Your activity: 46 p.v.
your limit has been reached. plz Donate us to allow your ip full access, Email: sshnevis@outlook.com

Systemic chemotherapy for metastatic colorectal cancer: General principles

Systemic chemotherapy for metastatic colorectal cancer: General principles
Authors:
Jeffrey W Clark, MD
Hanna K Sanoff, MD, MPH
Section Editor:
Richard M Goldberg, MD
Deputy Editor:
Diane MF Savarese, MD
Literature review current through: Dec 2022. | This topic last updated: Sep 14, 2022.

INTRODUCTION — The majority of patients with metastatic colorectal cancer (mCRC) cannot be cured, although a subset of patients with liver and/or lung-isolated metastatic disease, local recurrence, or limited intra-abdominal disease are potentially curable with surgery. For other patients with mCRC, treatment is palliative and generally consists of systemic chemotherapy. (See "Potentially resectable colorectal cancer liver metastases: Integration of surgery and chemotherapy" and "Surgical resection of pulmonary metastases: Outcomes by histology" and "Locoregional methods for management and palliation in patients who present with stage IV colorectal cancer", section on 'Aggressive cytoreduction and intraperitoneal chemotherapy for peritoneal metastases'.)

This topic review will cover general principles that underlie chemotherapy treatment of mCRC, including the goals of therapy in patients with potentially resectable metastatic disease versus those with categorically unresectable disease, benefits of treatment compared with supportive care alone, issues related to timing and duration of treatment in patients with unresectable metastatic disease, and assessment during therapy. Specific systemic treatment recommendations for patients with unresectable mCRC, issues relevant to treatment of mCRC in the elderly and those with a poor performance status, and the use of systemic therapy for the purpose of downsizing potentially resectable CRC liver metastases are discussed elsewhere. (See "Systemic therapy for nonoperable metastatic colorectal cancer: Selecting the initial therapeutic approach" and "Systemic therapy for nonoperable metastatic colorectal cancer: Approach to later lines of systemic therapy" and "Therapy for metastatic colorectal cancer in older adult patients and those with a poor performance status" and "Potentially resectable colorectal cancer liver metastases: Integration of surgery and chemotherapy", section on 'Patients with initially unresectable metastases'.)

CHEMOTHERAPY OPTIONS — For decades, fluorouracil (FU) was the sole active agent for advanced CRC. This has changed markedly since the year 2000, with the approval of irinotecan; oxaliplatin; three humanized monoclonal antibodies (MoAbs) that target the vascular endothelial growth factor (bevacizumab), the vascular endothelial growth factor receptor (VEGF; ramucirumab), and the epidermal growth factor receptor (cetuximab and panitumumab); intravenous aflibercept, a fully humanized recombinant fusion protein consisting of VEGF binding portions from the human VEGF receptors 1 and 2 fused to the Fc portion of human immunoglobulin G1; regorafenib, an orally active inhibitor of angiogenic (including the VEGF receptors 1 to 3), stromal, and oncogenic kinases; trifluridine-tipiracil (TAS-102), an oral cytotoxic agent that consists of the nucleoside analog trifluridine (a cytotoxic antimetabolite that inhibits thymidylate synthetase and, after modification within tumor cells, is incorporated into deoxyribonucleic acid (DNA) causing strand breaks); and tipiracil, a potent thymidine phosphorylase inhibitor, which inhibits trifluridine metabolism and has antiangiogenic properties as well. In addition, other orally active fluoropyrimidines (capecitabine, S-1, tegafur-uracil [UFT]) are also available.

The best way to combine and sequence these agents is still not established. A compilation of commonly used chemotherapy protocols for CRC is available. (See "Treatment protocols for small and large bowel cancer" and "Systemic therapy for nonoperable metastatic colorectal cancer: Selecting the initial therapeutic approach".)

Predictive biomarkers — Increasingly, biomarker expression is driving therapeutic decision-making in oncology, including in mCRC:

Among patients with mCRC, RAS mutation status permits the selection of individuals who might benefit from strategies targeting the epidermal growth factor receptor (EGFR). Anti-EGFR monoclonal antibodies (cetuximab, panitumumab) should only be prescribed for patients whose tumors are RAS wild-type. Patients with BRAF-mutated tumors are also unlikely to respond to anti-EGFR antibodies alone, but responses are seen in some cases with the addition of a BRAF inhibitor (an approach that is generally used for later lines of therapy). (See 'BRAF' below and "Systemic therapy for nonoperable metastatic colorectal cancer: Approach to later lines of systemic therapy", section on 'RAS wild-type, BRAF mutated tumors'.)

Initial therapy with an immune checkpoint inhibitor should be considered for individuals whose tumors have deficient mismatch repair (dMMR)/high levels of microsatellite instability (MSI-H). (See 'Checkpoint inhibitor immunotherapy and mismatch repair' below.)

Unfortunately, there are as of yet no accepted biologic or molecular markers of responsiveness to bevacizumab, ramucirumab, aflibercept, regorafenib, or cytotoxic agents such as fluoropyrimidines, oxaliplatin, irinotecan, or trifluridine-tipiracil, although these are active areas of research. (See 'Other agents' below.)

The following sections will review the available data on predictive biomarkers for selecting therapy in mCRC.

Agents targeting the EGFR — We restrict the use of anti-EGFR monoclonal antibody therapy to those patients whose tumors lack mutations after extended RAS testing, and in those who lack a BRAF V600E mutation (except when used in combination with a BRAF inhibitor for second- or third-line of therapy).

RAS mutations — Activating mutations in RAS (most commonly KRAS but also NRAS), which result in constitutive activation of the RAS-RAF-ERK pathway, induce resistance to anti-EGFR therapy [1-13]. Activating mutations in KRAS or less commonly NRAS are detected in approximately 40 to 45 percent of mCRCs, with good concordance between the primary and synchronous distant metastases (but not lymph node metastases) [14,15]. The rate of discordant results in RAS mutation assessment when testing primary versus recurrent tumors may be higher (20 percent in one report [16]), and in some settings, rebiopsy of metastases for RAS mutation analysis (or assay of circulating tumor DNA [ctDNA]) may be warranted. (See 'Tissue versus liquid biopsy' below.)

In the United States and elsewhere, panitumumab and cetuximab were originally approved only for patients without detectable RAS mutations [17]. In mCRC, the most common RAS mutations are in KRAS, and mutations are mainly found in exon 2 (codons 12, 13) [18,19]. However, whether all exon 2 KRAS mutations (particularly the G13D mutation) confer resistance to EGFR-targeted agents is unclear; the data, including two different meta-analyses on this subject, are conflicting [20-26].

Extended RAS testing — Wild-type KRAS in exon 2 does not guarantee benefit from agents targeting the EGFR, since even in these cohorts, response rates to either drug are 40 percent or less [7,27,28]. Resistance to anti-EGFR therapies is also mediated by lower-frequency mutations in KRAS outside of exon 2 and in NRAS [9,15,29-34] and exclusion of patients with all RAS mutations identifies a population that is more likely to benefit from an anti-EGFR agent [29,35,36].

As an example, in the Panitumumab Randomized Trial in Combination with Chemotherapy for Metastatic Colorectal Cancer to Determine Efficacy (PRIME) trial, in which 1183 patients with previously untreated mCRC were randomly assigned to FOLFOX (oxaliplatin plus leucovorin [LV] and short-term infusional fluorouracil [FU]) with or without panitumumab, 108 patients (17 percent) without exon 2 KRAS mutations had other mutations in KRAS exons 3 and 4 and in NRAS exons 2, 3, and 4 [29]. These additional mutations predicted a lack of response to panitumumab, and in fact, their presence was associated with inferior progression-free and overall survival in patients receiving panitumumab plus FOLFOX compared with FOLFOX alone.

A year 2015 Provisional Clinical Opinion from the American Society of Clinical Oncology (ASCO) recommended that all patients who are candidates for anti-EGFR therapy should have their tumor tested for mutations in both KRAS and NRAS exons 2 (codons 12 and 13), 3 (codons 59 and 61), and 4 (codons 117 and 146), and if those mutations are found anti-EGFR therapy is not useful [37]. Guidelines from the National Comprehensive Cancer Network (NCCN) also mandate comprehensive testing for mutations in KRAS and NRAS exons 2, 3, and 4 in patients being considered for an anti-EGFR agent [38].

In July 2017, the US Food and Drug Administration approved the PRAXIS Extended RAS Panel, a next-generation sequencing test to detect the presence of 56 specific mutations in KRAS exons 2, 3, and 4 and NRAS exons 2, 3, and 4 in the tumor tissue of patients with mCRC [39].

Tissue versus liquid biopsy — While tumor tissue remains the "gold standard" for genetic analysis in cancer patients, ctDNA can be detected and quantified in the blood of cancer patients and used for detection of tumor-specific genetic alterations, including RAS mutations. The overall concordance between tumor and plasma RAS mutational status (a summation of true positives and true negatives) is 82 to 93 percent in some reports [40-46], although lower rates (64 to 78 percent) are reported by others, especially in certain metastatic sites including the lung and peritoneum [42,43,47]. One meta-analysis of 21 studies evaluating the effectiveness of ctDNA for detection of KRAS mutations concluded that sensitivity and specificity rates were 67 (95% CI 55-78) and 96 (95% CI 95-98) percent, respectively [46].

One advantage of "liquid biopsy" is the potential for reducing data turnaround time [42]. However, only a limited number of cases have been studied, and all of the analyses are retrospective. Thus, in our view, additional data are needed before treatment decisions in mCRC can be made based upon ctDNA. A year 2018 joint review by ASCO and the College of American Pathologists concluded that the evidence shows discordance between the results of ctDNA assays and genotyping tumor specimens, and supports tumor tissue genotyping to confirm results from ctDNA tests [48]. This is an area of active research and is likely to evolve over the coming years.

BRAF — BRAF is a component of the RAS-RAF-MAPK signaling pathway. Activating mutations, which are mutually exclusive with KRAS mutations, are found in approximately 5 to 12 percent of mCRCs. BRAF mutations (most of which are V600E mutations) have consistently been associated with poor prognosis overall [49-55]. (See "Pathology and prognostic determinants of colorectal cancer", section on 'RAS and BRAF'.)

V600E mutations – Evidence increasingly supports the view that response to EGFR-targeted agents (either alone or in combination with chemotherapy) is unlikely in patients whose tumors harbor BRAF V600E mutations, even if they are RAS wild-type [56-58]:

One analysis of 10 randomized trials comparing cetuximab or panitumumab alone or plus chemotherapy with standard therapy or best supportive care included one phase II and nine phase III trials; six were conducted in the first-line treatment setting, two for second-line therapy, and two in patients with chemorefractory disease [56]. Among patients with RAS wild-type/BRAF V600E mutant tumors, compared with control regimens, the addition of an anti-EGFR monoclonal antibody did not significantly improve progression-free survival (hazard ratio [HR] 0.88, 95% CI 0.67-1.14), overall survival (HR 0.91, 95% CI 0.62-1.34), or objective response rate.

A similar conclusion was reached in an individual patient data analysis derived from the ARCAD database of ten randomized trials of first-line targeted therapies [58].

Another analysis included eight randomized trials, four conducted in the first-line setting, three in the second-line setting, and one in patients with chemorefractory disease [57]. Among patients with RAS wild-type/BRAF V600E mutant mCRC, there was no significant overall survival benefit for the addition of an anti-EGFR monoclonal antibodies (HR 0.97, 95% CI 0.67-1.41). By contrast, overall survival was significantly greater in patients with RAS wild-type BRAF wild-type tumors (HR 0.81, 95% CI 0.7-0.95). When comparing the overall survival benefit between BRAF V600E mutant and BRAF wild-type tumors, the test for interaction was not statistically significant, leading the authors to conclude that the observed differences in the effect of anti-EGFR monoclonal antibodies on overall survival according to BRAF V600E mutation status could have been due to chance, and that the evidence was insufficient to state that mutant tumors attain a different treatment benefit from anti-EGFR agents compared with individuals with BRAF wild-type tumors.

Additional information is available from a preliminary report of the prospective randomized FIRE-4.5 trial, in which 108 patients with previously untreated RAS wild-type BRAF V600E-mutated mCRC were randomly assigned to oxaliplatin plus irinotecan, leucovorin, and short-term infusional FU (FOLFOXIRI) plus either bevacizumab or cetuximab [59]. In the intent to treat analysis, the patients receiving cetuximab had a lower objective response rate (the primary endpoint, 40 versus 51 percent), significantly inferior median progression-free survival (PFS; 6 versus 8.3 months), and a trend towards inferior overall survival although the results were not mature.

Given these results, in our view and that of others, the preponderance of the available evidence is that response to EGFR-targeted agents, either as single agents or in combination with chemotherapy, is unlikely in patients whose tumors harbor a BRAF V600E mutation. The American Joint Committee on Cancer (AJCC), in its most recent 2017 tumor, node, metastasis (TNM) staging revision, considers that there is level I evidence to support a lack of effect of anti-EGFR antibody therapy in patients whose tumors harbor a BRAF V600E mutation [60]. Furthermore, consensus-based guidelines from the NCCN and the European Society for Medical Oncology [38,61] both suggest not using cetuximab or panitumumab for patients with BRAF V600E mutated cancers.

Resistance to EGFR-targeted agents in patients who have mutations in BRAF V600E but RAS wild-type disease can be overcome by the addition of BRAF inhibitors. This subject is discussed in more detail elsewhere. (See "Systemic therapy for nonoperable metastatic colorectal cancer: Approach to later lines of systemic therapy", section on 'RAS wild-type, BRAF mutated tumors'.)

Other BRAF mutations – Less is known about BRAF mutations outside of codon 600, which account for about one-fifth of all BRAF mutations in mCRC [62]. From a prognostic standpoint, patients with mCRC whose tumors harbor a non-V600 mutation seem to have a better median overall survival than do those with either a V600E mutation or a BRAF wild-type tumor (61 versus 11 versus 43 months, respectively) [62]. However, there are very few data addressing the predictive value of non-V600 BRAF mutations for response to anti-EGFR agents [63-65], and this remains an area of active investigation. (See "Pathology and prognostic determinants of colorectal cancer", section on 'RAS and BRAF'.)

EGFR amplification — Some [66-68], but not all studies [5,69-71], suggest an association between EGFR copy number and response to anti-EGFR therapy. The clinical use of EGFR amplification to select patients for therapy is limited by the lack of standardization of fluorescence in situ hybridization technology and scoring [67,70].

Other biomarkers — Even tumors that are wild type after extended RAS and BRAF testing do not consistently respond to EGFR-targeted therapies. Several other mechanisms of resistance have been explored [28,31,67,72-84], none of which have been incorporated into clinical practice. It seems likely that a comprehensive biomarker analysis will be required to identify the subgroup of patients with mCRC who will truly benefit from treatment with an anti-EGFR agent [85].

Checkpoint inhibitor immunotherapy and mismatch repair — Approximately 3.5 to 6.5 percent of stage IV CRCs have dMMR enzymes, the biologic footprint of which is MSI-H. Cancers with dMMR/MSI-H appear to be uniquely susceptible to immune checkpoint inhibitors, and this is a reasonable first-line approach for appropriately selected patients. (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 "Tissue-agnostic cancer therapy: DNA mismatch repair deficiency, tumor mutational burden, and response to immune checkpoint blockade in solid tumors", section on 'Biology of mismatch repair and tumor mutational burden'.)

HER2-targeted agents — Approximately 3 to 5 percent of CRCs have amplification of the human epidermal growth factor receptor 2 (HER2) oncogene or overexpress its protein product, HER2. Accumulating data provide proof-of-principle support for the potential benefit from HER2-targeted therapies (eg, trastuzumab plus pertuzumab or lapatinib, fam-trastuzumab deruxtecan) in these patients, and HER2-targeted therapy represents a standard treatment for HER2-overexpressing mCRC after failure of conventional chemotherapy, although the optimal treatment regimen remains uncertain [86]. (See "Systemic therapy for nonoperable metastatic colorectal cancer: Approach to later lines of systemic therapy", section on 'HER2 overexpressors'.)

HER2-targeted therapy may also be considered as a first-line treatment in patients who are not candidates for more intensive therapy; however, it is not our preferred approach. (See "Systemic therapy for nonoperable metastatic colorectal cancer: Selecting the initial therapeutic approach", section on 'Not candidates for intensive therapy'.)

Other agents

Bevacizumab and ramucirumab – Attempts to discover molecular or pathologic predictive factors for bevacizumab (and another antiangiogenic monoclonal antibody [MoAb], ramucirumab) efficacy in order to identify subgroups of patients who gain greater or lesser degrees of benefit from these drugs have not led to clinically useful predictive biomarkers, although this is an active area of research [87-93]. An important issue is that it is not tumor tissue that is the target for these drugs but instead host endothelial cells, which interact with tumor on the microenvironmental level.

Cytotoxic agents – There are no prospectively validated predictive biomarkers for conventional cytotoxic agents, although this is an area of active research [94-100].

Combination versus sequential single agents — The available evidence continues to support initial combination chemotherapy for most patients, particularly for those whose metastases might be potentially resectable after an initial chemotherapy response. However, the main benefits are higher response rate and delayed tumor progression. Given the lack of a demonstrable survival benefit from initial combination chemotherapy, the use of sequential single agents might represent a reasonable alternative for a patient who desires to minimize treatment-related toxicity. (See "Potentially resectable colorectal cancer liver metastases: Integration of surgery and chemotherapy", section on 'Patients with initially unresectable metastases'.)

First-line therapy with combinations of fluoropyrimidines, oxaliplatin, irinotecan, and biologic agents such as bevacizumab has markedly improved response rates, PFS, and survival compared with fluoropyrimidines alone or doublet chemotherapy regimens with or without a biologic agent. However, even in patients treated initially with fluoropyrimidine monotherapy, survival is positively impacted by subsequent lines of therapy, and upfront combination therapy (particularly when oxaliplatin and irinotecan are combined) also increases toxicity and cost. (See "Systemic therapy for nonoperable metastatic colorectal cancer: Selecting the initial therapeutic approach", section on 'Overview of the therapeutic approach'.)

The question of whether patients should receive initial combination therapy or fluoropyrimidine monotherapy has been addressed in two randomized trials, neither of which showed that survival was adversely impacted by initial single-agent therapy [101,102]. However, the median survival for all groups in both trials (which ranged from 13.9 to 17.4 months) was lower than expected for modern combination chemotherapy. (See "Systemic therapy for nonoperable metastatic colorectal cancer: Selecting the initial therapeutic approach", section on 'Overview of the therapeutic approach'.)

One possible reason is the low number of patients who eventually received all three active drugs in both trials. The proportion of patients receiving all active agents correlates strongly with median survival [103,104]. Another issue is that neither trial used bevacizumab or cetuximab as either first-line or salvage therapy. These agents improve PFS, and bevacizumab also improves overall survival when used in the first-line regimen. (See "Systemic therapy for nonoperable metastatic colorectal cancer: Selecting the initial therapeutic approach", section on 'Role of biologics'.)

Some of these issues were addressed in a third trial, the XELAVIRI trial, which randomly assigned 421 patients with untreated metastatic CRC to a fluoropyrimidine plus bevacizumab, followed by the addition of irinotecan at progression, versus initial combined therapy with all three agents [105]. Only 63 percent of patients treated with initial sequential therapy received irinotecan at some point in the course of their treatment, compared with 100 percent in the combination therapy group. Sequential therapy was noninferior to combination therapy for time to failure of strategy (the primary endpoint), and survival was not significantly different (median overall survival 23.5 versus 21.1 months). An unplanned subgroup analysis suggested that initial combination therapy might have particularly benefited patients with wild-type RAS/BRAF tumors.

These trial results and the implications for clinical practice are discussed in detail elsewhere. (See "Systemic therapy for nonoperable metastatic colorectal cancer: Selecting the initial therapeutic approach", section on 'Initial doublet combinations versus sequential single agents'.)

TREATMENT GOALS — The goals of chemotherapy for mCRC differ according to the clinical scenario. For most patients, treatment will be palliative and not curative (a fact that may not be understood by patients [106]), and the treatment goals are to prolong overall survival and maintain quality of life (QOL) for as long as possible.

Potentially resectable disease — However, some patients with stage IV disease (particularly those with liver-limited metastases) can be surgically cured of their disease. Even selected patients with initially unresectable liver metastases may become eligible for resection if the response to chemotherapy is sufficient.

This approach has been termed "conversion therapy" [107] to distinguish it from "neoadjuvant therapy," which applies to preoperative chemotherapy given to patients who present upfront with apparently resectable disease. The key parameter for selecting the specific regimen in this scenario is not survival or improved QOL, but instead, response rate (ie, the ability of the regimen to shrink metastases) [108]. (See "Potentially resectable colorectal cancer liver metastases: Integration of surgery and chemotherapy", section on 'Patients with initially unresectable metastases'.)

Nonresectable disease — The following general principles guide the use of palliative chemotherapy in the setting of nonoperable disease:

In general, for patients without symptomatic disease (ie, the majority of patients), induction of a tumor response is not as important as is delaying tumor progression for as long as possible. In the palliative setting, objective response rate is not the best indicator of treatment benefit (prolonged survival and/or progression-free survival [PFS]) [109-111]. Thus, achieving stable disease as the best response to therapy might be considered a treatment success. (See 'Assessment during therapy' below.)

Patients benefit more from access to all active agents than from a particular treatment sequence of specific regimens used as individual "lines" of therapy. In all large published phase III trials testing various combinations of cytotoxic agents and targeted agents conducted over the last decade, the proportion of patients receiving all active agents has correlated strongly with median survival [103,112]. Although no such analysis has yet been performed after the introduction of biologic agents, it is conceivable that the overall principle of optimizing outcomes through exposure to all active agents is still valid.

Despite these findings, the available evidence suggests that only a minority of American patients with mCRC are exposed to all active agents in the course of their therapy for mCRC [113].

Because of the survival benefit from second- and even third-line chemotherapy, the routine practice of crossover in clinical trials severely limits the ability to detect an overall survival advantage of one treatment regimen over another. Therefore, the actual activity of a new agent or combination regimen is better captured by the endpoint PFS, in particular in the first-line setting. Improvements in PFS correlate with longer survival [114-116] and are not affected by crossover or subsequent therapy.

These concepts can be illustrated by results from the EPIC trial, in which patients failing initial oxaliplatin-based therapy were randomly assigned to irinotecan with or without cetuximab [117]. There were significant differences in PFS, objective response, and disease control rates that favored combined therapy, but no overall survival advantage. This was attributed to the fact that 50 percent of the patients in the irinotecan arm crossed over to cetuximab at progression. (See "Systemic therapy for nonoperable metastatic colorectal cancer: Approach to later lines of systemic therapy", section on 'Patients not initially treated with cetuximab/panitumumab'.)

Endpoints other than PFS (eg, duration of disease control, time to failure of strategy) have been proposed, but none are widely used [118,119].

The model of distinct "lines" of chemotherapy (in which regimens containing non-cross-resistant drugs are each used in succession until disease progression) is being abandoned in incurable metastatic mCRC in favor of a "continuum of care" approach [120]. This implies an individualized treatment strategy that may include phases of maintenance chemotherapy interspersed with more aggressive treatment protocols, as well as reutilization of previously administered chemotherapy agents in combination with other active drugs.

The following sections will emphasize the practical issues that arise when choosing the appropriate treatment strategy for individual patients with inoperable mCRC. Specific recommendations for therapy as well as management of patients with potentially resectable liver metastases are discussed elsewhere. (See "Systemic therapy for nonoperable metastatic colorectal cancer: Selecting the initial therapeutic approach" and "Systemic therapy for nonoperable metastatic colorectal cancer: Approach to later lines of systemic therapy" and "Therapy for metastatic colorectal cancer in older adult patients and those with a poor performance status" and "Potentially resectable colorectal cancer liver metastases: Integration of surgery and chemotherapy".)

CHEMOTHERAPY VERSUS SUPPORTIVE CARE — Systemic fluorouracil (FU)-based chemotherapy produces meaningful improvements in median survival and progression-free survival (PFS) compared with best supportive care (BSC) alone [121-123]. These benefits are most pronounced with regimens containing irinotecan or oxaliplatin in combination with FU. Although no trial has compared these regimens with BSC alone, median survival durations in clinical trials of oxaliplatin- and irinotecan-containing chemotherapy now consistently exceed two years; by contrast, for patients with unresectable mCRC who receive best supportive care (BSC) alone, median survival is approximately five to six months [121-123].

Long-term survival is improving over time with the availability of more active anticancer agents [124-127]. As an example, in a report of pooled data from North Center Cancer Treatment Group (NCCTG) trials conducted in the FU plus leucovorin (LV) era, only 1.1 percent of patients were alive at five years [128]. By contrast, in a report from the phase III FIRE-3 trial (first-line irinotecan with short-term infusional FU plus LV [FOLFIRI] plus either bevacizumab or cetuximab), the five-year survival rate for patients with RAS wild-type tumors treated with FOLFIRI plus cetuximab was approximately 20 percent [129]. Although many of the survival gains are attributable to advances in chemotherapy treatment, more aggressive use of surgical resection of metastatic disease has also contributed [126]. (See "Potentially resectable colorectal cancer liver metastases: Integration of surgery and chemotherapy" and "Surgical resection of pulmonary metastases: Outcomes by histology", section on 'Colorectal cancer'.)

TIMING OF CHEMOTHERAPY — Although the value of early chemotherapy versus treatment deferral until symptoms develop is controversial, we suggest instituting chemotherapy at diagnosis for patients with categorically unresectable mCRC, and when possible, before patients become symptomatic.

Many patients with mCRC are asymptomatic. Data are limited on optimal timing of chemotherapy, and the only randomized trials directly addressing this issue studied older regimens like fluorouracil (FU) and leucovorin (LV):

In an early trial in which 182 patients with asymptomatic mCRC were randomly assigned to initial or deferred chemotherapy with sequential methotrexate, FU, and LV, earlier treatment was associated with improvements in median survival (14 versus 9 months), symptom-free interval, and progression-free survival (PFS) [121].

In a combined analysis of 168 asymptomatic patients who were enrolled in two trials randomly testing early versus delayed FU-based chemotherapy, there was a non-statistically significant two-month benefit in median survival with early treatment (13 versus 11 months) [130].

Whether these results can be extrapolated to patients treated with irinotecan, oxaliplatin, or biologic therapies, especially in the era of modern diagnostic procedures that can detect lower volume metastatic disease, is unclear. Regimens such as these are associated with clear-cut survival benefits, particularly if patients are serially exposed to all active agents. (See "Systemic therapy for nonoperable metastatic colorectal cancer: Selecting the initial therapeutic approach", section on 'Initial doublet combinations versus sequential single agents'.)

The only available data from the era of modern chemotherapy come from a retrospective report of 736 patients with mCRC diagnosed between January 2003 and December 2010 at a single Australian center; 377 (51 percent) received immediate chemotherapy, 167 (23 percent) did not because they were deemed inappropriate for therapy or refused, and 192 (26 percent) adopted a "watch and wait" policy initially, 168 of whom eventually received chemotherapy (at a median of 3.7 months from diagnosis) [131]. Compared with immediate treatment, the fraction of patients in the delayed chemotherapy group who eventually received treatment with all active agents was slightly less (30 versus 39 percent), but the median survival was superior (27 versus 17 months).

Importantly, these data are not from a randomized trial, and interpretation is limited by the potential for selection bias (ie, patients who had treatment deferred were likely to be those with favorable biology [asymptomatic, lower volume metastatic disease, better performance status]), all of which could have contributed to the longer survival in this group. At least in the United States, most patients institute treatment at a time when they are still asymptomatic from their cancer. An alternate approach, which may be particularly appropriate for asymptomatic elderly patients, is an initial period of observation to judge the tempo of disease progression.

CHEMOTHERAPY DOSING IN OBESE PATIENTS — For cancer patients with a large body surface area (BSA), chemotherapy drug doses are often reduced because of concern for excess toxicity. However, there is no evidence that fully dosed obese patients experience greater toxicity from chemotherapy for mCRC; furthermore, obese patients who are given reduced doses may have inferior outcomes [132]. Although limited, the available data do not support the policy of routine dose reduction (or capping the maximal BSA to 2.0 m2) for obese patients with mCRC. Guidelines from the American Society of Clinical Oncology recommend that full weight-based cytotoxic chemotherapy doses be used to treat obese patients with cancer [133]. (See "Dosing of anticancer agents in adults", section on 'Dosing for overweight/obese patients'.)

CONTINUOUS VERSUS INTERMITTENT THERAPY — The optimal duration of initial chemotherapy for unresectable mCRC is controversial. The decision to permit treatment breaks for responding patients must be individualized and based upon the regimen being used, tolerance of and response to chemotherapy, disease bulk and location, symptomatology, and patient preference. For many patients with chemotherapy responsive disease who do not have bulky or severely symptomatic disease, intermittent rather than continuous therapy may mitigate treatment-related toxicity, and does not appear to adversely impact overall survival. For patients initially treated with oxaliplatin, a complete break in therapy represents a valid alternative to fluoropyrimidine-based maintenance chemotherapy without oxaliplatin for patients who have responding or stable disease after the initial course of chemotherapy, particularly for those with a complete clinical response or small-volume metastatic disease.

Rationale for intermittent therapy — When fluorouracil (FU) was the only treatment alternative, patients generally stayed on treatment until their disease progressed or they developed unacceptable toxicity. This typically meant that patients were treated for four to six months (the median progression-free survival [PFS] duration) and then were placed on supportive care alone until they died (median duration of survival approximately one year).

Compared with FU alone, newer combinations are more effective (median survival durations now consistently approach two years), but they are also more toxic. This is particularly true for oxaliplatin-containing regimens, which cause cumulative neurotoxicity; several studies have shown that more patients come off of therapy because of toxic effects than because of progressive disease [134,135]. Intermittent rather than continuous chemotherapy has the potential to improve outcomes and reduce toxicity as well as cost.

However, intermittent therapy may be appropriate for some patients and not others:

There are many patients with small volume but multiple sites of disease who respond well to chemotherapy or have a prolonged period of disease stability. Even if their disease triples in volume off therapy, they will not likely be symptomatic or develop organ dysfunction. Patients with favorable characteristics may be able to tolerate chemotherapy-free (or at least oxaliplatin-free) intervals of multiple months per year and go on to respond favorably to drugs for many years.

On the other end of the spectrum are patients with retained primary tumors, bulky disease, poor performance scores due to tumor related symptoms, peritoneal disease that may lead to unsalvageable bowel obstruction as the first sign of progression, and those with extensive symptomatic disease who progress through treatment regimens in quick succession with either short-lived responses or no response. These patients may be better approached with continuous chemotherapy. (See "Systemic therapy for nonoperable metastatic colorectal cancer: Selecting the initial therapeutic approach", section on 'Duration of initial chemotherapy'.)

Whether continued chemotherapy provides better outcomes than intermittent therapy to best response followed by a chemotherapy "holiday" has been addressed in several trials, most of which have studied chemotherapy regimens that contain oxaliplatin, a drug that is associated with dose-limiting neurotoxicity. Intermittent oxaliplatin-free therapy can be achieved through a complete break in therapy or the use of a non-oxaliplatin-containing "maintenance regimen." (See "Overview of neurologic complications of platinum-based chemotherapy", section on 'Cumulative sensory neuropathy' and "Systemic therapy for nonoperable metastatic colorectal cancer: Selecting the initial therapeutic approach", section on 'FOLFOX versus FOLFIRI'.)

Patients receiving oxaliplatin — Oxaliplatin-based regimens (eg, FOLFOX [oxaliplatin plus LV and short-term infusional FU]) are commonly used for first-line chemotherapy in mCRC [113]. However, oxaliplatin is associated with a cumulative sensory neuropathy, which may be dose limiting.

Whether long-term neurotoxicity can by mitigated by intermittent oxaliplatin-free intervals has been addressed in several trials. The following represents an overview of the most important findings.

Maintenance fluoropyrimidines only

OPTIMOX-1 – The OPTIMOX1 trial randomly assigned 620 previously untreated patients to FOLFOX, administered every two weeks until disease progression (arm A), or FOLFOX (table 1) for six cycles only, followed by reintroduction of oxaliplatin at the time of progression after 12 cycles of a non-oxaliplatin-containing maintenance regimen (leucovorin-modulated FU) [118]. The duration of disease control and overall survival between the continuous (arm A) and maintenance (arm B) approaches were very similar (9 versus 8.7 months, and 19.3 versus 21.2 months, respectively). Individuals in arm B had a significantly lower risk of developing grade 3 or 4 toxicity during cycles 6 to 18 (but not overall) [136].

OPTIMOX-2 – The subsequent OPTIMOX-2 trial was initially designed as a 600 patient phase III trial, but when bevacizumab became available, accrual was halted with 202 patients enrolled [137]. OPTIMOX-2 compared six cycles of modified FOLFOX7 followed by maintenance with 5FU/LV (arm A) to six cycles of modified FOLFOX7 followed by a complete stop in chemotherapy (arm B). The primary endpoint was the duration of disease control, calculated as the sum of the duration of PFS both following the initial three-month course of modified FOLFOX7 (mFOLFOX7) (table 1), as well as after the subsequent reintroduction of oxaliplatin. An important characteristic of OPTIMOX-2 was that randomization occurred after six cycles of therapy regardless of response, and metastases were allowed to progress back to baseline levels before FOLFOX was reintroduced.

Complete discontinuation of therapy seemed to have an adverse impact on prognosis; the group receiving maintenance therapy had significantly longer median duration of disease control and median PFS from the time of randomization; there was also a trend toward improved median overall survival (24 versus 20 months, p = 0.42). These data mandate caution and both careful patient selection and vigilant patient monitoring so that therapy can be reinstated promptly at progression when considering chemotherapy-free intervals.

Another multicenter trial, the CONcePT trial, in which patients were randomly assigned to continuous versus intermittent oxaliplatin (alternating every eight cycles with and without oxaliplatin) also confirmed the benefit of intermittent rather than continuous oxaliplatin for increasing time on first-line therapy for oxaliplatin/bevacizumab-based combinations [138]. Rates of peripheral sensory neuropathy were significantly lower in the intermittent therapy group.

Maintenance bevacizumab — Several trials have explored the benefit of maintenance bevacizumab in patients initially treated with a bevacizumab-containing regimen, both alone and in combination with a fluoropyrimidine.

Bevacizumab plus a fluoropyrimidine

CAIRO3 – The utility of maintenance treatment with capecitabine plus bevacizumab was addressed in the Dutch CAIRO3 trial, which randomly assigned 558 patients with stable disease or better after six cycles of XELOX plus bevacizumab who were not eligible for potentially curative metastasectomy to continued capecitabine (625 mg/m2 twice daily every day) plus bevacizumab (7.5 mg/kg every three weeks) or observation alone [139]. Upon first progression (PFS1), patients in both arms were supposed to be treated with XELOX plus bevacizumab until the second progression (PFS2) per protocol. The primary endpoint was PFS2, which was calculated from the time of randomization. Maintenance therapy was associated with a significantly longer PFS2 (11.7 versus 8.5 months, hazard ratio [HR] 0.67, p<0.0001), and there was a trend toward improved overall survival, as well (median 21.6 versus 18.1 months, HR 0.89, p = 0.22).

German AIO KRK 0207 trial – Similarly, a benefit for continued fluoropyrimidine plus bevacizumab as compared with observation alone was also shown in the German AIO KRK 0207 trial, in which patients without progressive disease after six months of oxaliplatin plus a fluoropyrimidine and bevacizumab were randomly assigned to maintenance with the same fluoropyrimidine plus bevacizumab, bevacizumab alone, or observation only [140]. The primary endpoint was the "time to failure of strategy" or TFS, which included the duration of maintenance plus the time from reinduction after first progression to a second disease progression. The trial was powered to demonstrate noninferiority with a noninferiority margin set at 3.5 months, corresponding to an HR of 1.42. The median TFS in the fluoropyrimidine plus bevacizumab and observations arms was not significantly different (6.9 and 6.4 months, respectively; HR 1.26, 95% CI 0.99-1.60). However, the observation arm was not non-inferior to fluoropyrimidine plus bevacizumab because the upper limit of the 95 percent confidence interval exceeded the threshold set for non-inferiority (1.43). Notably, few patients in either arm were exposed to reinduction treatment (19 percent with combined therapy, and 46 percent of those undergoing observation), rendering the primary endpoint, TFS, non-informative and clinically irrelevant.

STOP and GO trial – A slightly different approach was tested in the Turkish STOP and GO trial, in which, following six cycles of bevacizumab plus XELOX, 123 patients were randomly assigned to continued therapy or discontinuation of oxaliplatin and maintenance with bevacizumab plus capecitabine until progression [141]. The median PFS was significantly better in the group receiving maintenance therapy with bevacizumab plus capecitabine (11 versus 8.3 months), with less grade 3 or 4 diarrhea (3.3 versus 11.3 percent), hand-foot syndrome (1.6 versus 3.2 percent), and neuropathy (1.6 versus 8.1 percent).

Bevacizumab monotherapy – For patients who have no disease progression after an initial course of bevacizumab plus oxaliplatin-containing chemotherapy, we suggest not pursuing bevacizumab alone for maintenance therapy; this approach is also not recommended in consensus-based guidelines for the treatment of mCRC from National Comprehensive Cancer Network [142] and European Society for Medical Oncology [61].

The role of maintenance bevacizumab alone has been studied in three trials, all of which used different comparator arms, and all of which came to different conclusions:

MACRO – In the Spanish MACRO trial, patients received six cycles of first-line XELOX plus bevacizumab followed by a randomization to continued therapy or bevacizumab maintenance therapy alone until progression or treatment intolerance [143]. There was no arm in which patients received no maintenance therapy. The median PFS and overall survival durations in patients treated with maintenance bevacizumab alone were not significantly worse, and rates of severe neurotoxicity, hand-foot syndrome, and fatigue were significantly lower. However, the trial failed to achieve its primary endpoint of non-inferiority for PFS, because the projected upper limit of the 95 percent confidence interval for PFS exceeded the preset limit.

SAKK 41-06 – In the Swiss SAKK 41-06 trial, 262 patients with mCRC were randomly assigned to bevacizumab continuation versus no maintenance after four to six months of first-line bevacizumab-containing chemotherapy (62 percent oxaliplatin-containing, 31 percent irinotecan-containing, and the rest fluoropyrimidine alone) [144]. Like the MACRO trial, the trial failed to achieve its primary endpoint of non-inferiority for TTP with the projected upper limit of the 95 percent confidence interval for TTP exceeding the preset limit. The median TTP was 4.1 for bevacizumab continuation versus 2.9 months for no continuation (HR 0.74, 95% CI 0.57-0.95). However, in our view, this study has significant limitations; it includes trials conducted over almost two decades, contains a very heterogenous patient population, and it is heavily influenced by the COIN trial due to its size. As a result, it should not be used to justify use of bevacizumab alone as effective maintenance therapy.

German AIO KRK 0207 trial – On the other hand, noninferiority of bevacizumab alone compared with bevacizumab plus a fluoropyrimidine was shown in the German AIO KRK 0207 trial, described above [140]. The primary endpoint (the median time to failure of strategy, TFS) in the fluoropyrimidine plus bevacizumab and bevacizumab alone arms was 6.9 and 6.1 months, respectively. Compared with fluoropyrimidine plus bevacizumab, the bevacizumab only arm was non-inferior (HR 1.08, 95% CI 0.85-1.37). However, the upper boundary of the noninferiority margin was very generous (HR 1.43). Notably, few patients in either arm were exposed to reinduction treatment (19 percent with combined therapy, and 43 percent of those receiving bevacizumab alone), rendering the primary endpoint, TFS, non-informative and clinically irrelevant.

Patients initially treated with an EGFR inhibitor — For patients initially treated with an agent targeting the epidermal growth factor receptor (EGFR), we suggest maintenance therapy using fluorouracil plus the anti-EGFR agent rather than an anti-EGFR agent or fluoropyrimidine alone.

Benefit from anti-EGFR therapies is limited to patients whose tumors lack mutations in one of the RAS oncogenes (ie, wild-type RAS). (See 'Agents targeting the EGFR' above.)

Three trials have addressed the benefit of maintenance therapy with an EGFR inhibitor after initial treatment with FOLFOX plus an EGFR inhibitor:

MACRO – The phase II MACRO-2 trial randomly assigned 193 patients with KRAS (exon 2 only) wild-type tumors to receive FOLFOX plus cetuximab for four months (eight courses) followed by either continued therapy with the same regimen or cetuximab monotherapy alone (250 mg/m2 weekly) [145]. Cetuximab monotherapy was noninferior to the combination of continued FOLFOX plus cetuximab, as judged by the primary endpoint, the proportion of patients who were progression free at nine months (60 versus 72 percent, HR 0.60, 95% CI 0.31-1.15).

VALENTINO – On the other hand, results with panitumumab alone were inferior to maintenance treatment with FU/LV plus panitumumab following four months of induction therapy with FOLFOX plus panitumumab in the phase II noninferiority VALENTINO trial [146]. Ten-month PFS was inferior with panitumumab alone (49 versus 60 percent).

PANAMA – A slightly different question, the benefit of adding panitumumab to leucovorin (LV) modulated FU versus FU/LV alone after six cycles of induction therapy with FOLFOX plus panitumumab in RAS wild-type advanced CRC was addressed in the phase III PANAMA trial [147]. Median PFS, the primary endpoint, was significantly better with combined therapy as compared with leucovorin-modulated FU alone (8.8 versus 5.7 months, HR 0.72, 95% CI 0.60-0.85), and there was also a trend to better overall survival that also favored maintenance panitumumab.

Irinotecan — While intermittent treatment approaches appear to be almost mandatory for the majority of patients receiving oxaliplatin because of cumulative neurotoxicity, there are no cumulative dose-dependent toxicities from irinotecan. For most patients, we treat as long as tumor shrinkage continues and treatment is tolerated. Thereafter, as intermittent treatment does not appear to compromise outcomes, treatment breaks could be considered in responding patients, especially those receiving concomitant therapy with an anti-EGFR agent.

The benefits/risks of intermittent chemotherapy with an irinotecan-containing regimen have been addressed in the following reports:

One trial demonstrated that patients started on FOLFIRI (irinotecan with short-term infusional FU plus LV (table 2)) as first-line therapy had similar overall outcome (PFS and overall survival) whether or not the regimen was administered continuously until progression or toxicity or in "two months on/two months off" intervals [148]. The mean chemotherapy-free period in the intermittent treatment group was only three months. However, there were no demonstrable differences in treatment-related toxicity between the continuous versus intermittent treatment groups. Of note, further second- and third-line therapy did not follow a "stop-and-go" approach, so that for overall survival, any potential differences obtained in first-line therapy could have been obscured by subsequent treatment. (See "Systemic therapy for nonoperable metastatic colorectal cancer: Selecting the initial therapeutic approach", section on 'Irinotecan-based regimens'.)

On the other hand, patients treated initially with an irinotecan plus an anti-EGFR agent might benefit from intermittent as compared with continuous therapy. This issue was directly addressed in the prospective IMPROVE trial, in which 137 patients with unresectable, previously untreated RAS/BRAF wild-type mCRC were randomly assigned to FOLFIRI plus panitumumab until disease progression on treatment, or a fixed eight cycles followed by a treatment-free interval, and reintroduction of the same regimen at disease progression [149]. In a preliminary report presented at the 2022 annual American Society of Clinical Oncology meeting, at a median follow-up of 18 months, the overall disease control rate was similar with intermittent as compared with continuous therapy (90 versus 94 percent), and more patients were alive without progression at one year with intermittent therapy (60.8 versus 52.1 percent, median PFS 17.1 versus 13.1 months). The intermittent strategy yielded lower rates of grade ≥3 skin toxicity (13 versus 27 percent), and fewer patients discontinuing therapy for toxicity. The results of this study do have the caveat of a relatively small sample size.

A lack of benefit for maintenance bevacizumab versus no treatment until progression following six months of induction FOLFIRI plus bevacizumab was shown in the randomized phase III PRODIGE 9 trial [150].

Complete break in therapy — The data on maintenance therapy described above have led to the general conclusion that some form of maintenance therapy is preferred rather than a complete break in therapy in patients who are responding to or have stable disease after induction chemotherapy therapy. However, while maintenance therapy prolongs PFS compared with no maintenance therapy, none of the trials described above have shown that this approach is associated with better overall survival compared with a complete break in therapy.

At least two meta-analyses and a more recent trial have specifically addressed the role of observation (ie, a complete break in therapy) versus maintenance treatment in patients initially treated with either oxaliplatin or irinotecan-based initial systemic therapy for mCRC, all of which have concluded that overall survival is not adversely impacted by a complete break in treatment:

A network meta-analysis included 12 randomized trials comparing the different treatment strategies of continued chemotherapy, observation, and maintenance therapy (including fluoropyrimidine alone, bevacizumab alone, or fluoropyrimidine plus bevacizumab) [151]. Different induction regimens were used in the different trials, including an oxaliplatin-based regimen in nine [118,137-141,143,144,152,153], an irinotecan-based regimen in two [150,154], and mixed regimens in one trial [144].

Comparisons of any maintenance therapy versus observation demonstrated that maintenance therapy was associated with improved PFS in both direct (HR 0.63, 95% CI 0.45-0.86) and indirect analyses (HR 0.58, 95% CI 0.43-0.77), but the effect on overall survival was not significant (HR for the indirect analysis 0.91, 95% CI 0.83-1.01). Analyses of each specific maintenance strategy (fluoropyrimidine alone, bevacizumab alone, fluoropyrimidine plus bevacizumab) versus observation alone also found improved PFS but not overall survival for all comparisons.

Similarly, an individual patient data meta-analysis of more than 4000 patients enrolled on nine trials evaluating intermittent therapy after successful completion of induction therapy (six with planned stopping of all therapy, the other three discontinuing oxaliplatin with continuation of the other regimen components as maintenance therapy) also concluded that a complete break in therapy did not adversely impact survival [155]. The overall analysis of intermittent versus continuous therapy showed no significant overall survival detriment from intermittent therapy (HR 1.03, 95% CI 0.93-1.14), whether from complete break (HR 1.04, 95% CI 0.87-1.26) or maintenance (HR 0.99, 95% CI 0.87-1.13). PFS results were broadly consistent with the overall survival results. In a preplanned analysis, thrombocytosis was confirmed as a poor prognostic factor, but it did not predict for inferior survival from a complete treatment break compared with continuous therapy (interaction HR 0.97, 95% CI 0.66-1.40).

Additional data are available from the randomized FOCUS4-N trial, in which 254 patients with stable or responding disease after 16 weeks of induction therapy with a variety of regimens were randomly assigned to a complete break in therapy with active monitoring versus single-agent capecitabine (1250 mg/m2 twice daily on days 1 through 14 of each 21-day cycle), until progression [156]. Maintenance therapy with capecitabine doubled the time to progression and return to full-dose chemotherapy (median PFS 3.88 versus 1.87 months, HR 0.40, 95% CI 0.21-0.75), but had no impact on median overall survival (14.8 versus 15.2 months, adjusted HR 0.93, 95% CI 0.69-1.27). Furthermore, those assigned to maintenance capecitabine had significant higher rates of cumulative toxicity, especially diarrhea, fatigue, nausea, and palmar plantar erythrodysesthesia, although these were primarily low grade.

ASSESSMENT DURING THERAPY — During chemotherapy, response is typically assessed by periodic assay (every one to three months) of serum carcinoembryonic antigen (CEA) levels, if initially elevated, and interval radiographic evaluation (typically every 8 to 12 weeks, or as prompted by a rising CEA level). Although persistently rising CEA levels are highly correlated with disease progression, confirmatory radiologic confirmatory studies should be obtained prior to a change in therapeutic strategy, with the notable exception of confirmed peritoneal carcinomatosis that is not radiographically measurable.

Radiographic response — Radiographic tumor response is usually quantified using Response Evaluation Criteria In Solid Tumors (RECIST) (table 3) [157,158].

Immunotherapy using immune checkpoint inhibitors is increasingly being integrated into the care of patients with mismatch repair-deficient/microsatellite instability-high (dMMR/MSI-H) mCRC. (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'.)

Individuals treated with immune checkpoint inhibitors for dMMR/MSI-H mCRC can have pseudoprogression [159], and objective response criteria specifically developed for these drugs should be used (eg, immune-modified RECIST [imRECIST] (table 4)). (See "Principles of cancer immunotherapy", section on 'Immunotherapy response criteria'.)

Serum tumor markers — If initially elevated, a 50 percent or greater declines in CEA from baseline to first restaging can predict disease nonprogression and correlate with favorable long-term outcomes [160]. On the other hand, persistently rising CEA levels (particularly rapidly rising levels [161]) are highly correlated with disease progression [162,163]. However, confirmatory radiologic studies are generally recommended in both settings, particularly if a change in therapeutic strategy is being considered because of a rising CEA. Caution should be used when interpreting a rising CEA level during the first four to six weeks of a new therapy, since spurious early elevation in serum CEA may occur, especially after oxaliplatin [164-166].

Circulating tumor DNA (ctDNA) is the fraction of circulating DNA that is derived from a patient's cancer. Colorectal cancers shed DNA into the blood, and interest in using ctDNA as a surrogate indicator of treatment response has grown as techniques to detect and quantify such DNA have improved. A meta-analysis of 24 studies on patients with mCRC reporting on the predictive or prognostic value of ctDNA concluded that a small or no early decrease in ctDNA levels during treatment was associated with short progression-free and overall survival, but the majority of included studies had a high risk of bias [167].

Few large prospective validation studies have been performed on ctDNA-based treatment monitoring. At least in advanced breast cancer, there are some data that suggest that ctDNA responses do not always parallel imaging-based responses [168], and no studies convincingly demonstrate improved patient outcomes or any cost savings when compared with standard of care monitoring approaches.

Thus, in our view, there is not yet enough known about the mechanisms controlling ctDNA change and how well radiologic responses or CEA changes and ctDNA markers correlate with each other to understand whether ctDNA can replace or supplement periodic assay of CEA or radiologic assessment, and the clinical utility or serial assay of ctDNA during remains uncertain. This position is consistent with a year 2018 joint review of the utility of ctDNA analysis in patients with cancer by American Society of Clinical Oncology and the College of American Pathologists, which concluded that there is insufficient evidence of clinical validity and utility for the majority of ctDNA assays in advanced cancer [48]. However, this remains an active area of research with a number of ongoing studies that should impact information on the potential future utility of ctDNA analysis in this setting.

SPECIAL CONSIDERATIONS DURING THE COVID-19 PANDEMIC — The COVID-19 pandemic has increased the complexity of cancer care. Important issues in areas where viral transmission rates are high 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 undergoing palliative chemotherapy for stage IV colorectal cancer include establishing goals of care and discussing advance care planning, utilizing oral rather than intravenous therapy, where appropriate, transitioning outpatient care (eg, pump disconnection) to home whenever possible, and using intermittent rather than continuous therapy (with or without maintenance therapy), where feasible. (See 'Continuous versus intermittent therapy' above and "Systemic therapy for nonoperable metastatic colorectal cancer: Selecting the initial therapeutic approach", section on 'Not candidates for intensive therapy'.)

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

General considerations – Systemic chemotherapy produces meaningful improvements in survival that are most pronounced in patients who are exposed to all active drugs. Understanding of how to combine and sequence drugs for metastatic colorectal cancer (mCRC) is evolving. (See 'Chemotherapy versus supportive care' above and 'Chemotherapy options' above.)

Predictive biomarkers – Increasingly, biomarker expression is driving therapeutic decision-making in oncology, including in mCRC. We restrict anti-epidermal growth factor receptor (EGFR) monoclonal antibody therapy to those patients whose tumors lack mutations after extended RAS testing, and in those who lack a BRAF V600E mutation (except when used in combination with a BRAF inhibitor for second- or third-line of therapy). The evidence suggests that these mutations confer resistance. (See 'Predictive biomarkers' above.)

Sequential single agent versus combination therapy – We suggest initial combination chemotherapy rather than sequential single agents for most patients, particularly for those whose metastases might be potentially resectable after an initial chemotherapy response (Grade 2C). Given the lack of a demonstrable survival benefit from multiagent chemotherapy, the use of sequential single agents might represent a reasonable alternative for a patient who desires to minimize treatment-related toxicity. (See 'Combination versus sequential single agents' above.)

Treatment goals – Some patients with stage IV disease can be surgically cured of their disease, and the goal of initial chemotherapy is maximal reduction in tumor burden. For most, treatment is palliative, and the goals are to prolong overall survival and maintain quality of life (QOL) for as long as possible. (See 'Treatment goals' above.)

Timing of therapy – For most patients, we suggest early rather than deferred initiation of chemotherapy, and when possible, before patients become symptomatic (Grade 2C). (See 'Timing of chemotherapy' above.)

Duration of initial therapy – The optimal duration of initial chemotherapy for unresectable mCRC is controversial. The decision to permit treatment breaks for responding patients must be individualized and based upon the regimen being used, tolerance of and response to chemotherapy, disease bulk and location, symptomatology, and patient preference. For many patients with chemotherapy responsive disease who do not have bulky or severely symptomatic disease, intermittent rather than continuous therapy may mitigate treatment-related toxicity, and does not appear to adversely impact overall survival. (See 'Continuous versus intermittent therapy' above.)

Patients receiving oxaliplatin – For most patients who are responding to an oxaliplatin-based initial regimen, we suggest discontinuing oxaliplatin before the onset of severe neurotoxicity (usually after three to four months of therapy) while continuing the other agents in the regimen (Grade 2C). Continuing oxaliplatin is a reasonable alternative for patients who have an ongoing response and no clinically significant neuropathy. (See 'Patients receiving oxaliplatin' above.)

A complete break in therapy is also a valid option, particularly if a complete clinical response is observed or for those with small-volume metastatic disease who have a partial response or stable disease to the initial course of chemotherapy. Decision-making should also consider patient preference. In such cases, close follow-up with tumor assessment at two-month intervals and early resumption of chemotherapy at the first sign of progression is recommended. (See 'Complete break in therapy' above.)

Patients receiving irinotecan – The advantages of intermittent treatment with irinotecan-based regimens are less clear, and for most patients, we continue treatment for as long as tolerability and tumor shrinkage continue. Intermittent treatment is an option for responding patients who desire a break in therapy, particularly for those receiving concomitant therapy with an anti-EGFR agent. (See 'Irinotecan' above.)

Response assessment – Response to chemotherapy is typically assessed by periodic assay of serum carcinoembryonic antigen (CEA) levels, if initially elevated, and interval radiographic evaluation. Although persistently rising CEA levels are highly correlated with disease progression, confirmatory radiologic confirmatory studies should be obtained prior to a change in therapeutic strategy, with the notable exception of confirmed peritoneal carcinomatosis that is not radiographically measurable. (See 'Assessment during therapy' above.)

ACKNOWLEDGMENT — The UpToDate editorial staff acknowledges Axel Grothey, MD, who contributed to an earlier version of this topic review.

  1. de Reyniès A, Boige V, Milano G, et al. KRAS mutation signature in colorectal tumors significantly overlaps with the cetuximab response signature. J Clin Oncol 2008; 26:2228.
  2. Van Cutsem E, Köhne CH, Hitre E, et al. Cetuximab and chemotherapy as initial treatment for metastatic colorectal cancer. N Engl J Med 2009; 360:1408.
  3. Bokemeyer C, Bondarenko I, Makhson A, et al. Fluorouracil, leucovorin, and oxaliplatin with and without cetuximab in the first-line treatment of metastatic colorectal cancer. J Clin Oncol 2009; 27:663.
  4. Karapetis CS, Khambata-Ford S, Jonker DJ, et al. K-ras mutations and benefit from cetuximab in advanced colorectal cancer. N Engl J Med 2008; 359:1757.
  5. Khambata-Ford S, Garrett CR, Meropol NJ, et al. Expression of epiregulin and amphiregulin and K-ras mutation status predict disease control in metastatic colorectal cancer patients treated with cetuximab. J Clin Oncol 2007; 25:3230.
  6. Lièvre A, Bachet JB, Boige V, et al. KRAS mutations as an independent prognostic factor in patients with advanced colorectal cancer treated with cetuximab. J Clin Oncol 2008; 26:374.
  7. Amado RG, Wolf M, Peeters M, et al. Wild-type KRAS is required for panitumumab efficacy in patients with metastatic colorectal cancer. J Clin Oncol 2008; 26:1626.
  8. Di Fiore F, Blanchard F, Charbonnier F, et al. Clinical relevance of KRAS mutation detection in metastatic colorectal cancer treated by Cetuximab plus chemotherapy. Br J Cancer 2007; 96:1166.
  9. Loupakis F, Ruzzo A, Cremolini C, et al. KRAS codon 61, 146 and BRAF mutations predict resistance to cetuximab plus irinotecan in KRAS codon 12 and 13 wild-type metastatic colorectal cancer. Br J Cancer 2009; 101:715.
  10. Richman SD, Seymour MT, Chambers P, et al. KRAS and BRAF mutations in advanced colorectal cancer are associated with poor prognosis but do not preclude benefit from oxaliplatin or irinotecan: results from the MRC FOCUS trial. J Clin Oncol 2009; 27:5931.
  11. Tabernero J, Cervantes A, Rivera F, et al. Pharmacogenomic and pharmacoproteomic studies of cetuximab in metastatic colorectal cancer: biomarker analysis of a phase I dose-escalation study. J Clin Oncol 2010; 28:1181.
  12. Dahabreh IJ, Terasawa T, Castaldi PJ, Trikalinos TA. Systematic review: Anti-epidermal growth factor receptor treatment effect modification by KRAS mutations in advanced colorectal cancer. Ann Intern Med 2011; 154:37.
  13. Tougeron D, Lecomte T, Pagès JC, et al. Effect of low-frequency KRAS mutations on the response to anti-EGFR therapy in metastatic colorectal cancer. Ann Oncol 2013; 24:1267.
  14. Han CB, Li F, Ma JT, Zou HW. Concordant KRAS mutations in primary and metastatic colorectal cancer tissue specimens: a meta-analysis and systematic review. Cancer Invest 2012; 30:741.
  15. Peeters M, Kafatos G, Taylor A, et al. Prevalence of RAS mutations and individual variation patterns among patients with metastatic colorectal cancer: A pooled analysis of randomised controlled trials. Eur J Cancer 2015; 51:1704.
  16. Lee KH, Kim JS, Lee CS, Kim JY. KRAS discordance between primary and recurrent tumors after radical resection of colorectal cancers. J Surg Oncol 2015; 111:1059.
  17. Information on the conditional approval of panitumumab (Vectibix) by the EMEA www.emea.europa.eu/pdfs/human/press/pr/32370307en.pdf (Accessed on April 20, 2011).
  18. The full document from the College of American Pathologists available at www.cap.org/POET (Accessed on April 20, 2011).
  19. Jimeno A, Messersmith WA, Hirsch FR, et al. KRAS mutations and sensitivity to epidermal growth factor receptor inhibitors in colorectal cancer: practical application of patient selection. J Clin Oncol 2009; 27:1130.
  20. Tejpar S, Celik I, Schlichting M, et al. Association of KRAS G13D tumor mutations with outcome in patients with metastatic colorectal cancer treated with first-line chemotherapy with or without cetuximab. J Clin Oncol 2012; 30:3570.
  21. Gajate P, Sastre J, Bando I, et al. Influence of KRAS p.G13D mutation in patients with metastatic colorectal cancer treated with cetuximab. Clin Colorectal Cancer 2012; 11:291.
  22. Peeters M, Douillard JY, Van Cutsem E, et al. Mutant KRAS codon 12 and 13 alleles in patients with metastatic colorectal cancer: assessment as prognostic and predictive biomarkers of response to panitumumab. J Clin Oncol 2013; 31:759.
  23. Mao C, Huang YF, Yang ZY, et al. KRAS p.G13D mutation and codon 12 mutations are not created equal in predicting clinical outcomes of cetuximab in metastatic colorectal cancer: a systematic review and meta-analysis. Cancer 2013; 119:714.
  24. Schirripa M, Loupakis F, Lonardi S, et al. Phase II study of single-agent cetuximab in KRAS G13D mutant metastatic colorectal cancer. Ann Oncol 2015; 26:2503.
  25. Rowland A, Dias MM, Wiese MD, et al. Meta-analysis comparing the efficacy of anti-EGFR monoclonal antibody therapy between KRAS G13D and other KRAS mutant metastatic colorectal cancer tumours. Eur J Cancer 2016; 55:122.
  26. Segelov E, Thavaneswaran S, Waring PM, et al. Response to Cetuximab With or Without Irinotecan in Patients With Refractory Metastatic Colorectal Cancer Harboring the KRAS G13D Mutation: Australasian Gastro-Intestinal Trials Group ICECREAM Study. J Clin Oncol 2016; 34:2258.
  27. Di Nicolantonio F, Martini M, Molinari F, et al. Wild-type BRAF is required for response to panitumumab or cetuximab in metastatic colorectal cancer. J Clin Oncol 2008; 26:5705.
  28. Garm Spindler KL, Pallisgaard N, Rasmussen AA, et al. The importance of KRAS mutations and EGF61A>G polymorphism to the effect of cetuximab and irinotecan in metastatic colorectal cancer. Ann Oncol 2009; 20:879.
  29. Douillard JY, Oliner KS, Siena S, et al. Panitumumab-FOLFOX4 treatment and RAS mutations in colorectal cancer. N Engl J Med 2013; 369:1023.
  30. Fornaro L, Lonardi S, Masi G, et al. FOLFOXIRI in combination with panitumumab as first-line treatment in quadruple wild-type (KRAS, NRAS, HRAS, BRAF) metastatic colorectal cancer patients: a phase II trial by the Gruppo Oncologico Nord Ovest (GONO). Ann Oncol 2013; 24:2062.
  31. De Roock W, Claes B, Bernasconi D, et al. Effects of KRAS, BRAF, NRAS, and PIK3CA mutations on the efficacy of cetuximab plus chemotherapy in chemotherapy-refractory metastatic colorectal cancer: a retrospective consortium analysis. Lancet Oncol 2010; 11:753.
  32. Peeters M, Oliner KS, Price TJ, et al. Analysis of KRAS/NRAS mutations in phase 3 study 20050181 of panitumumab (pmab) plus FOLFIRI versus FOLFIRI for second-line treatment (tx) of metastatic colorectal cancer (mCRC). J Clin Oncol 2014; 32S:ASCO #LBA387.
  33. Schwartzberg LS, Rivera F, Karthaus M, et al. Analysis of KRAS/NRAS mutations in PEAK: A randomized phase II study of FOLFOX6 plus panitumumab (pmab) or bevacizumab (bev) as first-line treatment (tx) for wild‑type (WT) KRAS (exon 2) metastatic colorectal cancer (mCRC). J Clin Oncol 2013; 31S:ASCO #3631.
  34. Stintzing S, Jung A, Rossius L, Modest DP. Analysis of KRAS/NRAS and BRAF mutations in FIRE-3: A randomized phase III study of FOLFIRI plus cetuximab or bevacizumab as first-line treatment for wild-type (WT) KRAS (exon 2) metastatic colorectal cancer (mCRC) patients. Eur J Cancer 2013; 49S:ECC #17.
  35. Sorich MJ, Wiese MD, Rowland A, et al. Extended RAS mutations and anti-EGFR monoclonal antibody survival benefit in metastatic colorectal cancer: a meta-analysis of randomized, controlled trials. Ann Oncol 2015; 26:13.
  36. Peeters M, Oliner KS, Price TJ, et al. Analysis of KRAS/NRAS Mutations in a Phase III Study of Panitumumab with FOLFIRI Compared with FOLFIRI Alone as Second-line Treatment for Metastatic Colorectal Cancer. Clin Cancer Res 2015; 21:5469.
  37. Allegra CJ, Rumble RB, Hamilton SR, et al. Extended RAS Gene Mutation Testing in Metastatic Colorectal Carcinoma to Predict Response to Anti-Epidermal Growth Factor Receptor Monoclonal Antibody Therapy: American Society of Clinical Oncology Provisional Clinical Opinion Update 2015. J Clin Oncol 2016; 34:179.
  38. NCCN Clinical Practice Guidelines in Oncology. Available at: https://www.nccn.org/professionals/physician_gls/default.aspx (Accessed on October 27, 2021).
  39. US Food and Drug Administraton Premarket approval for PRAXIS Extended RAS panel available online at https://www.accessdata.fda.gov/scripts/cdrh/cfdocs/cfPMA/pma.cfm?id=P160038&et_cid=39413534&et_rid=907466112&linkid=https%3a%2f%2fwww.accessdata.fda.gov%2fscripts%2fcdrh%2fcfdocs%2fcfPMA%2fpma.cfm%3fid%3dP160038 (Accessed on July 24, 2017).
  40. Spindler KG, Boysen AK, Pallisgård N, et al. Cell-Free DNA in Metastatic Colorectal Cancer: A Systematic Review and Meta-Analysis. Oncologist 2017; 22:1049.
  41. García-Foncillas J, Alba E, Aranda E, et al. Incorporating BEAMing technology as a liquid biopsy into clinical practice for the management of colorectal cancer patients: an expert taskforce review. Ann Oncol 2017; 28:2943.
  42. Thierry AR, El Messaoudi S, Mollevi C, et al. Clinical utility of circulating DNA analysis for rapid detection of actionable mutations to select metastatic colorectal patients for anti-EGFR treatment. Ann Oncol 2017; 28:2149.
  43. Normanno N, Esposito Abate R, Lambiase M, et al. RAS testing of liquid biopsy correlates with the outcome of metastatic colorectal cancer patients treated with first-line FOLFIRI plus cetuximab in the CAPRI-GOIM trial. Ann Oncol 2018; 29:112.
  44. Grasselli J, Elez E, Caratù G, et al. Concordance of blood- and tumor-based detection of RAS mutations to guide anti-EGFR therapy in metastatic colorectal cancer. Ann Oncol 2017; 28:1294.
  45. Vidal J, Muinelo L, Dalmases A, et al. Plasma ctDNA RAS mutation analysis for the diagnosis and treatment monitoring of metastatic colorectal cancer patients. Ann Oncol 2017; 28:1325.
  46. Hao YX, Fu Q, Guo YY, et al. Effectiveness of circulating tumor DNA for detection of KRAS gene mutations in colorectal cancer patients: a meta-analysis. Onco Targets Ther 2017; 10:945.
  47. Kagawa Y, Elez E, García-Foncillas J, et al. Combined Analysis of Concordance between Liquid and Tumor Tissue Biopsies for RAS Mutations in Colorectal Cancer with a Single Metastasis Site: The METABEAM Study. Clin Cancer Res 2021; 27:2515.
  48. Merker JD, Oxnard GR, Compton C, et al. Circulating Tumor DNA Analysis in Patients With Cancer: American Society of Clinical Oncology and College of American Pathologists Joint Review. J Clin Oncol 2018; 36:1631.
  49. Yuan ZX, Wang XY, Qin QY, et al. The prognostic role of BRAF mutation in metastatic colorectal cancer receiving anti-EGFR monoclonal antibodies: a meta-analysis. PLoS One 2013; 8:e65995.
  50. Lochhead P, Kuchiba A, Imamura Y, et al. Microsatellite instability and BRAF mutation testing in colorectal cancer prognostication. J Natl Cancer Inst 2013; 105:1151.
  51. Van Cutsem E, Köhne CH, Láng I, et al. Cetuximab plus irinotecan, fluorouracil, and leucovorin as first-line treatment for metastatic colorectal cancer: updated analysis of overall survival according to tumor KRAS and BRAF mutation status. J Clin Oncol 2011; 29:2011.
  52. Maughan TS, Adams RA, Smith CG, et al. Addition of cetuximab to oxaliplatin-based first-line combination chemotherapy for treatment of advanced colorectal cancer: results of the randomised phase 3 MRC COIN trial. Lancet 2011; 377:2103.
  53. Tran B, Kopetz S, Tie J, et al. Impact of BRAF mutation and microsatellite instability on the pattern of metastatic spread and prognosis in metastatic colorectal cancer. Cancer 2011; 117:4623.
  54. Venderbosch S, Nagtegaal ID, Maughan TS, et al. Mismatch repair status and BRAF mutation status in metastatic colorectal cancer patients: a pooled analysis of the CAIRO, CAIRO2, COIN, and FOCUS studies. Clin Cancer Res 2014; 20:5322.
  55. Chu JE, Johnson B, Kugathasan L, et al. Population-based Screening for BRAFV600E in Metastatic Colorectal Cancer Reveals Increased Prevalence and Poor Prognosis. Clin Cancer Res 2020; 26:4599.
  56. Pietrantonio F, Petrelli F, Coinu A, et al. Predictive role of BRAF mutations in patients with advanced colorectal cancer receiving cetuximab and panitumumab: a meta-analysis. Eur J Cancer 2015; 51:587.
  57. Rowland A, Dias MM, Wiese MD, et al. Meta-analysis of BRAF mutation as a predictive biomarker of benefit from anti-EGFR monoclonal antibody therapy for RAS wild-type metastatic colorectal cancer. Br J Cancer 2015; 112:1888.
  58. Cohen R, Liu H, Fiskum J, et al. BRAF V600E Mutation in First-Line Metastatic Colorectal Cancer: An Analysis of Individual Patient Data From the ARCAD Database. J Natl Cancer Inst 2021; 113:1386.
  59. Steintzine S, et al. Randomized study to investigate FOLFOXIRI plus either bevacizumab or cetuximab as first-line treatment of BRAF V600E-mutant mCRC: The phase-II FIRE-4.5 study (AIO KRK-0116) (abstract). J Clin Oncol 30, 2021 (suppl 15; abstr 3502). Abstract available online at https://meetinglibrary.asco.org/record/195968/abstract (Accessed on June 09, 2021).
  60. Jessup JM, Goldberg RM, Asare EA, et al. Colon and Rectum. In: AJCC Cancer Staging Manual, 8th ed, Amin MB (Ed), AJCC, Chicago 2017. p.251.
  61. Van Cutsem E, Cervantes A, Adam R, et al. ESMO consensus guidelines for the management of patients with metastatic colorectal cancer. Ann Oncol 2016; 27:1386.
  62. Jones JC, Renfro LA, Al-Shamsi HO, et al. Non-V600 BRAF Mutations Define a Clinically Distinct Molecular Subtype of Metastatic Colorectal Cancer. J Clin Oncol 2017; :JCO2016714394.
  63. Shinozaki E, Yoshino T, Yamazaki K, et al. Clinical significance of BRAF non-V600E mutations on the therapeutic effects of anti-EGFR monoclonal antibody treatment in patients with pretreated metastatic colorectal cancer: the Biomarker Research for anti-EGFR monoclonal Antibodies by Comprehensive Cancer genomics (BREAC) study. Br J Cancer 2017; 117:1450.
  64. Johnson B, Loree JM, Jacome AA, et al. Atypical, Non-V600 BRAF Mutations as a Potential Mechanism of Resistance to EGFR Inhibition in Metastatic Colorectal Cancer. JCO Precis Oncol 2019; 3.
  65. Yaeger R, Kotani D, Mondaca S, et al. Response to Anti-EGFR Therapy in Patients with BRAF non-V600-Mutant Metastatic Colorectal Cancer. Clin Cancer Res 2019; 25:7089.
  66. Moroni M, Veronese S, Benvenuti S, et al. Gene copy number for epidermal growth factor receptor (EGFR) and clinical response to antiEGFR treatment in colorectal cancer: a cohort study. Lancet Oncol 2005; 6:279.
  67. Laurent-Puig P, Cayre A, Manceau G, et al. Analysis of PTEN, BRAF, and EGFR status in determining benefit from cetuximab therapy in wild-type KRAS metastatic colon cancer. J Clin Oncol 2009; 27:5924.
  68. Cappuzzo F, Varella-Garcia M, Finocchiaro G, et al. Primary resistance to cetuximab therapy in EGFR FISH-positive colorectal cancer patients. Br J Cancer 2008; 99:83.
  69. Italiano A, Follana P, Caroli FX, et al. Cetuximab shows activity in colorectal cancer patients with tumors for which FISH analysis does not detect an increase in EGFR gene copy number. Ann Surg Oncol 2008; 15:649.
  70. Personeni N, Fieuws S, Piessevaux H, et al. Clinical usefulness of EGFR gene copy number as a predictive marker in colorectal cancer patients treated with cetuximab: a fluorescent in situ hybridization study. Clin Cancer Res 2008; 14:5869.
  71. Randon G, Yaeger R, Hechtman JF, et al. EGFR Amplification in Metastatic Colorectal Cancer. J Natl Cancer Inst 2021; 113:1561.
  72. Mao C, Yang ZY, Hu XF, et al. PIK3CA exon 20 mutations as a potential biomarker for resistance to anti-EGFR monoclonal antibodies in KRAS wild-type metastatic colorectal cancer: a systematic review and meta-analysis. Ann Oncol 2012; 23:1518.
  73. Tian S, Simon I, Moreno V, et al. A combined oncogenic pathway signature of BRAF, KRAS and PI3KCA mutation improves colorectal cancer classification and cetuximab treatment prediction. Gut 2013; 62:540.
  74. Sartore-Bianchi A, Martini M, Molinari F, et al. PIK3CA mutations in colorectal cancer are associated with clinical resistance to EGFR-targeted monoclonal antibodies. Cancer Res 2009; 69:1851.
  75. Oden-Gangloff A, Di Fiore F, Bibeau F, et al. TP53 mutations predict disease control in metastatic colorectal cancer treated with cetuximab-based chemotherapy. Br J Cancer 2009; 100:1330.
  76. Winder T, Zhang W, Yang D, et al. Germline polymorphisms in genes involved in the IGF1 pathway predict efficacy of cetuximab in wild-type KRAS mCRC patients. Clin Cancer Res 2010; 16:5591.
  77. Huang F, Xu LA, Khambata-Ford S. Correlation between gene expression of IGF-1R pathway markers and cetuximab benefit in metastatic colorectal cancer. Clin Cancer Res 2012; 18:1156.
  78. Ålgars A, Lintunen M, Carpén O, et al. EGFR gene copy number assessment from areas with highest EGFR expression predicts response to anti-EGFR therapy in colorectal cancer. Br J Cancer 2011; 105:255.
  79. Adams RA, James MD, Smith CG, et al. Epidermal growth factor receptor (EGFR) as a predictive and prognostic marker in patients with advanced colorectal cancer (aCRC): The MRC COIN trial experience. J Clin Oncol 2011; 29S:ASCO #359.
  80. Igarashi H, Kurihara H, Mitsuhashi K, et al. Association of MicroRNA-31-5p with Clinical Efficacy of Anti-EGFR Therapy in Patients with Metastatic Colorectal Cancer. Ann Surg Oncol 2015; 22:2640.
  81. Seligmann JF, Elliott F, Richman SD, et al. Combined Epiregulin and Amphiregulin Expression Levels as a Predictive Biomarker for Panitumumab Therapy Benefit or Lack of Benefit in Patients With RAS Wild-Type Advanced Colorectal Cancer. JAMA Oncol 2016.
  82. Bardelli A, Siena S. Molecular mechanisms of resistance to cetuximab and panitumumab in colorectal cancer. J Clin Oncol 2010; 28:1254.
  83. Siena S, Sartore-Bianchi A, Di Nicolantonio F, et al. Biomarkers predicting clinical outcome of epidermal growth factor receptor-targeted therapy in metastatic colorectal cancer. J Natl Cancer Inst 2009; 101:1308.
  84. Dienstmann R, Cervantes A. Heterogeneity of driver genes and therapeutic implications in colorectal cancer. Ann Oncol 2015; 26:1523.
  85. Morano F, Corallo S, Lonardi S, et al. Negative Hyperselection of Patients With RAS and BRAF Wild-Type Metastatic Colorectal Cancer Who Received Panitumumab-Based Maintenance Therapy. J Clin Oncol 2019; 37:3099.
  86. Brown TJ, Massa RC. Challenges in the Management of Patients With HER2-Amplified Colorectal Cancer. JCO Oncol Pract 2022; 18:555.
  87. Lambrechts D, Lenz HJ, de Haas S, et al. Markers of response for the antiangiogenic agent bevacizumab. J Clin Oncol 2013; 31:1219.
  88. Weickhardt AJ, Williams DS, Lee CK, et al. Vascular endothelial growth factor D expression is a potential biomarker of bevacizumab benefit in colorectal cancer. Br J Cancer 2015; 113:37.
  89. Sunakawa Y, Stintzing S, Cao S, et al. Variations in genes regulating tumor-associated macrophages (TAMs) to predict outcomes of bevacizumab-based treatment in patients with metastatic colorectal cancer: results from TRIBE and FIRE3 trials. Ann Oncol 2015; 26:2450.
  90. Bais C, Mueller B, Brady MF, et al. Tumor Microvessel Density as a Potential Predictive Marker for Bevacizumab Benefit: GOG-0218 Biomarker Analyses. J Natl Cancer Inst 2017; 109.
  91. Tabernero J, Hozak RR, Yoshino T, et al. Analysis of angiogenesis biomarkers for ramucirumab efficacy in patients with metastatic colorectal cancer from RAISE, a global, randomized, double-blind, phase III study. Ann Oncol 2018; 29:602.
  92. van Dijk E, Biesma HD, Cordes M, et al. Loss of Chromosome 18q11.2-q12.1 Is Predictive for Survival in Patients With Metastatic Colorectal Cancer Treated With Bevacizumab. J Clin Oncol 2018; 36:2052.
  93. Nixon AB, Sibley AB, Liu Y, et al. Plasma Protein Biomarkers in Advanced or Metastatic Colorectal Cancer Patients Receiving Chemotherapy With Bevacizumab or Cetuximab: Results from CALGB 80405 (Alliance). Clin Cancer Res 2022; 28:2779.
  94. Braun MS, Richman SD, Quirke P, et al. Predictive biomarkers of chemotherapy efficacy in colorectal cancer: results from the UK MRC FOCUS trial. J Clin Oncol 2008; 26:2690.
  95. Dopeso H, Mateo-Lozano S, Elez E, et al. Aprataxin tumor levels predict response of colorectal cancer patients to irinotecan-based treatment. Clin Cancer Res 2010; 16:2375.
  96. Boige V, Mendiboure J, Pignon JP, et al. Pharmacogenetic assessment of toxicity and outcome in patients with metastatic colorectal cancer treated with LV5FU2, FOLFOX, and FOLFIRI: FFCD 2000-05. J Clin Oncol 2010; 28:2556.
  97. Ebert MP, Tänzer M, Balluff B, et al. TFAP2E-DKK4 and chemoresistance in colorectal cancer. N Engl J Med 2012; 366:44.
  98. Lv H, Li Q, Qiu W, et al. Genetic polymorphism of XRCC1 correlated with response to oxaliplatin-based chemotherapy in advanced colorectal cancer. Cancer Invest 2013; 31:24.
  99. Moutinho C, Martinez-Cardús A, Santos C, et al. Epigenetic inactivation of the BRCA1 interactor SRBC and resistance to oxaliplatin in colorectal cancer. J Natl Cancer Inst 2014; 106:djt322.
  100. Abraham JP, Magee D, Cremolini C, et al. Clinical Validation of a Machine-learning-derived Signature Predictive of Outcomes from First-line Oxaliplatin-based Chemotherapy in Advanced Colorectal Cancer. Clin Cancer Res 2021; 27:1174.
  101. Koopman M, Antonini NF, Douma J, et al. Sequential versus combination chemotherapy with capecitabine, irinotecan, and oxaliplatin in advanced colorectal cancer (CAIRO): a phase III randomised controlled trial. Lancet 2007; 370:135.
  102. Seymour MT, Maughan TS, Ledermann JA, et al. Different strategies of sequential and combination chemotherapy for patients with poor prognosis advanced colorectal cancer (MRC FOCUS): a randomised controlled trial. Lancet 2007; 370:143.
  103. Grothey A, Sargent D, Goldberg RM, Schmoll HJ. Survival of patients with advanced colorectal cancer improves with the availability of fluorouracil-leucovorin, irinotecan, and oxaliplatin in the course of treatment. J Clin Oncol 2004; 22:1209.
  104. Varol U, Cirak Y, Cakar B, et al. Survival analysis of metastatic colorectal cancer patients who were treated with the five major therapeutic agents over the course of disease. J BUON 2013; 18:647.
  105. Modest DP, Fischer von Weikersthal L, Decker T, et al. Sequential Versus Combination Therapy of Metastatic Colorectal Cancer Using Fluoropyrimidines, Irinotecan, and Bevacizumab: A Randomized, Controlled Study-XELAVIRI (AIO KRK0110). J Clin Oncol 2019; 37:22.
  106. Weeks JC, Catalano PJ, Cronin A, et al. Patients' expectations about effects of chemotherapy for advanced cancer. N Engl J Med 2012; 367:1616.
  107. Petrelli NJ. Plenary program discussion. 43rd Annual Meeting of the American Society of Clinical Oncology, Chicago, IL, June 4, 2007.
  108. Folprecht G, Grothey A, Alberts S, et al. Neoadjuvant treatment of unresectable colorectal liver metastases: correlation between tumour response and resection rates. Ann Oncol 2005; 16:1311.
  109. Grothey A, Hedrick EE, Mass RD, et al. Response-independent survival benefit in metastatic colorectal cancer: a comparative analysis of N9741 and AVF2107. J Clin Oncol 2008; 26:183.
  110. Siena S, Peeters M, Van Cutsem E, et al. Association of progression-free survival with patient-reported outcomes and survival: results from a randomised phase 3 trial of panitumumab. Br J Cancer 2007; 97:1469.
  111. Giessen C, Laubender RP, Ankerst DP, et al. Progression-free survival as a surrogate endpoint for median overall survival in metastatic colorectal cancer: literature-based analysis from 50 randomized first-line trials. Clin Cancer Res 2013; 19:225.
  112. Grothey A, Sargent D. Overall survival of patients with advanced colorectal cancer correlates with availability of fluorouracil, irinotecan, and oxaliplatin regardless of whether doublet or single-agent therapy is used first line. J Clin Oncol 2005; 23:9441.
  113. Abrams TA, Meyer G, Schrag D, et al. Chemotherapy usage patterns in a US-wide cohort of patients with metastatic colorectal cancer. J Natl Cancer Inst 2014; 106:djt371.
  114. Tang PA, Bentzen SM, Chen EX, Siu LL. Surrogate end points for median overall survival in metastatic colorectal cancer: literature-based analysis from 39 randomized controlled trials of first-line chemotherapy. J Clin Oncol 2007; 25:4562.
  115. Buyse M, Burzykowski T, Carroll K, et al. Progression-free survival is a surrogate for survival in advanced colorectal cancer. J Clin Oncol 2007; 25:5218.
  116. Shi Q, de Gramont A, Grothey A, et al. Individual patient data analysis of progression-free survival versus overall survival as a first-line end point for metastatic colorectal cancer in modern randomized trials: findings from the analysis and research in cancers of the digestive system database. J Clin Oncol 2015; 33:22.
  117. Eng C, Maurel J, Scheithauer W, et al. Impact on quality of life of adding cetuximab to irinotecan in patients who have failed prior oxaliplatin-based therapy: The EPIC trial. J Clin Oncol 2007; 25S: ASCO #4003.
  118. Tournigand C, Cervantes A, Figer A, et al. OPTIMOX1: a randomized study of FOLFOX4 or FOLFOX7 with oxaliplatin in a stop-and-Go fashion in advanced colorectal cancer--a GERCOR study. J Clin Oncol 2006; 24:394.
  119. Allegra C, Blanke C, Buyse M, et al. End points in advanced colon cancer clinical trials: a review and proposal. J Clin Oncol 2007; 25:3572.
  120. Goldberg RM, Rothenberg ML, Van Cutsem E, et al. The continuum of care: a paradigm for the management of metastatic colorectal cancer. Oncologist 2007; 12:38.
  121. Nordic Gastrointestinal Tumor Adjuvant Therapy Group. Expectancy or primary chemotherapy in patients with advanced asymptomatic colorectal cancer: a randomized trial. J Clin Oncol 1992; 10:904.
  122. Scheithauer W, Rosen H, Kornek GV, et al. Randomised comparison of combination chemotherapy plus supportive care with supportive care alone in patients with metastatic colorectal cancer. BMJ 1993; 306:752.
  123. Simmonds PC. Palliative chemotherapy for advanced colorectal cancer: systematic review and meta-analysis. Colorectal Cancer Collaborative Group. BMJ 2000; 321:531.
  124. Sanoff HK, Sargent DJ, Campbell ME, et al. Five-year data and prognostic factor analysis of oxaliplatin and irinotecan combinations for advanced colorectal cancer: N9741. J Clin Oncol 2008; 26:5721.
  125. Renouf DJ, Lim HJ, Speers C, et al. Survival for metastatic colorectal cancer in the bevacizumab era: a population-based analysis. Clin Colorectal Cancer 2011; 10:97.
  126. Jawed I, Wilkerson J, Prasad V, et al. Colorectal Cancer Survival Gains and Novel Treatment Regimens: A Systematic Review and Analysis. JAMA Oncol 2015; 1:787.
  127. Shen C, Tannenbaum D, Horn R, et al. Overall Survival in Phase 3 Clinical Trials and the Surveillance, Epidemiology, and End Results Database in Patients With Metastatic Colorectal Cancer, 1986-2016: A Systematic Review. JAMA Netw Open 2022; 5:e2213588.
  128. Dy GK, Hobday TJ, Nelson G, et al. Long-term survivors of metastatic colorectal cancer treated with systemic chemotherapy alone: a North Central Cancer Treatment Group review of 3811 patients, N0144. Clin Colorectal Cancer 2009; 8:88.
  129. Heinemann V, von Weikersthal LF, Decker T, et al. FOLFIRI plus cetuximab versus FOLFIRI plus bevacizumab as first-line treatment for patients with metastatic colorectal cancer (FIRE-3): a randomised, open-label, phase 3 trial. Lancet Oncol 2014; 15:1065.
  130. Ackland SP, Jones M, Tu D, et al. A meta-analysis of two randomised trials of early chemotherapy in asymptomatic metastatic colorectal cancer. Br J Cancer 2005; 93:1236.
  131. Voskoboynik M, Bae S, Ananda S, et al. An initial watch and wait approach is a valid strategy for selected patients with newly diagnosed metastatic colorectal cancer. Ann Oncol 2012; 23:2633.
  132. Chambers P, Daniels SH, Thompson LC, Stephens RJ. Chemotherapy dose reductions in obese patients with colorectal cancer. Ann Oncol 2012; 23:748.
  133. Griggs JJ, Bohlke K, Balaban EP, et al. Appropriate Systemic Therapy Dosing for Obese Adult Patients With Cancer: ASCO Guideline Update. J Clin Oncol 2021; 39:2037.
  134. Saltz LB, Clarke S, Díaz-Rubio E, et al. Bevacizumab in combination with oxaliplatin-based chemotherapy as first-line therapy in metastatic colorectal cancer: a randomized phase III study. J Clin Oncol 2008; 26:2013.
  135. Goldberg RM, Sargent DJ, Morton RF, et al. A randomized controlled trial of fluorouracil plus leucovorin, irinotecan, and oxaliplatin combinations in patients with previously untreated metastatic colorectal cancer. J Clin Oncol 2004; 22:23.
  136. de Gramont A, Buyse M, Abrahantes JC, et al. Reintroduction of oxaliplatin is associated with improved survival in advanced colorectal cancer. J Clin Oncol 2007; 25:3224.
  137. Chibaudel B, Maindrault-Goebel F, Lledo G, et al. Can chemotherapy be discontinued in unresectable metastatic colorectal cancer? The GERCOR OPTIMOX2 Study. J Clin Oncol 2009; 27:5727.
  138. Hochster HS, Grothey A, Hart L, et al. Improved time to treatment failure with an intermittent oxaliplatin strategy: results of CONcePT. Ann Oncol 2014; 25:1172.
  139. Simkens LH, van Tinteren H, May A, et al. Maintenance treatment with capecitabine and bevacizumab in metastatic colorectal cancer (CAIRO3): a phase 3 randomised controlled trial of the Dutch Colorectal Cancer Group. Lancet 2015; 385:1843.
  140. Hegewisch-Becker S, Graeven U, Lerchenmüller CA, et al. Maintenance strategies after first-line oxaliplatin plus fluoropyrimidine plus bevacizumab for patients with metastatic colorectal cancer (AIO 0207): a randomised, non-inferiority, open-label, phase 3 trial. Lancet Oncol 2015; 16:1355.
  141. Yalcin S, Uslu R, Dane F, et al. Bevacizumab + capecitabine as maintenance therapy after initial bevacizumab + XELOX treatment in previously untreated patients with metastatic colorectal cancer: phase III 'Stop and Go' study results--a Turkish Oncology Group Trial. Oncology 2013; 85:328.
  142. 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).
  143. Díaz-Rubio E, Gómez-España A, Massutí B, et al. First-line XELOX plus bevacizumab followed by XELOX plus bevacizumab or single-agent bevacizumab as maintenance therapy in patients with metastatic colorectal cancer: the phase III MACRO TTD study. Oncologist 2012; 17:15.
  144. Koeberle D, Betticher DC, von Moos R, et al. Bevacizumab continuation versus no continuation after first-line chemotherapy plus bevacizumab in patients with metastatic colorectal cancer: a randomized phase III non-inferiority trial (SAKK 41/06). Ann Oncol 2015; 26:709.
  145. Aranda E, García-Alfonso P, Benavides M, et al. First-line mFOLFOX plus cetuximab followed by mFOLFOX plus cetuximab or single-agent cetuximab as maintenance therapy in patients with metastatic colorectal cancer: Phase II randomised MACRO2 TTD study. Eur J Cancer 2018; 101:263.
  146. Pietrantonio F, Morano F, Corallo S, et al. Maintenance Therapy With Panitumumab Alone vs Panitumumab Plus Fluorouracil-Leucovorin in Patients With RAS Wild-Type Metastatic Colorectal Cancer: A Phase 2 Randomized Clinical Trial. JAMA Oncol 2019; 5:1268.
  147. Modest DP, Karthaus M, Fruehauf S, et al. Panitumumab Plus Fluorouracil and Folinic Acid Versus Fluorouracil and Folinic Acid Alone as Maintenance Therapy in RAS Wild-Type Metastatic Colorectal Cancer: The Randomized PANAMA Trial (AIO KRK 0212). J Clin Oncol 2022; 40:72.
  148. Labianca R, Sobrero A, Isa L, et al. Intermittent versus continuous chemotherapy in advanced colorectal cancer: a randomised 'GISCAD' trial. Ann Oncol 2011; 22:1236.
  149. Avallone A, Giuliani F, Nasto F, et al. Randomized intermittent or continuous panitumumab plus FOLFIRI (FOLFIRI/PANI) for first-line treatment of patients (pts) with RAS/BRAF wild-type (wt) metastatic colorectal cancer (mCRC): The IMPROVE study (abstract). Abstract available online at https://meetings.asco.org/2022-asco-annual-meeting/14359?presentation=208329#208329 (Accessed on June 09, 2022).
  150. Aparicio T, Ghiringhelli F, Boige V, et al. Bevacizumab Maintenance Versus No Maintenance During Chemotherapy-Free Intervals in Metastatic Colorectal Cancer: A Randomized Phase III Trial (PRODIGE 9). J Clin Oncol 2018; 36:674.
  151. Sonbol MB, Mountjoy LJ, Firwana B, et al. The Role of Maintenance Strategies in Metastatic Colorectal Cancer: A Systematic Review and Network Meta-analysis of Randomized Clinical Trials. JAMA Oncol 2020; 6:e194489.
  152. Adams RA, Meade AM, Seymour MT, et al. Intermittent versus continuous oxaliplatin and fluoropyrimidine combination chemotherapy for first-line treatment of advanced colorectal cancer: results of the randomised phase 3 MRC COIN trial. Lancet Oncol 2011; 12:642.
  153. Luo HY, Li YH, Wang W, et al. Single-agent capecitabine as maintenance therapy after induction of XELOX (or FOLFOX) in first-line treatment of metastatic colorectal cancer: randomized clinical trial of efficacy and safety. Ann Oncol 2016; 27:1074.
  154. Berry SR, Cosby R, Asmis T, et al. Continuous versus intermittent chemotherapy strategies in metastatic colorectal cancer: a systematic review and meta-analysis. Ann Oncol 2015; 26:477.
  155. Adams R, Goey K, Chibaudel B, et al. Treatment breaks in first line treatment of advanced colorectal cancer: An individual patient data meta-analysis. Cancer Treat Rev 2021; 99:102226.
  156. Adams RA, Fisher DJ, Graham J, et al. Capecitabine Versus Active Monitoring in Stable or Responding Metastatic Colorectal Cancer After 16 Weeks of First-Line Therapy: Results of the Randomized FOCUS4-N Trial. J Clin Oncol 2021; 39:3693.
  157. Eisenhauer EA, Therasse P, Bogaerts J, et al. New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1). Eur J Cancer 2009; 45:228.
  158. Therasse P, Arbuck SG, Eisenhauer EA, et al. New guidelines to evaluate the response to treatment in solid tumors. European Organization for Research and Treatment of Cancer, National Cancer Institute of the United States, National Cancer Institute of Canada. J Natl Cancer Inst 2000; 92:205.
  159. Colle R, Radzik A, Cohen R, et al. Pseudoprogression in patients treated with immune checkpoint inhibitors for microsatellite instability-high/mismatch repair-deficient metastatic colorectal cancer. Eur J Cancer 2021; 144:9.
  160. Gulhati P, Yin J, Pederson L, et al. Threshold Change in CEA as a Predictor of Non-Progression to First-Line Systemic Therapy in Metastatic Colorectal Cancer Patients With Elevated CEA. J Natl Cancer Inst 2020; 112:1127.
  161. Iwanicki-Caron I, Di Fiore F, Roque I, et al. Usefulness of the serum carcinoembryonic antigen kinetic for chemotherapy monitoring in patients with unresectable metastasis of colorectal cancer. J Clin Oncol 2008; 26:3681.
  162. Shani A, O'Connell MJ, Moertel CG, et al. Serial plasma carcinoembryonic antigen measurements in the management of metastatic colorectal carcinoma. Ann Intern Med 1978; 88:627.
  163. Trillet-Lenoir V, Chapuis F, Touzet S, et al. Any clinical benefit from the use of oncofoetal markers in the management of chemotherapy for patients with metastatic colorectal carcinomas? Clin Oncol (R Coll Radiol) 2004; 16:196.
  164. Sørbye H, Dahl O. Carcinoembryonic antigen surge in metastatic colorectal cancer patients responding to oxaliplatin combination chemotherapy: implications for tumor marker monitoring and guidelines. J Clin Oncol 2003; 21:4466.
  165. Ailawadhi S, Sunga A, Rajput A, et al. Chemotherapy-induced carcinoembryonic antigen surge in patients with metastatic colorectal cancer. Oncology 2006; 70:49.
  166. Strimpakos AS, Cunningham D, Mikropoulos C, et al. The impact of carcinoembryonic antigen flare in patients with advanced colorectal cancer receiving first-line chemotherapy. Ann Oncol 2010; 21:1013.
  167. Callesen LB, Hamfjord J, Boysen AK, et al. Circulating tumour DNA and its clinical utility in predicting treatment response or survival in patients with metastatic colorectal cancer: a systematic review and meta-analysis. Br J Cancer 2022; 127:500.
  168. García-Saenz JA, Ayllón P, Laig M, et al. Tumor burden monitoring using cell-free tumor DNA could be limited by tumor heterogeneity in advanced breast cancer and should be evaluated together with radiographic imaging. BMC Cancer 2017; 17:210.
Topic 15802 Version 65.0

References