, et al. High-dose chemoradiotherapy and watchful waiting for distal rectal cancer: a prospective observational study. Lancet Oncol 2015; 16:919.
  182. Habr-Gama A, Gama-Rodrigues J, São Julião GP, et al. Local recurrence after complete clinical response and watch and wait in rectal cancer after neoadjuvant chemoradiation: impact of salvage therapy on local disease control. Int J Radiat Oncol Biol Phys 2014; 88:822.
  183. Habr-Gama A, São Julião GP, Gama-Rodrigues J, et al. Baseline T Classification Predicts Early Tumor Regrowth After Nonoperative Management in Distal Rectal Cancer After Extended Neoadjuvant Chemoradiation and Initial Complete Clinical Response. Dis Colon Rectum 2017; 60:586.
  184. Martens MH, Maas M, Heijnen LA, et al. Long-term Outcome of an Organ Preservation Program After Neoadjuvant Treatment for Rectal Cancer. J Natl Cancer Inst 2016; 108.
  185. Smith RK, Fry RD, Mahmoud NN, Paulson EC. Surveillance after neoadjuvant therapy in advanced rectal cancer with complete clinical response can have comparable outcomes to total mesorectal excision. Int J Colorectal Dis 2015; 30:769.
  186. Dossa F, Chesney TR, Acuna SA, Baxter NN. A watch-and-wait approach for locally advanced rectal cancer after a clinical complete response following neoadjuvant chemoradiation: a systematic review and meta-analysis. Lancet Gastroenterol Hepatol 2017; 2:501.
  187. van der Valk MJM, Hilling DE, Bastiaannet E, et al. Long-term outcomes of clinical complete responders after neoadjuvant treatment for rectal cancer in the International Watch & Wait Database (IWWD): an international multicentre registry study. Lancet 2018; 391:2537.
  188. Smith JJ, Strombom P, Chow OS, et al. Assessment of a Watch-and-Wait Strategy for Rectal Cancer in Patients With a Complete Response After Neoadjuvant Therapy. JAMA Oncol 2019; 5:e185896.
  189. Spiegel DY, Boyer MJ, Hong JC, et al. Long-term Clinical Outcomes of Nonoperative Management With Chemoradiotherapy for Locally Advanced Rectal Cancer in the Veterans Health Administration. Int J Radiat Oncol Biol Phys 2019; 103:565.
  190. Sanford NN, Dee EC, Ahn C, et al. Recent Trends and Overall Survival of Young Versus Older Adults With Stage II to III Rectal Cancer Treated With and Without Surgery in the United States, 2010-2015. Am J Clin Oncol 2020; 43:694.
  191. Hallam S, Messenger DE, Thomas MG. A Systematic Review of Local Excision After Neoadjuvant Therapy for Rectal Cancer: Are ypT0 Tumors the Limit? Dis Colon Rectum 2016; 59:984.
  192. Wyatt JNR, Powell SG, Altaf K, et al. Completion Total Mesorectal Excision After Transanal Local Excision of Early Rectal Cancer: A Systematic Review and Meta-analysis. Dis Colon Rectum 2022; 65:628.
  193. Sprenger T, Rothe H, Conradi LC, et al. Stage-Dependent Frequency of Lymph Node Metastases in Patients With Rectal Carcinoma After Preoperative Chemoradiation: Results from the CAO/ARO/AIO-94 Trial and From a Comparative Prospective Evaluation With Extensive Pathological Workup. Dis Colon Rectum 2016; 59:377.
  194. Smith JJ, Chow OS, Gollub MJ, et al. Organ Preservation in Rectal Adenocarcinoma: a phase II randomized controlled trial evaluating 3-year disease-free survival in patients with locally advanced rectal cancer treated with chemoradiation plus induction or consolidation chemotherapy, and total mesorectal excision or nonoperative management. BMC Cancer 2015; 15:767.
  195. Hiotis SP, Weber SM, Cohen AM, et al. Assessing the predictive value of clinical complete response to neoadjuvant therapy for rectal cancer: an analysis of 488 patients. J Am Coll Surg 2002; 194:131.
  196. Stipa F, Zernecke A, Moore HG, et al. Residual mesorectal lymph node involvement following neoadjuvant combined-modality therapy: rationale for radical resection? Ann Surg Oncol 2004; 11:187.
  197. Zmora O, Dasilva GM, Gurland B, et al. Does rectal wall tumor eradication with preoperative chemoradiation permit a change in the operative strategy? Dis Colon Rectum 2004; 47:1607.
  198. Habr-Gama A, Perez RO, Nadalin W, et al. Long-term results of preoperative chemoradiation for distal rectal cancer correlation between final stage and survival. J Gastrointest Surg 2005; 9:90.
  199. Patel UB, Taylor F, Blomqvist L, et al. Magnetic resonance imaging-detected tumor response for locally advanced rectal cancer predicts survival outcomes: MERCURY experience. J Clin Oncol 2011; 29:3753.
  200. Kristiansen C, Loft A, Berthelsen AK, et al. PET/CT and histopathologic response to preoperative chemoradiation therapy in locally advanced rectal cancer. Dis Colon Rectum 2008; 51:21.
  201. Gollub MJ, Gultekin DH, Akin O, et al. Dynamic contrast enhanced-MRI for the detection of pathological complete response to neoadjuvant chemotherapy for locally advanced rectal cancer. Eur Radiol 2012; 22:821.
  202. Perez RO, Habr-Gama A, Gama-Rodrigues J, et al. Accuracy 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). Cancer 2012; 118:3501.
  203. Zhang C, Tong J, Sun X, et al. 18F-FDG-PET evaluation of treatment response to neo-adjuvant therapy in patients with locally advanced rectal cancer: a meta-analysis. Int J Cancer 2012; 131:2604.
  204. Guillem JG, Ruby JA, Leibold T, et al. Neither FDG-PET Nor CT can distinguish between a pathological complete response and an incomplete response after neoadjuvant chemoradiation in locally advanced rectal cancer: a prospective study. Ann Surg 2013; 258:289.
  205. van der Paardt MP, Zagers MB, Beets-Tan RG, et al. Patients who undergo preoperative chemoradiotherapy for locally advanced rectal cancer restaged by using diagnostic MR imaging: a systematic review and meta-analysis. Radiology 2013; 269:101.
  206. Zhao RS, Wang H, Zhou ZY, et al. Restaging of locally advanced rectal cancer with magnetic resonance imaging and endoluminal ultrasound after preoperative chemoradiotherapy: a systemic review and meta-analysis. Dis Colon Rectum 2014; 57:388.
  207. Hanly AM, Ryan EM, Rogers AC, et al. Multicenter Evaluation of Rectal cancer ReImaging pOst Neoadjuvant (MERRION) Therapy. Ann Surg 2014; 259:723.
  208. Memon S, Lynch AC, Akhurst T, et al. Systematic review of FDG-PET prediction of complete pathological response and survival in rectal cancer. Ann Surg Oncol 2014; 21:3598.
  209. Maffione AM, Marzola MC, Capirci C, et al. Value of (18)F-FDG PET for Predicting Response to Neoadjuvant Therapy in Rectal Cancer: Systematic Review and Meta-Analysis. AJR Am J Roentgenol 2015; 204:1261.
  210. de Jong EA, ten Berge JC, Dwarkasing RS, et al. The accuracy of MRI, endorectal ultrasonography, and computed tomography in predicting the response of locally advanced rectal cancer after preoperative therapy: A metaanalysis. Surgery 2016; 159:688.
  211. Barbaro B, Fiorucci C, Tebala C, et al. Locally advanced rectal cancer: MR imaging in prediction of response after preoperative chemotherapy and radiation therapy. Radiology 2009; 250:730.
  212. Dresen RC, Beets GL, Rutten HJ, et al. Locally advanced rectal cancer: MR imaging for restaging after neoadjuvant radiation therapy with concomitant chemotherapy. Part I. Are we able to predict tumor confined to the rectal wall? Radiology 2009; 252:71.
  213. Jonas J, Bähr R. Neoadjuvant chemoradiation treatment impairs accuracy of MRI staging in rectal carcinoma. Gut 2006; 55:1214.
  214. Joye I, Deroose CM, Vandecaveye V, Haustermans K. The role of diffusion-weighted MRI and (18)F-FDG PET/CT in the prediction of pathologic complete response after radiochemotherapy for rectal cancer: a systematic review. Radiother Oncol 2014; 113:158.
  215. Dworak O, Keilholz L, Hoffmann A. Pathological features of rectal cancer after preoperative radiochemotherapy. Int J Colorectal Dis 1997; 12:19.
  216. Mandard AM, Dalibard F, Mandard JC, et al. Pathologic assessment of tumor regression after preoperative chemoradiotherapy of esophageal carcinoma. Clinicopathologic correlations. Cancer 1994; 73:2680.
  217. Pang X, Xie P, Yu L, et al. A new magnetic resonance imaging tumour response grading scheme for locally advanced rectal cancer. Br J Cancer 2022; 127:268.
  218. Fernández-Martos C, Pericay C, Aparicio J, et al. Phase II, randomized study of concomitant chemoradiotherapy followed by surgery and adjuvant capecitabine plus oxaliplatin (CAPOX) compared with induction CAPOX followed by concomitant chemoradiotherapy and surgery in magnetic resonance imaging-defined, locally advanced rectal cancer: Grupo cancer de recto 3 study. J Clin Oncol 2010; 28:859.
  219. Schrag D, Weiser MR, Goodman KA, et al. Neoadjuvant chemotherapy without routine use of radiation therapy for patients with locally advanced rectal cancer: a pilot trial. J Clin Oncol 2014; 32:513.
  220. Rouanet P, Rullier E, Lelong B, et al. Tailored Strategy for Locally Advanced Rectal Carcinoma (GRECCAR 4): Long-term Results From a Multicenter, Randomized, Open-Label, Phase II Trial. Dis Colon Rectum 2022; 65:986.
  221. Cassidy RJ, Liu Y, Patel K, et al. Can we eliminate neoadjuvant chemoradiotherapy in favor of neoadjuvant multiagent chemotherapy for select stage II/III rectal adenocarcinomas: Analysis of the National Cancer Data base. Cancer 2017; 123:783.
  222. Papke DJ Jr, Yurgelun MB, Noffsinger AE, et al. Prevalence of Mismatch-Repair Deficiency in Rectal Adenocarcinomas. N Engl J Med 2022; 387:1714.
  223. de Rosa N, Rodriguez-Bigas MA, Chang GJ, et al. DNA Mismatch Repair Deficiency in Rectal Cancer: Benchmarking Its Impact on Prognosis, Neoadjuvant Response Prediction, and Clinical Cancer Genetics. J Clin Oncol 2016; 34:3039.
  224. Ludford K, Cohen R, Svrcek M, et al. Pathological Tumor Response Following Immune Checkpoint Blockade for Deficient Mismatch Repair Advanced Colorectal Cancer. J Natl Cancer Inst 2021; 113:208.
  225. Krishnamoorthy M, Lenehan JG, Maleki Vareki S. Neoadjuvant Immunotherapy for High-Risk, Resectable Malignancies: Scientific Rationale and Clinical Challenges. J Natl Cancer Inst 2021; 113:823.
  226. Chalabi M, Fanchi LF, Dijkstra KK, et al. Neoadjuvant immunotherapy leads to pathological responses in MMR-proficient and MMR-deficient early-stage colon cancers. Nat Med 2020; 26:566.
  227. Kothari A, White MG, Peacock O, et al. Pathological response following neoadjuvant immunotherapy in mismatch repair-deficient/microsatellite instability-high locally advanced, non-metastatic colorectal cancer. Br J Surg 2022; 109:489.
  228. Verschoor YL, van den Berg J, Beets G, et al.Neoadjuvant nivolumab, ipilimumab, and celecoxib in MMR-proficient and MMR-deficient colon cancers: Final clinical analysis of the NICHE study (abstract). J Clin Oncol 40, 2022 (suppl 16; abstr 3511) https://meetings.asco.org/2022-asco-annual-meeting/14360?presentation=208373#208373 (Accessed on June 15, 2022).
  229. Wang QX, Xiao BY, Cheng Y, et al. Anti-PD-1-based immunotherapy as curative-intent treatment in dMMR/MSI-H rectal cancer: A multicentre cohort study. Eur J Cancer 2022; 174:176.
  230. https://clinicaltrials.gov/ct2/show/NCT04751370?term=neoadjuvant+immunotherapy&cond=Rectal+Cancer+Stage+III&draw=2&rank=2 (Accessed on March 31, 2021).
  231. Cercek A, Lumish M, Sinopoli J, et al. PD-1 Blockade in Mismatch Repair-Deficient, Locally Advanced Rectal Cancer. N Engl J Med 2022; 386:2363.
  232. Herman J, Messersmith W, Suh WW, et al. ACR Appropriateness Criteria: rectal cancer-metastatic disease at presentation. Curr Probl Cancer 2010; 34:201.
  233. Kim JW, Kim YB, Kim NK, et al. The role of adjuvant pelvic radiotherapy in rectal cancer with synchronous liver metastasis: a retrospective study. Radiat Oncol 2010; 5:75.
  234. Butte JM, Gonen M, Ding P, et al. Patterns of failure in patients with early onset (synchronous) resectable liver metastases from rectal cancer. Cancer 2012; 118:5414.
  235. Shin SJ, Yoon HI, Kim NK, et al. Upfront systemic chemotherapy and preoperative short-course radiotherapy with delayed surgery for locally advanced rectal cancer with distant metastases. Radiat Oncol 2011; 6:99.
  236. Chang CY, Kim HC, Park YS, et al. The effect of postoperative pelvic irradiation after complete resection of metastatic rectal cancer. J Surg Oncol 2012; 105:244.
  237. van Dijk TH, Tamas K, Beukema JC, et al. Evaluation of short-course radiotherapy followed by neoadjuvant bevacizumab, capecitabine, and oxaliplatin and subsequent radical surgical treatment in primary stage IV rectal cancer. Ann Oncol 2013; 24:1762.
  238. Tyc-Szczepaniak D, Wyrwicz L, Kepka L, et al. Palliative radiotherapy and chemotherapy instead of surgery in symptomatic rectal cancer with synchronous unresectable metastases: a phase II study. Ann Oncol 2013; 24:2829.
  239. Crane CH, Janjan NA, Abbruzzese JL, et al. Effective pelvic symptom control using initial chemoradiation without colostomy in metastatic rectal cancer. Int J Radiat Oncol Biol Phys 2001; 49:107.
  240. Chiorean EG, Nandakumar G, Fadelu T, et al. Treatment of Patients With Late-Stage Colorectal Cancer: ASCO Resource-Stratified Guideline. JCO Glob Oncol 2020; 6:414.
Topic 2468 Version 136.0

References