Purpose:

The RAS/RAF/MEK/ERK signaling pathway is critical to the development of colorectal cancers, and KRAS, NRAS, and BRAF mutations foster resistance to radiation. We performed a phase I trial to determine the safety of trametinib, a potent MEK1/2 inhibitor, with 5-fluorouracil (5-FU) chemoradiation therapy (CRT) in patients with locally advanced rectal cancer (LARC).

Patients and Methods:

Patients with stage II/III rectal cancer were enrolled on a phase I study with 3+3 study design, with an expansion cohort of 9 patients at the MTD. Following a 5-day trametinib lead-in, with pre- and posttreatment tumor biopsies, patients received trametinib and CRT, surgery, and adjuvant chemotherapy. Trametinib was given orally daily at 3 dose levels: 0.5 mg, 1 mg, and 2 mg. CRT consisted of infusional 5-FU 225 mg/m2/day and radiation dose of 28 daily fractions of 1.8 Gy (total 50.4 Gy). The primary endpoint was to identify the MTD and recommended phase II dose. IHC staining for phosphorylated ERK (pERK) and genomic profiling was performed on the tumor samples.

Results:

Patients were enrolled to all dose levels, and 18 patients were evaluable for toxicities and responses. Treatment was well tolerated, and there was one dose-limiting toxicity of diarrhea, which was attributed to CRT rather than trametinib. At the 2 mg dose level, 25% had pathologic complete response. IHC staining confirmed dose-dependent decrease in pERK with increasing trametinib doses.

Conclusions:

The combination of trametinib with 5-FU CRT is safe and well tolerated, and may warrant additional study in a phase II trial, perhaps in a RAS/RAF-mutant selected population.

This article is featured in Highlights of This Issue, p. 3071

Translational Relevance

Colorectal cancers harboring KRAS-, NRAS-, or BRAF-activating mutations are known to have worse prognoses, and preclinical studies have shown these mutations to promote resistance to radiation. For stage II/III rectal cancers, standard of care includes trimodality treatment with 5-FU–based chemoradiation, 5-FU and oxaliplatin chemotherapy, and surgery. In this phase I study, we report the safety of combining trametinib, a potent and selective MEK1/2 inhibitor, at the dose of 2 mg daily with 5-FU chemoradiation in patients with locally advanced rectal cancer (LARC). We also demonstrate pharmacodynamic studies showing a dose-dependent decrease in phosphorylated ERK in the tumor tissue, and preliminary antitumor activity. A phase II study may be warranted to determine the efficacy of trametinib and chemoradiotherapy in patients with rectal tumors that harbor KRAS, NRAS, and BRAF mutations, which are indicative of a more aggressive pathology.

Colorectal cancer is one of the most common malignancies in the Western world, and the second leading cause of cancer-related deaths in the United States. In 2018, there were an estimated 43,030 new cases of rectal cancer in the United States, and the majority have locally advanced disease (1). Although surgical total mesorectal excision (TME) is the basis of curative-intent treatment for locally advanced rectal cancer, patients with clinically staged T3/T4 tumors or positive nodal stage also undergo preoperative chemoradiation therapy (CRT) to decrease pelvic recurrence rates and improve tumor downstaging prior to surgery. Phase III trials of neoadjuvant 5-fluorouracil (5-FU) CRT have demonstrated better local-regional control with 5-year local recurrence rates of 6%–9% and 5-year overall survival rates of 65%–75% (2–4). Despite numerous phase II/III trials that have added potentially synergistic agents (e.g., cetuximab and oxaliplatin) to the 5-FU CRT backbone, none have shown additional survival benefit so far (5–7).

The RAS/RAF/MEK/ERK signaling pathway is a critical signal transduction pathway that contributes to tumor cell proliferation, invasion, migration, and inhibition of apoptosis (8, 9). Activation of the MAPK pathway is initiated by ligand binding to a receptor tyrosine kinase such as EGFR, leading to KRAS activation, and subsequent activation of RAF isoforms. RAF isoforms then activate MEK1/2, ultimately leading to ERK1/2 activation and nuclear translocation. KRAS-, NRAS-, and BRAF-activating mutations have been well described in colorectal cancer, and occur at a frequency of about 35%–45%, 2%–5%, and 8%–10%, respectively (10–15). These mutations are mutually exclusive, which may indicate that they play an important role in the MAPK pathway. Thus, inhibition of proteins that are downstream of KRAS, BRAF, and NRAS, such as MEK1/2, are attractive targets in the treatment of RAS/RAF-mutant colorectal cancer.

Interestingly, ionizing radiation causes activation of the RAS-MAPK pathway in a dose-dependent fashion, and this activation is blunted by MEK inhibition (16, 17). RAS-activating mutations promote resistance to radiation in tumor cells in preclinical and clinical studies, including colorectal cancer and non–small cell lung cancer (18–26). Preclinical studies demonstrate that genetic downregulation of KRAS or chemical inhibition of KRAS with farnesyltransferase inhibitors resensitizes cells to radiation (27–29). More recently, MEK inhibitors have been shown to effectively radiosensitize RAS-mutant tumor cells in vitro and in vivo, in many preclinical studies, including colorectal cancer (30–34). These studies have elicited potential mechanisms of radiosensitization including altered DNA repair, in part, through DNA double-strand break (DSB) repair pathways, and prevention of a HIF1-mediated hypoxic response (30, 32, 34). Preclinical studies have also established the efficacy of combining MEK inhibition with 5-FU, with or without radiation both in cell lines and tumor xenografts (31, 35).

Trametinib (GSK1120212, MEKinist) is a potent and selective inhibitor of mitogen-activated extracellular signal regulated kinase 1 (MEK1) and MEK2, and is approved for the treatment of BRAF-mutant melanoma (36). Trametinib has been shown to effectively radiosensitize RAS-mutant tumor cells, and has also been shown to have anti-proliferative properties in vitro and in vivo when added to human colon cancer cell lines that harbor KRAS or BRAF mutations (34, 35). Given the substantial preclinical data supporting MEK inhibition as a radiosensitizing strategy for the treatment of colorectal cancer, a cancer enriched with RAS/RAF-activating mutations, we conducted a phase I trial to test the safety and tolerability of trametinib in combination with 5-FU–based CRT prior to surgery for locally advanced rectal cancer (LARC).

Participants and eligibility

Patients with histologically confirmed, locally advanced, not otherwise pretreated stage II or III rectal adenocarcinoma, cT3/4 and/or cN+, M0 (AJCC 7th edition) were recruited to two centers (The Ohio State University, Columbus, OH and Vanderbilt University, Nashville, TN). Investigators obtained informed written consent from each participant or participant's guardian prior to enrollment. The research was conducted in accordance with the recognized ethical guidelines of the Declaration of Helsinki, CIOMS, Belmont report, and U.S. Common Rule. Clinical study was performed after the approval by an institutional review board. Patients were aged ≥18 years and had adequate hepatic, bone marrow, and renal organ function and an Eastern Cooperative Oncology Group (ECOG) performance status of 0 or 1. Initially, the trial was open only to patients with a KRAS gene mutation (codon 12, 13, 61), NRAS mutation (codon 12, 13, 61), or BRAF gene mutation, but due to slow accrual and the nature of the phase I trial to determine the recommended phase II dose of trametinib, it was opened to all KRAS, NRAS, and BRAF wild-type patients as well (beginning with patient #6).

Procedures and screening

Before the amendment to allow for KRAS/NRAS/BRAF wild-type patients, DNA was isolated from primary tumor samples before treatment and tested for KRAS, NRAS, and BRAF mutational status in the local institutional CLIA-certified pathology laboratory by Sanger sequencing. Baseline CT of the thorax/abdomen and pelvis was performed, and all patients underwent either endoscopic rectal ultrasound and/or MRI of the pelvis for adequate staging. Adequate archival pretreatment (diagnostic) tumor tissue was required for correlative studies; if not available, a repeat tumor biopsy was performed prior to study therapy.

Study design

The primary objective of the trial was to determine the maximally tolerated dose of trametinib in combination with 5-FU and radiation in patients with locally advanced rectal cancer. Patients were recruited in a cohort-of-3 trial design (see below for statistical design). Secondary objectives included evaluation of tolerability and safety of the combination, evaluation of pathologic response and clinical outcomes (local control, disease-free survival, and overall survival), evaluation of pharmacodynamics effects of trametinib on RAS/MAPK signaling pathways in tumor biopsies, and performance of tumor mutational profiling to correlate with clinical response.

Trametinib was given orally for a 5-day lead-in period daily Monday–Friday starting at day –7 and for the duration of radiotherapy (days 1–38). Supplementary Figs. S1 and S2 show the flowchart and schema, respectively. Three dose levels of trametinib were tested in escalated fashion: 0.5 mg (dose level 1), 1.0 mg (dose level 2), and 2 mg (dose level 3; Table 1). During trametinib monotherapy, research biopsies of tumor and normal tissue for correlative studies were obtained on day –4 or day –3. All patients received a total radiation dose of 45 Gy applied in 25 fractions of 1.8 Gy over 5 weeks using conventional three-dimensional conformal radiation technique or intensity-modulated radiotherapy, followed by a cone-down boost of 5.4 Gy in 1.8 Gy fractions to the primary tumor and sacral hollow. Infusional 5-FU was delivered as 225 mg/m2/day over 120 hours (Monday–Friday) during days of radiotherapy (days 1–38). An expansion cohort for an additional 6 patients treated at the MTD was completed to provide a total of 12 patients treated at the MTD. Surgery, specifically total mesorectal excision, was recommended to occur after a 6- to 10-week rest period after completion of chemoradiotherapy. Surgical samples were assessed by a board-certified gastrointestinal pathologist and examined for response, including tumor regression grade (score 0–3) based on the modified Ryan scheme (37, 38). Postoperative chemotherapy was given at the discretion of the treating physician but was encouraged in patients with pN+ disease and to begin between 4–12 weeks following surgical resection.

Table 1.

Dose levels/cohorts.

Dose levelTrametinib (mg) Monday–Friday5-FU (mg/m2/day over 120 hr, Monday–Friday)Total radiation dose in 28 fractions (Gy)
−1 0.5 200 50.4 
0.5 225 50.4 
225 50.4 
2a 1.5 225 50.4 
225 50.4 
Dose levelTrametinib (mg) Monday–Friday5-FU (mg/m2/day over 120 hr, Monday–Friday)Total radiation dose in 28 fractions (Gy)
−1 0.5 200 50.4 
0.5 225 50.4 
225 50.4 
2a 1.5 225 50.4 
225 50.4 

Toxicities and statistical design

This is a phase I study designed to determine the MTD level of trametinib when given in combination with 5-FU and radiotherapy in patients with LARC, using the standard cohort-of-3 phase I clinical trial design. Dose-limiting toxicities (DLT) were defined according to CTCAE v.4.0 as any grade 4 neutropenia, any grade 3–5 thrombocytopenia, or any nonhematologic toxicity of grade 3–5 attributable to the three therapies (trametinib, 5-FU, and radiotherapy) that resulted in 7 days or greater interruption in 5-FU and radiotherapy. The DLT observation period (i.e., the period during which DLTs observed was used to inform dose escalation decisions) was 8.5 weeks in this trial (during trametinib monotherapy, chemoradiation, and up to 3 weeks after). The MTD was defined as the maximum dose level of trametinib where at most 1 of 6 patients experience a DLT according to the following rules. If no DLTs were observed at dose level 3 and the MTD level may indeed be at higher doses, dose level 3 would be considered the MTD. If dose level 2 was deemed safe and dose level 3 was found to exceed the MTD (i.e., 2+ patients with an observed DLT), then the intermediate dose level 2a would be evaluated (see Table 1). Once the MTD was determined, an additional 6 patients (for a total of 12 patients at the MTD) were accrued.

IHC

Sections (4–6 μm thick) of formalin-fixed paraffin-embedded (FFPE) tissue were sectioned, placed on plus slides, and baked at 65°C for 1 hour. Immunostaining was performed on the fully automated Bond RX autostaining system (Leica Biosystems). Briefly, heat-induced antigen retrieval was done using Epitope Retrieval Solution 2 (Leica Biosystems) for 20 minutes, and slides were stained with a rabbit mAb to phosphorylated-p44/42 MAPK (Erk1/2; Thr202/Tyr204; clone D13.14.4E, catalog no. 4370, Cell Signaling Technology) at a 1:150 dilution for 30 minutes. The Bond Polymer Refine (DAB) detection system (Leica Biosystems) was used. Slides were evaluated for degree of phosphorylated ERK (pERK) staining in cells by acquiring multispectral brightfield images from each tissue section using the automated Vectra 3.0 automated quantitative pathology imaging system. A board-certified gastrointestinal pathologist (W. Chen) then identified and labeled both the tumor and stromal tissue contained in each image. Using inForm Cell Analysis software, manual tissue segmentation was performed for each image accordingly to separately analyze tumor and stromal tissue. Once tissue segmentation was complete, tumor and stromal nuclear segmentations were performed separately by using the unmixed hematoxylin image to find each nucleus. Cytoplasmic segmentation was then performed. To separate cytoplasm from the nuclei, only pixels that contained a significant ocular density (OD) for DAB, but not hematoxylin, were counted in the cytoplasmic segmentation. For each image, the average DAB per cell of tumor tissue was calculated by extracting the total ocular density (OD) within each tissue segment divided by the tissue cell count. Cells at the edge of either the image or tissue segment were excluded to obtain more accurate whole-cell readings. A minimum of 1,000 tumor cells were quantified per patient.

Targeted next-generation sequencing

DNA was extracted from FFPE tissue scrolls using the Maxwell 16 FFPE DNA purification kit (Promega, Inc) and quantitated using pico green on a Qubit (Life Technologies). Libraries were generated using the ThruPLEX DNA-seq kit (Rubicon Genomics) and custom-targeted DNA probes from Integrated DNA Technologies (IDT). Briefly, 200 ng of FFPE-derived gDNA was sheared using a Covaris instrument, and samples were end-repaired, adaptor and index-ligated, and PCR-amplified. Following the PCR, 500 ng of the prepared DNA libraries was hybridized to the probes overnight. Hybridized probes were captured on magnetic beads, washed, and PCR-amplified. The target-enriched indexed libraries were pooled and underwent paired-end 150 bp sequencing on an Illumina HiSeq4000 to an average depth of 281.2.

Alignment was performed with bowtie2 v2.2.6 using local option to human reference genome hg19 (39). Alignment was converted to bam with Samtools v1.3.1 (40). After adding read groups, marking duplicates, and sorting with picard 2.4.1 (http://broadinstitute.github.io/picard), GATK v3.6 (41) was used to realign around indels. Mpileup format was generated with Samtools v1.3.1 requiring quality > 1 and variants were called with Varscan v2.4.1 (42). Variants were annotated with Variant Effect Predictor from Ensembl (43) and filtered for location (excluding noncoding variants), coding impact (excluding LOW impact variants—those unlikely to change protein behavior), allele frequency in public databases [excluding common variants found in 1000g (44) and ExAC (45)], and known variants [excluding variants listed in NCBI dbSNP (46) but not in COSMIC databases]. Variants were visually inspected in IGV (44).

Patients and toxicities

Nineteen patients were enrolled in dose levels 1 (cohort 1, 0.5 mg), 2 (cohort 2, 1.0 mg), and 3 (cohort 3, 2.0 mg) between August 2013 and May 2017. One patient was deemed unevaluable due to suspected dihydropyrimidine dehydrogenase (DPD) deficiency. Baseline patient characteristics of the 18 evaluable patients are shown in Table 2. Median follow-up for all patients was 37.1 months. Three patients each were enrolled on dose levels 1 and 2, and no DLTs were encountered. Only 1 DLT was observed in patient #12 in dose level 3 [diarrhea (grade 3)] which was attributed to 5-FU chemoradiation. The maximum tolerated dose of trametinib was determined to be 2.0 mg daily (dose level 3), and a total of 12 patients were enrolled at this dose, including the expansion cohort. For all dose levels, no grade 4 or 5 toxicities were observed. The most common adverse event observed that was attributed to trametinib was a maculopapular rash (11 grade 1, 2 grade 2, and 2 grade 3; Table 3). This maculopapular rash led to trametinib discontinuation after 2 weeks of therapy in 1 patient, and trametinib being held for the last 3 days of chemoradiation for 2 other patients. The other common side effects observed were diarrhea, rectal pain, nausea, oral mucositis, and fatigue, which are commonly encountered with 5-FU chemoradiation.

Table 2.

Patient characteristics, staging, and surgical outcomes.

PatientAge (years at enrollment)GenderECOG at enrollmentTrametinib dose (mg)Clinical stagePathologic stageDownstaging?TRGMutation status for KRAS, NRAS, BRAFType of surgery
1. 58 0.5 T4N1 T4N1c No KRAS G13D Abdominoperineal resection 
2. 48 0.5 T3N1 T2N0 Yes KRAS G12V Abdominoperineal resection 
3. 59 0.5 T3N1 T1N0 Yes KRAS G12V Low anterior resection 
4. 54 T3N1 T2N0 Yes KRAS G12V Abdominoperineal resection 
5. 52 T3N0 T3N0 No KRAS G12V Low anterior resection 
6. 58 T3N2 T3N0 Yes WT Low anterior resection 
7. 38 T3N0 CR Yes KRAS G12A Abdominoperineal resection 
8. 55 T3N1 T2N0 Yes BRAF D594N Low anterior resection 
9. 64 T3N0 T3N1 No KRAS G12V Low anterior resection 
10. 52 T3N0 CR Yes WT Low anterior resection 
11. 34 T3N0 CR Yes WT Low anterior resection 
12. 65 T3N1 T3N2b No WT Low anterior resection 
13. 49 T4N2 T1N0 Yes WT Low anterior resection 
14. 74 T3N2b T3N0 Yes WT Low anterior resection 
15. 48 T3N0 T2N1 Yes WT Low anterior resection 
16. 50 T3N1 T1N0 Yes WT Low anterior resection 
17. 47 T3N1 T1N0 Yes WT Abdominoperineal resection 
18. 66 T4N1 T3N0 Yes WT Low anterior resection 
PatientAge (years at enrollment)GenderECOG at enrollmentTrametinib dose (mg)Clinical stagePathologic stageDownstaging?TRGMutation status for KRAS, NRAS, BRAFType of surgery
1. 58 0.5 T4N1 T4N1c No KRAS G13D Abdominoperineal resection 
2. 48 0.5 T3N1 T2N0 Yes KRAS G12V Abdominoperineal resection 
3. 59 0.5 T3N1 T1N0 Yes KRAS G12V Low anterior resection 
4. 54 T3N1 T2N0 Yes KRAS G12V Abdominoperineal resection 
5. 52 T3N0 T3N0 No KRAS G12V Low anterior resection 
6. 58 T3N2 T3N0 Yes WT Low anterior resection 
7. 38 T3N0 CR Yes KRAS G12A Abdominoperineal resection 
8. 55 T3N1 T2N0 Yes BRAF D594N Low anterior resection 
9. 64 T3N0 T3N1 No KRAS G12V Low anterior resection 
10. 52 T3N0 CR Yes WT Low anterior resection 
11. 34 T3N0 CR Yes WT Low anterior resection 
12. 65 T3N1 T3N2b No WT Low anterior resection 
13. 49 T4N2 T1N0 Yes WT Low anterior resection 
14. 74 T3N2b T3N0 Yes WT Low anterior resection 
15. 48 T3N0 T2N1 Yes WT Low anterior resection 
16. 50 T3N1 T1N0 Yes WT Low anterior resection 
17. 47 T3N1 T1N0 Yes WT Abdominoperineal resection 
18. 66 T4N1 T3N0 Yes WT Low anterior resection 

Note: Clinical/pathological staging is AJCC 7th edition.

Abbreviations: ECOG, Eastern Cooperative Oncology Group performance score; TRG, tumor regression grade; WT, wild-type.

Table 3.

Toxicities for all patients.

ToxicityGrade 1Grade 2Grade 3
Maculopapular rash 11 
Diarrhea 10 
Rectal pain 
Nausea 
Oral mucositis 
Fatigue 
Pruritus 
Abdominal cramps 
Anorexia 
Proctitis 
Hand-foot syndrome 
Lymphopenia 12 
Anemia 
Leucopenia 
Thrombocytopenia 
ToxicityGrade 1Grade 2Grade 3
Maculopapular rash 11 
Diarrhea 10 
Rectal pain 
Nausea 
Oral mucositis 
Fatigue 
Pruritus 
Abdominal cramps 
Anorexia 
Proctitis 
Hand-foot syndrome 
Lymphopenia 12 
Anemia 
Leucopenia 
Thrombocytopenia 

Surgical toxicities and postoperative chemotherapy

Six surgeons operated on this clinical trial, and the majority of patients had a temporary ostomy at the time of the initial TME (13 of 18 patients). All patients successfully completed TME (5 abdominoperineal resections and 13 low anterior resections) within the timeframe recommended per the clinical trial, and all patients had R0 resections. Overall, patients healed well after surgery. Patient #1 had T4 involvement requiring cystoprostatectomy during abdominoperineal resection (APR), with flap reconstruction. He had an area of poor healing in the flap requiring wound debridement within 4 weeks of initial surgery, which ultimately healed, but he was recommended not to have postoperative chemotherapy. Patient #2 developed a surgical site infection and distal colostomy ischemia within 2 weeks of surgery that was managed expectantly, and this patient went on to receive postoperative chemotherapy. Patient #4 had delayed healing of a perineal wound which eventually healed, but he opted against receiving postoperative chemotherapy. Patients #10, 13, and 14 were readmitted within 30 days of surgery for dehydration, nausea, and vomiting which resolved with medical management. Patient #11 returned on postoperative day 12 for wound infection which ultimately healed, and the patient did receive postoperative chemotherapy. Patient #18 developed an anastomotic leak (coloanal) and abscess within 2 weeks of low anterior resection (LAR) requiring takedown of coloanal anastomosis and end colostomy. This patient had a prolonged hospitalization and ultimately did not receive postoperative chemotherapy. Overall, 5 patients did not start postoperative chemotherapy either due to surgical complications or patient refusal. Of note, only one out of the 5 patients who did not receive adjuvant therapy had node-positive disease identified on pathologic surgical specimen evaluation.

Efficacy and outcomes

Analysis of the whole patient cohort revealed tumor downstaging in 77.8% (14/18) of patients, with pathological complete response seen in 16.7% (3/18) patients. Four patients did not have tumor downstaging from therapy, one of whom experienced a DLT (diarrhea, patient #12), leading to an interrupted and suboptimal chemoradiation course. In this case, tumor upstaging was actually observed (from cN1 to pN2b). At the MTD and recommended phase II dose (2 mg dose level), 75% (9/12) of patients had tumor downstaging, 25% (3/12) had pathologic complete response (pCR), and an additional 25% (3/12) patients had near pCR (only microscopic foci of residual tumor cells). This led to a 50% (6/12) pCR or near-pCR rate at the recommended phase II dose. In terms of disease control outcomes, only 1 patient developed a local (pelvic) failure which occurred in combination with pulmonary metastasis. Another patient developed isolated pulmonary metastasis. One patient died at 62 months after enrollment, but had no signs of recurrence, and death was attributed to noncancer-related causes. Median local recurrence-free survival, median disease-free survival, and overall survival were not reached (Fig. 1AC). Actuarial local-recurrence free survival, disease-free survival, and cancer-specific survival are high at 94.4%, 88.9%, and 100%.

Figure 1.

Kaplan–Meier curves for local (pelvic) recurrence-free survival (A), disease-free survival (B), and overall survival (C). Of the 2 patients with longest follow-up, one patient died at 62 months from noncancer-related causes and had no signs of recurrence prior to death.

Figure 1.

Kaplan–Meier curves for local (pelvic) recurrence-free survival (A), disease-free survival (B), and overall survival (C). Of the 2 patients with longest follow-up, one patient died at 62 months from noncancer-related causes and had no signs of recurrence prior to death.

Close modal

Correlation of pERK with dose level and response

Quantitative immunohistochemistry (qIHC) was performed on matched tumor specimens from pretreatment and day −4/−3 tumor tissue obtained during trametinib monotherapy. For patients with high quality and sufficient tissue available (2 each from dose levels 1 and 2, and 11 from dose level 3), qIHC revealed a dose-dependent decrease in pERK compared with pretreatment pERK levels with increasing dose levels of trametinib (Fig. 2). Maximal decrease in pERK was observed in patients receiving 2 mg daily in cohort 3 which was significantly different compared with pretreatment levels (P = 0.006), confirming the pharmacodynamic inhibitory effects of trametinib on activated ERK signaling within tumor cells. We also assessed the levels of pretreatment pERK in tumor and interestingly found a strong correlation with pathologic response. Higher pERK levels at baseline (pretherapy) were associated with higher tumor regression grade score (TRG, P = 0.0007), indicating that higher pERK levels appear to be correlated with poor response to neoadjuvant therapy (Supplementary Fig. S3). However, there was only a trend toward higher pERK levels at baseline being associated with RAS/RAF mutations (P = 0.203; Supplementary Fig. S4). In addition, a higher degree of reduction in pERK levels between pretreatment and after trametinib monotherapy did not correlate significantly with RAS/RAF mutation status or TRG score, but did show trends for an association with pCR in the whole cohort (P = 0.204) and 2 mg cohort (P = 0.078; Supplementary Fig. S5).

Figure 2.

Quantitative IHC on pretreatment and posttreatment tumor biopsies. Top left, representative photomicrographs of pERK1/2 IHC from pretreatment biopsy and research tumor biopsies done after 4 or 5 days of trametinib monotherapy. Both patient #7 and #8 were in cohort 3 (2.0 mg trametinib daily dose cohort). Note the considerable decrease in DAB (brown) staining. Nuclei counterstained with hematoxylin. Top right, quantitation of average optical density of DAB staining per cell comparing pretreatment to posttreatment (day −4/−3). Note dose-dependent increase in pERK staining reduction with increasing dose of trametinib in cohorts 1–3. Bottom, average optical density of DAB staining/cell in each cohort from pretreatment to 4 or 5 days of trametinib monotherapy. Note the significant decrease in cohort 3 (P = 0.006).

Figure 2.

Quantitative IHC on pretreatment and posttreatment tumor biopsies. Top left, representative photomicrographs of pERK1/2 IHC from pretreatment biopsy and research tumor biopsies done after 4 or 5 days of trametinib monotherapy. Both patient #7 and #8 were in cohort 3 (2.0 mg trametinib daily dose cohort). Note the considerable decrease in DAB (brown) staining. Nuclei counterstained with hematoxylin. Top right, quantitation of average optical density of DAB staining per cell comparing pretreatment to posttreatment (day −4/−3). Note dose-dependent increase in pERK staining reduction with increasing dose of trametinib in cohorts 1–3. Bottom, average optical density of DAB staining/cell in each cohort from pretreatment to 4 or 5 days of trametinib monotherapy. Note the significant decrease in cohort 3 (P = 0.006).

Close modal

Targeted next-generation sequencing

We performed targeted next-generation sequencing on DNA isolated from tumor research biopsies using an in-house custom panel of 279 cancer-associated genes (46). Mutations were found in 94.4% (17/18) of patients (Supplementary Table S1). We found 38.9% (7/18), 5.6% (1/18), and 0% of patients' tumors had KRAS, BRAF, and NRAS mutations, respectively. TP53 mutations were the most common, occurring in 77.7% (14/18). The next most common mutation was APC (61.1%), followed by lower frequency mutations in SMAD4 (16.7%), mTOR (11.1%), PIK3CA (11.1%), ATM (11.1%), and FBXW7 (11.1%). One patient (#11) was found to have microsatellite instability, indicative of mismatch repair defect and Lynch syndrome, which was confirmed by IHC studies of MLH1, PMS2, MSH2, and MSH6 as well as germline genetic testing. Of the 7 patients (#6, 7, 10, 11, 14, 15, and 16) who had an excellent response to neoadjuvant therapy with pCR or near pCR, one patient had a KRAS mutation (14.2%), 86% had TP53 mutations, and 57.1% had APC mutations, but there did not appear to be other frequently occurring mutations in this group of patients that could account for their excellent response. Of note, KRAS and BRAF mutations appeared to be enriched (63.7%) in the 11 patients who did not have a significant response to neoadjuvant therapy. No statistically significant relationship between the presence of KRAS, BRAF, APC, or TP53 mutations and tumor regression grade were identified, although individuals with a KRAS or BRAFmutation in their tumor had a trend towards a worse tumor regression grade (P = 0.076).

In this phase I trial of MEK1/2 inhibitor trametinib in combination with standard neoadjuvant chemoradiation for LARC, we found that the MTD of trametinib was 2.0 mg daily. This dose was chosen, due to the FDA-approved dose of trametinib 2 mg daily in melanoma and lung cancer, and due to concern of toxicities at higher doses when given in combination with 5-FU chemoradiation. Overall, treatment was relatively well tolerated, with no grade 4–5 toxicities observed and manageable and reversible grade 1–3 toxicities seen. Only one DLT was observed which was attributed to the standard therapy of chemotherapy and radiation. Furthermore, it did not appear that the addition of trametinib increased postoperative complications. With regards to efficacy, 12 patients were treated at the MTD and recommended phase II dose of 2.0 mg. In this group (cohort 3), we observed a relatively high rate of pCR of 25%, along with an additional 25% of patients with near pCR. Given the historical pCR rate seen for patients with neoadjuvant 5-FU chemoradiation alone is expected to be in the range of 10%–15%, these results may be worthy of further exploration, perhaps in a RAS/RAF-mutated patient population.

The landmark AIO-94 German rectal trial established the role of neoadjuvant 5-FU–based chemoradiation in stage II/III rectal cancers (4). The pCR was 8% in this study. NSABP R-03 showed that neoadjuvant therapy attained pCR in 15% of the patients (47). Recent strategies to improve neoadjuvant therapy response have included the concept of total neoadjuvant therapy (TNT), in which systemic chemotherapy is delivered in the neoadjuvant setting rather than the adjuvant setting (48). The rationale for TNT is that approximately 30%–40% of patients do not receive their adjuvant therapy due to postoperative healing problems or complications. Thus, the benefits from the TNT approach are to ensure that patients do receive their intended systemic chemotherapy. In addition, TNT allows for maximal downstaging with both chemotherapy and chemoradiation modalities prior to surgery.

Another strategy is to personalize treatment based on molecular knowledge of the pathways driving therapeutic resistance, such as targeting specific tumor genotypes. Although this strategy has proven to be successful in targeting stage IV disease in many cancers (e.g., EGFR-mutant or ALK fusion–positive NSCLC), tumor genotype has not yet driven specific radiotherapy treatments in solid tumors. Despite this, there is growing data on how genotype may inform radiotherapy response, particularly KRAS genotype. In early-stage NSCLC, localized KRAS-mutant lung tumors that were treated with stereotactic body radiotherapy (SBRT) had lower local control, when compared to KRAS wild-type tumors (21). A prospective study with SBRT for metastases to the liver from colorectal, gastroesophageal, and pancreatic cancers reported that the presence of a KRAS mutation was predictive of poor local control and radioresistance. One year local control for KRAS-mutant tumors was 20% versus 69.2% for KRAS wild-type tumors (P = 0.001; ref. 24). In a study specifically examining neoadjuvant therapy in patients with rectal cancer, there was a markedly lower pCR noted in the KRAS-mutant patient population (13 vs. 33%; P = 0.006; ref. 26). Taken together, there is growing clinical evidence that the presence of a KRAS mutation is associated with radioresistance and that these patients have the greatest need for an agent targeting the RAS/RAF pathway.

In our phase I study, most of the patients in the final dose cohort of 2 mg did not harbor KRAS/NRAS/BRAF mutations. To expedite patient accrual and establish the R2PD, we amended the protocol to omit the inclusion criteria that patients needed tumors that harbored KRAS, NRAS, or BRAF mutations. As a result, only 2 patients had tumors with KRAS codon 12 mutation in the R2PD dose: one had a pCR and the other had a poor response resulting in tumor up-staging. One patient had a BRAF D594N mutation, and there was evidence of tumor downstaging. As the primary goal of our study was to establish the safe and tolerable dose of trametinib to use in combination with chemoradiation, further study may be needed to truly establish if safety, response, and tumor downstaging for patients with LARC is improved with MEK inhibition in combination with chemoradiation. Regarding postoperative toxicity, we did note postoperative complications that may or may not be due to the study treatment combination. While we do not have historical institutional data available on surgical complications for patients treated with neoadjuvant chemoradiation, our anecdotal experience is that it is likely higher than 5%. We believe a larger study will help us determine whether postoperative complications are out of the ordinary for this treatment combination. Because of the limited scope of our study related to small patient numbers, it is difficult to attribute the few wound-healing complications we had to the novel treatment combination. Other studies have shown higher wound complication rates than this study, with complications ranging from 14% to 80% and need for reoperation in 6%–8% of patients, particularly in patients with APR (49–50). Thus, we feel that none of the individual surgical complications were outside the range and frequency of colorectal surgical complications we typically observe in historically treated patients with 5-FU–based chemoradiation.

Of particular interest for a future trial with regards to efficacy, is the evaluation of the treatment combination both in RAS/RAF wild-type and RAS/RAF-mutant populations to determine whether there is an improvement in response in one particular genomic subgroup more than the other, in comparison with historical results. Most of the preclinical data showing efficacy of MEK inhibition in combination with radiation was demonstrated in RAS/RAF-mutant cells, except the study by Chung and colleagues which demonstrated radiosensitization in DU145 prostate cancer cells. (30–34) Thus, we hypothesize that we would observe a greater degree of benefit with the treatment combination in the RAS/RAF-mutant population compared with the RAS/RAF wild-type population because MEK inhibition would abrogate downstream RAS–RAF–MEK–ERK aberrant signaling that promotes radioresistance. Finally, given the increasing interest and role of nonoperative management (organ preservation) for rectal cancer, novel strategies to improve clinical complete response and long-term durable pelvic control with definitive chemoradiation are also warranted, and may be achieved with strategies that enhance chemoradiosensitization.

In conclusion, our phase I evaluation of this treatment combination of trametinib, 5-FU, and radiation appears to be safe, well-tolerated, and demonstrates early signs of clinical efficacy at the 2 mg cohort, with confirmation of on-target biologic effects. Future prospective studies of this treatment combination may be warranted in rectal cancer, perhaps focusing on RAS/RAF-mutant rectal cancer.

C. Wu is a paid consultant/advisory board member for Array Biopharma and Signatera, and reports receiving other commercial research support from Vaccinex, Boston Biomedical Inc., Lycera, Seattle Genetics, Symphogen, and Rapt Therapeutics. K.K. Ciombor is an employee/paid consultant for Natera and Research to Practice, and reports receiving commercial research grants from Incyte, Array, Nucana, and Daiichi-Sankyo. No potential conflicts of interest were disclosed by the other authors.

Conception and design: C. Wu, T.M. Williams, L. Wei, T. Bekaii-Saab, E. Wuthrick

Development of methodology: C. Wu, T.M. Williams, R. Robb, K.K. Ciombor, C. Timmers, E. Wuthrick

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): C. Wu, T.M. Williams, R. Robb, S. Mikhail, K.K. Ciombor, D.B. Cardin, C. Timmers, S.G. Krishna, M. Arnold, A. Harzman, S. Abdel-Misih, S. Roychowdhury, T. Bekaii-Saab, E. Wuthrick

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): C. Wu, T.M. Williams, R. Robb, L. Wei, K.K. Ciombor, C. Timmers, M. Arnold, T. Bekaii-Saab, E. Wuthrick

Writing, review, and/or revision of the manuscript: C. Wu, T.M. Williams, R. Robb, L. Wei, W. Chen, S. Mikhail, K.K. Ciombor, D.B. Cardin, C. Timmers, S.G. Krishna, M. Arnold, A. Harzman, S. Abdel-Misih, T. Bekaii-Saab, E. Wuthrick

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): T.M. Williams, C. Timmers

Study supervision: C. Wu, T.M. Williams, T. Bekaii-Saab, E. Wuthrick

This study was approved and funded in part by the National Comprehensive Cancer Network (NCCN) Oncology Research Program from general research support provided by Novartis Pharmaceutical Corporation. We thank Dr. Tanios Bekaii-Saab for mentorship on this project. We also thank Hamida Umar and Barbara Kleiber for support in the Clinical Trials Office.

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1.
Siegel
RL
,
Miller
KD
,
Jemal
A
. 
Cancer statistics, 2018
.
CA Cancer J Clin
2018
;
68
:
7
30
.
2.
Bosset
JF
,
Collette
L
,
Calais
G
,
Mineur
L
,
Maingon
P
,
Radosevic-Melic
L
, et al
Chemotherapy with preoperative radiotherapy in rectal cancer
.
N Engl J Med
2006
;
355
:
1114
23
.
3.
Gerard
JP
,
Conroy
T
,
Bonnetain
F
,
Bouche
O
,
Chapet
O
,
Closon-Dejardin
MT
, et al
Preoperative radiotherapy with or without concurrent fluorouracil and leucovorin in T3–4 rectal cancers: results of FFCD 9203
.
J Clin Oncol
2006
;
24
:
4620
5
.
4.
Sauer
R
,
Liersch
T
,
Merkel
S
,
Fietkau
R
,
Hohenberger
W
,
Hess
C
, et al
Preoperative versus postoperative chemoradiotherapy for locally advanced rectal cancer: results of the German CAO/ARO/AIO-94 randomized phase III trial after a median follow-up of 11 years
.
J Clin Oncol
2012
;
30
:
1926
33
.
5.
Aschele
C
,
Cionini
L
,
Lonardi
S
,
Pinto
C
,
Cordio
S
,
Rosati
G
, et al
Primary tumor response to preoperative chemoradiation with or without oxaliplatin in locally advanced rectal cancer: pathologic results of the STAR-01 randomized phase III trial
.
J Clin Oncol
2011
;
29
:
2773
80
.
6.
Bertolini
F
,
Chiara
S
,
Bengala
C
,
Antognoni
P
,
Dealis
C
,
Zironi
S
, et al
Neoadjuvant treatment with single-agent cetuximab followed by 5-FU, cetuximab, and pelvic radiotherapy: a phase II study in locally advanced rectal cancer
.
Int J Radiat Oncol Biol Phys
2009
;
73
:
466
72
.
7.
Gerard
JP
,
Azria
D
,
Gourgou-Bourgade
S
,
Martel-Laffay
I
,
Hennequin
C
,
Etioone
PL
, et al
Comparison of two neoadjuvant chemoradiotherapy regimens for locally advanced rectal cancer: results of the phase III trial ACCORD 12/0405-Prodige 2
.
J Clin Oncol
2010
;
28
:
1638
44
.
8.
Dhillon
AS
,
Hagan
S
,
Rath
O
,
Kolch
W
. 
MAP kinase signalling pathways in cancer
.
Oncogene
2007
;
26
:
3279
90
.
9.
Sebolt-Leopold
JS
,
Herrera
R
. 
Targeting the mitogen-activated protein kinase cascade to treat cancer
.
Nat Rev Cancer
2004
;
4
:
937
47
.
10.
Brink
M
,
de Goeij
AF
,
Weijenberg
MP
,
Roemen
GM
,
Lentjes
MH
,
Pachen
MM
, et al
K-ras oncogene mutations in sporadic colorectal cancer in The Netherlands cohort study
.
Carcinogenesis
2003
;
24
:
703
710
.
11.
Jonsson
M
,
Ekstrand
A
,
Edekling
T
,
Eberhard
J
,
Grabau
D
,
Borg
D
, et al
Experiences from treatment-predictive KRAS testing; high mutation frequency in rectal cancers from females and concurrent mutations in the same tumor
.
BMC Clin Pathol
2009
;
9
:
8
.
12.
De Roock
W
,
Claes
B
,
Bernasconi
D
,
De Schutter
J
,
Biesmans
B
,
Fountzilas
G
, 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
62
.
13.
Wang
L
,
Cunningham
JM
,
Winters
JL
,
Guenther
JC
,
French
AJ
,
Boardman
LA
, et al
BRAF mutations in colon cancer are not likely attributable to defective DNA mismatch repair
.
Cancer Res
2003
;
63
:
5209
12
.
14.
De Roock
W
,
Lambrechts
D
,
Tejpar
S
. 
K-ras mutations and cetuximab in colorectal cancer
.
N Engl J Med
2009
;
360
:
834
.
15.
Cancer Genome Atlas Network
. 
Comprehensive molecular characterization of human colon and rectal cancer
.
Nature
2012
;
487
:
330
7
.
16.
Dent
P
,
Reardon
DB
,
Park
JS
,
Bowers
G
,
Logsdon
C
,
Valerie
K
, et al
Radiation-induced release of transforming growth factor alpha activates the epidermal growth factor receptor and mitogen-activated protein kinase pathway in carcinoma cells, leading to increased proliferation and protection from radiation-induced cell death
.
Mol Biol Cell
1999
;
10
:
2493
506
.
17.
Qiao
L
,
Yacoub
A
,
McKinstry
R
,
Park
JS
,
Caron
R
,
Fisher
PB
, et al
Pharmocologic inhibitors of the mitogen activated protein kinase cascade have the potential to interact with ionizing radiation exposure to induce cell death in carcinoma cells by multiple mechanisms
.
Cancer Biol Ther
2002
;
1
:
168
76
.
18.
Bernhard
EJ
,
Stanbridge
EJ
,
Gupta
S
,
Gupta
AK
,
Soto
D
,
Bakanauskas
VJ
, et al
Direct evidence for the contribution of activated N-ras and K-ras oncogenes to increased intrinsic radiation resistance in human tumor cell lines
.
Cancer Res
2000
;
60
:
6597
6600
.
19.
McKenna
WG
,
Muschel
RJ
,
Gupta
AK
,
Hahn
SM
,
Bernhard
EJ
. 
The RAS signal transduction pathway and its role in radiation sensitivity
.
Oncogene
2003
;
22
:
5866
75
.
20.
Sklar
MD
. 
The ras oncogenes increase the intrinsic resistance of NIH 3T3 cells to ionizing radiation
.
Science
1988
;
239
:
645
7
.
21.
Mak
RH
,
Hermann
G
,
Lewis
JH
,
Aerts
HJ
,
Baldini
EH
,
Chen
AB
, et al
Outcomes by tumor histology and KRAS mutation status after lung stereotactic body radiation therapy for early-stage non-small-cell lung cancer
.
Clin Lung Cancer
2015
;
16
:
24
32
.
22.
Russo
AL
,
Ryan
DP
,
Borger
DR
,
Wo
JY
,
Szymonifka
J
,
Liang
WY
, et al
Mutational and clinical predictors of pathologic complete response in the treatment of locally advanced rectal cancer
.
Journal of Gastroint Cancer
2014
;
45
:
34
9
.
23.
Cassidy
RJ
,
Zhang
X
,
Patel
PR
,
Shelton
JW
,
Escott
CE
,
Sica
GL
, et al
Next-generation sequencing and clinical outcomes of patients with lung adenocarcinoma treated with stereotactic body radiotherapy
.
Cancer
2017
;
123
:
3681
90
.
24.
Hong
TS
,
Wo
JY
,
Borger
DR
,
Yeap
BY
,
McDonnell
EI
,
Willers
H
, et al
Phase II study of proton-based stereotactic body radiation therapy for liver metastases: importance of tumor genotype
.
J Natl Cancer Inst
2017
;
109(9)
.
25.
Grimminger
PP
,
Danenberg
P
,
Dellas
K
,
Arnold
D
,
Rödel
C
,
Machiels
JP
, et al
Biomarkers for cetuximab-based neoadjuvant radiochemotherapy in locally advanced rectal cancer
.
Clin Cancer Res
2011
;
17
:
3469
77
.
26.
Duldulao
MP
,
Lee
W
,
Nelson
RA
,
Li
W
,
Chen
Z
,
Kim
J
, et al
Mutations in specific codons of the KRAS oncogene are associated with variable resistance to neoadjuvant chemoradiation therapy in patients with rectal adenocarcinoma
.
Ann Surg Oncol
2013
;
20
:
2166
71
.
27.
Bernhard
EJ
,
McKenna
WG
,
Hamilton
AD
,
Sebti
SM
,
Qian
Y
,
Wu
JM
, et al
Inhibiting Ras prenylation increases the radiosensitivity of human tumor cell lines with activating mutations of ras oncogenes
.
Cancer Res
1998
;
58
:
1754
61
.
28.
Brunner
TB
,
Cengel
KA
,
Hahn
SM
,
Wu
J
,
Fraker
DL
,
McKenna
WG
, et al
Pancreatic cancer cell radiation survival and prenyltransferase inhibition: the role of K-Ras
.
Cancer Res
2005
;
65
:
8433
41
.
29.
Cengel
KA
,
Deutsch
E
,
Stephens
TC
,
Voong
KR
,
Kao
GD
,
Bernhard
EJ
. 
Radiosensitizing effects of the prenyltransferase inhibitor AZD3409 against RAS mutated cell lines
.
Cancer Biol Ther
2006
;
5
:
1206
10
.
30.
Chung
EJ
,
Brown
AP
,
Asano
H
,
Mandler
M
,
Burgan
WE
,
Carter
D
, et al
In vitro and in vivo radiosensitization with AZD6244 (ARRY-142886), an inhibitor of mitogen-activated protein kinase/extracellular signal-regulated kinase 1/2 kinase
.
Clin Cancer Res
2009
;
15
:
3050
7
.
31.
Urick
ME
,
Chung
EJ
,
Shield
WP
 3rd
,
Gerber
N
,
White
A
,
Sowers
A
, et al
Enhancement of 5-fluorouracil-induced in vitro and in vivo radiosensitization with MEK inhibition
.
Clin Cancer Res
2011
;
17
:
5038
47
.
32.
Shannon
AM
,
Telfer
BA
,
Smith
PD
,
Babur
M
,
Logie
A
,
Wilkinson
RW
, et al
The mitogen-activated protein/extracellular signal-regulated kinase kinase 1/2 inhibitor AZD6244 (ARRY-142886) enhances the radiation responsiveness of lung and colorectal tumor xenografts
.
Clin Cancer Res
2009
;
15
:
6619
29
.
33.
Williams
TM
,
Flecha
AR
,
Keller
P
,
Ram
A
,
Karnak
D
,
Galbán
S
, et al
Cotargeting MAPK and PI3K signaling with concurrent radiotherapy as a strategy for the treatment of pancreatic cancer
.
Mol Cancer Ther
2012
;
11
:
1193
1202
.
34.
Estrada-Bernal
A
,
Chatterjee
M
,
Haque
SJ
,
Yang
L
,
Morgan
MA
,
Kotian
S
, et al
MEK inhibitor GSK1120212-mediated radiosensitization of pancreatic cancer cells involves inhibition of DNA double-strand break repair pathways
.
Cell Cycle
2015
;
14
:
3713
24
.
35.
Yamaguchi
T
,
Kakefuda
R
,
Tajima
N
,
Sowa
Y
,
Sakai
T
. 
Antitumor activities of JTP-74057 (GSK1120212), a novel MEK1/2 inhibitor, on colorectal cancer cell lines in vitro and in vivo
.
Int J Oncol
2011
;
39
:
23
31
.
36.
Flaherty
KT
,
Robert
C
,
Hersey
P
,
Nathan
P
,
Garbe
C
,
Milhem
M
, et al
Improved survival with MEK inhibition in BRAF-mutated melanoma
.
N Engl J Med
2012
;
367
:
107
14
.
37.
Ryan
R
,
Gibbons
D
,
Hyland
JM
,
Treanor
D
,
White
A
,
Mulcahy
HE
, et al
Pathological response following long-course neoadjuvant chemoradiotherapy for locally advanced rectal cancer
.
Histopathology
2005
;
47
:
141
6
.
38.
Washington
MK
,
Berlin
J
,
Branton
PA
,
Burgart
LJ
,
Carter
DK
,
Fitzgibbons
PL
, et al
Protocol for the examination of specimens from patients with primary carcinomas of the colon and rectum
.
Arch Pathol Lab Med
2008
;
132
:
1182
93
.
39.
Langmead
B
,
Salzberg
SL
. 
Fast gapped-read alignment with Bowtie 2
.
Nat Methods
2012
;
9
:
357
9
.
40.
Li
H
,
Handsaker
B
,
Wysoker
A
,
Fennell
T
,
Ruan
J
,
Homer
N
, et al
The sequence alignment/map format and SAMtools
.
Bioinformatics
2009
;
25
:
2078
9
.
41.
McKenna
A
,
Hanna
M
,
Banks
E
,
Sivachenko
A
,
Cibulskis
K
,
Kernytsky
A
, et al
The genome analysis toolkit: a MapReduce framework for analyzing next-generation DNA sequencing data
.
Genome Res
2010
;
20
:
1297
303
.
42.
Koboldt
DC
,
Zhang
Q
,
Larson
DE
,
Shen
D
,
McLellan
MD
,
Lin
L
, et al
VarScan 2: somatic mutation and copy number alteration discovery in cancer by exome sequencing
.
Genome Res
2012
;
22
:
568
76
.
43.
McLaren
W
,
Gil
L
,
Hunt
SE
,
Riat
HS
,
Ritchie
GR
,
Thormann
A
, et al
The ensembl variant effect predictor
.
Genome Biol
2016
;
17
:
122
.
44.
Thorvaldsdottir
H
,
Robinson
JT
,
Mesirov
JP
. 
Integrative Genomics Viewer (IGV): high-performance genomics data visualization and exploration
.
Brief Bioinform
2013
;
14
:
178
192
.
45.
Genomes Project
C
,
Auton
A
,
Brooks
LD
,
Durbin
RM
,
Garrison
EP
,
Kang
HM
, et al
A global reference for human genetic variation
.
Nature
2015
;
526
:
68
74
.
46.
Lek
M
,
Karczewski
KJ
,
Minikel
EV
,
Samocha
KE
,
Banks
E
,
Fennell
T
, et al
Analysis of protein-coding genetic variation in 60,706 humans
.
Nature
2016
;
536
:
285
291
.
47.
Roh
MS
,
Colangelo
LH
,
O'Connell
MJ
,
Yothers
G
,
Deutsch
M
,
Allegra
C
, et al
Preoperative multimodality therapy improves disease-free survival in patients with carcinoma of the rectum.: NSABP R-03
.
J Clin Oncol
2009
.
1
:
27
:
5125
30
.
48.
George
TJ
,
Franke
AJ
,
Chakravarthy
AB
,
Das
P
,
Dasari
A
,
El-Rayes
BF
, et al
National cancer institute state of the science: targeted radiosensitizers in colorectal cancer
.
Cancer
2019
;
125
:
2732
46
.
49.
Hawkins
AT
,
Albutt
K
,
Wise
PE
,
Alavi
K
,
Sudan
R
,
Kaiser
AM
, et al
Abdominoperineal resection for rectal cancer in the twenty-first century: indications, techniques, and outcomes
.
J Gastrointest Surg
2018
;
22
:
1477
87
.
50.
Saadat
LV
,
Fields
AC
,
Lyu
H
,
Urman
RD
,
Whang
EE
,
Goldberg
J
, et al
National Surgical Quality Improvement Program analysis of unplanned reoperation in patients undergoing low anterior resection or abdominoperineal resection for rectal cancer
.
Surgery
2019
;
165
:
602
7
.