Purpose:

Molecular properties associated with complete response or acquired resistance to concurrent chemotherapy and radiotherapy (CRT) are incompletely characterized.

Experimental Design: We performed integrated whole-exome/transcriptome sequencing and immune infiltrate analysis on rectal adenocarcinoma tumors prior to neoadjuvant CRT (pre-CRT) and at time of resection (post-CRT) in 17 patients [8 complete/partial responders, 9 nonresponders (NR)].

Results:

CRT was not associated with increased tumor mutational burden or neoantigen load and did not alter the distribution of established somatic tumor mutations in rectal cancer. Concurrent KRAS/TP53 mutations (KP) associated with NR tumors and were enriched for an epithelial–mesenchymal transition transcriptional program. Furthermore, NR was associated with reduced CD4/CD8 T-cell infiltrates and a post-CRT M2 macrophage phenotype. Absence of any local tumor recurrences, KP/NR status predicted worse progression-free survival, suggesting that local immune escape during or after CRT with specific genomic features contributes to distant progression.

Conclusions:

Overall, while CRT did not impact genomic profiles, CRT impacted the tumor immune microenvironment, particularly in resistant cases.

This article is featured in Highlights of This Issue, p. 5429

Translational Relevance

Integrated tumor profiling of patient-matched rectal adenocarcinomas before and after neoadjuvant chemo/radiotherapy reveals insights into tumor evolution and treatment resistance mechanisms. The inability of neoadjuvant therapy to enhance tumor mutational burden coupled with poor response and local immune escape, particularly in KRAS/TP53-mutated tumors, warrant novel treatment approaches.

Radiotherapy is used in the management of nearly two-thirds of cancers (1), often fulfilling the role of a curative treatment modality in place of surgery. Therapeutic radiation can be adapted to target tumors in various anatomic locations as well as various malignancies. The radiation dose and fractionation can be altered to maximize tumor killing while sparing normal tissues (2). Radiotherapy is typically combined with concurrent chemotherapy (CRT) in locally advanced disease. When used neoadjuvantly, pathologic downstaging is a surrogate of long-term outcome in many disease sites (3–7). For example, in rectal cancer, approximately 9%–20% of patients with locally advanced disease have a pathologic complete response (pCR) to neoadjuvant CRT (8), while 20%–40% of patients have little to no response (9, 10). Predictive biomarkers of pCR remain to be established.

The major mechanism of radiation-induced cell killing is likely through DNA damage. There is, however, emerging evidence that radiation also has effects on the tumor microenvironment with variation based on anatomic site, tumor histology, and multiple other characteristics (11). These cell killing effects can be further augmented by combining radiation with radiosensitizing systemic agents (12). In addition, there is recent interest in the utility of radiation to alter the adaptive immune response to improve treatment outcomes by creating a local antitumor immune response that may be modulated into a systemic antitumor immune response with the use of immunomodulatory agents (13–15). Proposed mechanisms include possible creation of increased neoantigens or tumor mutational burden (TMB) through the DNA-damaging effects of radiation (16, 17), the latter of which has been previously demonstrated to correlate with response after treatment with immune checkpoint inhibitors (18, 19). Despite the widespread use of radiotherapy for solid tumors, there has been slow progress in predicting treatment outcomes to radiation to allow for personalization of therapy on an individual level (12, 15). Biomarkers have been in use and ultimately transformed the field of systemic therapy, while few predictive biomarkers are available for radiotherapy (12).

Using rectal cancer as our model (3, 20), we hypothesized that a comprehensive assessment of patient-matched pre- and post-CRT specimens, examining both tumor-intrinsic and microenvironmental features from the tumor site, may reveal features associated with treatment response at the molecular level. To that end, we leveraged a cohort of patients with locally advanced rectal cancer who underwent fluoropyrimidine-based CRT to a dose of 50.4 Gy followed by surgical resection and analyzed genomic tumor changes in the matched pre- and posttreatment rectal tumor samples to identify drivers of resistance to neoadjuvant CRT and thereby identify biomarkers for patient stratification.

Patient population and samples

We retrospectively identified patients with biopsy-proven locally advanced rectal cancer (defined as T3–4 or N+) who received neoadjuvant fluoropyrimidine-based chemotherapy concurrently with 50.4 Gy radiotherapy, followed by surgical resection within 8–11 weeks between 2010 and 2016 (20). Patients then went on to receive adjuvant systemic therapy, which consisted of FOLFOX (21). Patients had to have documented written consent through the institutional review board–approved protocol that collects tissue and whole-blood specimens on patients with gastrointestinal malignancies in accordance with the Declaration of Helsinki and all applicable legal regulatory requirements. There were 77 patients who met initial criteria. Eligible patients had to have sufficient tumor tissue in study specimens of formalin-fixed, paraffin-embedded (FFPE) tissue sections from surgical samples, as well as a germline DNA specimen that was extracted from either peripheral mononuclear cells or histologically normal rectal tissue. Twenty patients were identified with sufficient tissue available. All patients were arbitrarily identified with no prior knowledge of genomic tumor status. All samples had to pass standard quality control measures. We identified 34 pre- and post-CRT–matched tumor samples from 17 patients in our final cohort. Nine and 8 patients were classified as nonresponders (no evidence of any pathologic downstaging, NR) and responders (pathologic complete response or pathologic partial response, R), respectively, at surgery based on pathologic evaluation.

DNA extraction and whole-exome sequencing

DNA extraction, whole-exome library preparation, and sequencing were performed for the samples as described previously (22, 23). Slides were cut from FFPE blocks and examined by a board-certified pathologist to select high-density cancer blocks and ensure high purity of cancer DNA. Biopsy cores were taken from the corresponding tissue block for DNA extraction. DNA was extracted using Qiagen's QIAamp DNA FFPE Tissue Kit Quantitation Reagent (Invitrogen). DNA was stored at −20°C.

Whole-exome capture libraries were constructed from 100 ng of DNA from tumor and normal tissue after sample shearing, end repair, and phosphorylation and ligation to barcoded sequencing adaptors. Ligated DNA was size selected for lengths between 200 and 350 bp and subjected to exonic hybrid capture using The Broad Institute Genomics Platform Custom Illumina bait. The Illumina exome specifically targets approximately 37.7 Mb of mainly exonic territory made up of all targets from the Agilent exome design (Agilent SureSelect All Exon V2), all coding regions of Gencode V11 genes, and all coding regions of RefSeq gene and KnownGene tracks from the UCSC genome browser (http://genome.ucsc.edu). The sample was multiplexed and sequenced using Illumina HiSeq technology.

Sequencing was performed to an average depth of 150 ×. Data were analyzed using the Broad Picard Pipeline, which includes demultiplexing and data aggregation.

Quality control and variant calling

Initial data processing and analysis of exome sequence data were performed using Broad Institute pipelines as described previously (23). Using the Broad Picard Pipeline for alignment, BAM files were uploaded into the Firehose infrastructure to manage intermediate analysis files executed by analysis pipelines. Quality control modules in Firehose (24) were run to compare the tumor and normal genotypes and ensure concordance between samples. Of samples from 20 initial patients, six samples from 3 patients were abandoned because of high estimates of tumor contamination (25), inadequate coverage (<40× tumor average coverage), or low tumor purity (26). This yielded a final number of 17 total pairs of pre- and posttreatment tumors for analysis.

The MuTect algorithm (27) was applied to identify somatic single-nucleotide variants in targeted exons. Strelka (28) was used to identify small deletions or insertions, and alterations were annotated with Oncotator (29). Mutations were examined for distribution and type and confirmed using the integrative genomics viewer (30, 31).

Transcriptome capture method cDNA library construction

Using established protocols (32), total RNA was assessed for quality using the Caliper LabChip GX2. The percentage of fragments with a size greater than 200 nucleotides (DV200) was calculated using software. An aliquot of 200 ng of RNA was used as the input for first strand cDNA synthesis using Illumina's TruSeq RNA Access Library Prep Kit. Synthesis of the second strand of cDNA was followed by indexed adapter ligation. Subsequent PCR amplification enriched for adapted fragments. The amplified libraries were quantified using an automated PicoGreen assay.

A total of 200 ng of each cDNA library, not including controls, were combined into 4-plex pools. Capture probes that target the exome were added and hybridized for recommended time. Following hybridization, streptavidin magnetic beads were used to capture the library-bound probes from the previous step. Two wash steps effectively remove any nonspecifically bound products. These same hybridization, capture, and wash steps are repeated to assure high specificity. A second round of amplification enriches the captured libraries. After enrichment the libraries were quantified with qPCR using the KAPA Library Quantification Kit for Illumina Sequencing Platforms and then pooled equimolarly. The entire process is in 96-well format and all pipetting is done by either Agilent Bravo or Hamilton Starlet.

Pooled libraries were normalized to 2 nmol/L and denatured using 0.1 N NaOH prior to sequencing. Flowcell cluster amplification and sequencing were performed according to the manufacturer's protocols using HiSeq 2500. Each run was a 76 bp paired-end with an eight-base index barcode read. Data was analyzed using the Broad Picard Pipeline, which includes demultiplexing and data aggregation.

Neoantigen prediction

HLA-type was inferred using POLYSOLVER (33) which uses a normal tissue BAM file as input. It then employs a Bayesian classifier to determine the genotype for each patient. Neoantigens were predicted for each patient by defining all novel amino acid 9mers and 10mers resulting from mutations (23). We filtered out mutations with <3 supportive reads, or <30 total reads at the position. Neoantigen prediction continued on the basis of whether predicted binding affinity to the patient's germline HLA alleles was <500 nmol/L using NetMHCpan (34). Correlations and associated P values between neoantigen load and R versus NR was performed using Mann–Whitney U tests, P < 0.05 was considered significant.

Purity/ploidy and clonal/subclonal mutational calls

Purity and ploidy for each sample was estimated using ABSOLUTE algorithm (35). This algorithm integrates variant allele frequency distributions and copy number variants to estimate absolute tumor purity and ploidy and infer cancer cell fraction (CCF), which is the proportion of cancer cells in the sample which contain each mutation. An ABSOLUTE extension algorithm (35) was used to construct an inferred phylogenetic tree with clones, subclones, and evolutionary relationships in pre- and posttreatment tumor samples. As described by Brastianos and colleagues (36), clones and subclones were determined through Markov Chain Monte Carlo sampling using Dirichlet process Mixture Models on pre- and post-CRT mutation CCFs, which assigns mutations to subclones without prespecifying the number of subclones. Mutations inferred to be in a subclone with a CCF ≥ 0.8 were described as “clonal,” while those inferred to be in a subclone with CCF < 0.8 were called “subclonal.” For each subclone, two CCFs were inferred; one CCF in the pretreatment tumor and CCF in the posttreatment tumor (23).

Changes in mutational and neoantigen load

Changes in mutational, neoantigen, and indel load were calculated using a paired t test of changes in paired samples with a null hypothesis of a difference of 0 (23). P < 0.05 was considered to be statistically significant.

Discovery of resistance or sensitivity biomarkers

We used MutSig2CV (26) to identify significantly mutated genes across our cohort of pre-CRT and post-CRT tumors. Each altered gene in the pretreatment tumors had a P value calculated for mutational significance considering only mutations private to these samples. Similarly, a P value of mutational significance considering only those mutations private to the posttreatment tumor was calculated. Adjustment for hypothesis testing was performed using a Benjamini–Hochberg FDR of 0.1 (23).

Gene expression profiling

Available RNA-Seq data were analyzed as described previously (37). Briefly, expression data were examined and adjusted for batch effects using ComBat (38) using the R Bioconductor package “sva” V3.8 (39). Gene set enrichment analysis (40) was run using https://genepattern.broadinstitute.org using 50 “Hallmark” gene sets to investigate differences in gene set expression in R versus NR (pre-CRT R vs. pre-CRT NR; post-CRT R vs. post-CRT NR) with 1,000 permutations, type “gene_set.” Gene level transcripts per million (TPM) were the input. Family-wise error rates were calculated to identify significant gene sets.

To determine the relationship between CRT and the immune landscape, we analyzed matched transcriptomes from the tumors using CIBERSORT (41) to deconvolute immune cell populations from bulk transcriptome data using immune cell–associated signatures. From this, we inferred overall immune infiltrate and relative immune cell populations in both the pre-CRT and post-CRT specimens. This was run using the CIBERSORT interface (https://cibersort.stanford.edu). The analysis was set to absolute quantification output. Input was gene level TPM and leukocyte gene signature matrix (LM22; ref. 41) was used to deconvolve 22 immune cell subset populations. Absolute quantification normalizing by the 50th percentile of overall gene expression generated a metric that is comparable between samples. Correlations and associated P values between groups of pre-CRT versus post-CRT and R versus NR was performed using Mann–Whitney U tests, P < 0.05 was considered significant. To account for multiple hypothesis testing, a Benjamini–Hochberg FDR of 0.1 was used to identify highly significant associations.

IHC

Details of the six antibodies (PD1, PD-L1, PD-L2, CTLA4, CD4, and CD8), host species, clone, and dilatation are given in Supplementary Table S1. IHC was performed automatically using a Benchmark XT/Discovery ULTRA Staining Module (Ventana Medical Systems, Inc.) using established protocols (42). In brief, protocols consisted of pretreatment with CC1 (pH 8.0), incubation with primary antibodies, and detection using a DAB-system (catalog no. 760-500, Ventana Medical Systems, Inc) including ultraview inhibitor, horseradish peroxidase, multimer chromogen, H2O2, and copper. In brief, sections were washed for 5 minutes (xylene × 3, 100% ethanol × 2, 95% ethanol × 1, 70% ethanol × 1, and PBS × 1). Staining properties and specificity have been determined previously (refs. 37, 43–47; Supplementary Table S1), which we additionally ascertained using negative and positive controls (Tonsil).

Microscopy and quantification

For light microscopy, we captured images using an Olympus DP27 camera attached to an Olympus BX40 light microscope (Olympus America). All markers were evaluated on tumor and nontumor compartments and scored as positive versus negative using established cut-off points (48–50). For CD4 and CD8 we additionally captured four images (high power field, 400×) and applied established image quantification tools. Briefly, segmentation of cells was achieved using threshold filters in combination with circularity and size cutoffs using “cell counter” and “analyze particle” plug-ins in ImageJ Software (NIH, Bethesda, MD; ref. 42). For statistical analysis of CD4 and CD8 staining of immune infiltrates, we took the average and median of four independent regions of interest. Differences in CD4 and CD8 T-cell infiltrates between pre-/post-CRT samples were calculated using a t test of changes with a null hypothesis of a difference of 0. P < 0.05 was considered to be statistically significant. Correlations and associated P values between groups of pre-CRT versus post-CRT, R versus NR, and KP genotype versus no KP genotype were performed using Mann–Whitney U tests, P < 0.05 was considered significant.

Outcome analysis

We analyzed the association between R versus NR and KRAS/TP53 mutation genotype versus no KRAS/TP53 mutation genotype with progression-free survival (PFS) using the Kaplan–Meier method. All statistical tests were performed using R version 3.5.2 and Prism 8 Software (GraphPad).

Data availability

All BAMS for the matched pre- and posttreatment tumors will be deposited in dbGAP (phs001829.v1.p1).

Chemoradiation does not increase TMB or neoantigen load

We assembled a cohort of 17 patients with locally advanced rectal carcinoma, of whom 9 were characterized pathologically as R and 8 as NR following neoadjuvant CRT (Materials and Methods). Tumor genotype was unknown at the time of case identification. These patients had sufficient pre-CRT biopsy tissue and post-CRT surgical resection tissue available for multiple analytical pipelines including deep whole-exome sequencing (Fig. 1A). Demographic, treatment, and tumor characteristics are summarized in Supplementary Tables S2 and S3. All tumors demonstrated microsatellite stability. Median follow-up of the cohort was 47.1 months (range, 5.8–90.6). There were no local tumor failures. Overall, NR status was associated with reduced PFS compared with R with 5-year PFS 44% versus 100%, respectively (log-rank P = 0.02; Fig. 1B). Median PFS for NR and R was 24.8 months and not reached, respectively.

Figure 1.

Integrated molecular characterization. A, Sample inclusion and analytic workflow. B, PFS by response. NR predicted poorer PFS compared with R with 5-year PFS of 44% versus 100% (log-rank P = 0.02). C, Mutational burden in cohort (paired t test; P = 0.4). Patients are ordered by response group (Rs and NRs), with tumor mutation burden in decreasing order within each response category. D, Neoantigen load in cohort (paired t test; P = 0.12). E, Mutations in the cohort. Shown are the genes that were most commonly mutated as assessed by MutSig2CV analysis. WES, whole-exome sequencing; RNA, RNA-sequencing.

Figure 1.

Integrated molecular characterization. A, Sample inclusion and analytic workflow. B, PFS by response. NR predicted poorer PFS compared with R with 5-year PFS of 44% versus 100% (log-rank P = 0.02). C, Mutational burden in cohort (paired t test; P = 0.4). Patients are ordered by response group (Rs and NRs), with tumor mutation burden in decreasing order within each response category. D, Neoantigen load in cohort (paired t test; P = 0.12). E, Mutations in the cohort. Shown are the genes that were most commonly mutated as assessed by MutSig2CV analysis. WES, whole-exome sequencing; RNA, RNA-sequencing.

Close modal

No statistically significant change in TMB before and after exposure to CRT was observed in our cohort (P = 0.40; Fig. 1C). A similar analysis of predicted neoantigen burden between pre- and post-CRT tumors also demonstrated no statistically significant change (P = 0.12; Fig. 1D). Neither pre- nor post-CRT neoantigen load were associated with treatment response (P = 0.81; Supplementary Fig. S1 and P = 0.42; Supplementary Fig. S2, respectively). We also found no difference in indel loads between pre- and posttreatment samples (P = 0.20; Supplementary Figure S3). As has been demonstrated previously (51–54), the most frequently mutated genes pre- and post-CRT included KRAS, TP53, and APC (Fig. 1E). Thus, global somatic mutations were not impacted by exposure to CRT in this cohort.

Presence of KRAS and TP53 comutation predicts resistance to chemoradiation

In evaluating differences in specific somatic mutations between R versus NR cases, we observed that NR tumors were enriched for concurrent KRAS and TP53 mutations (KP genotype) in contrast to R tumors (Fisher exact P = 0.05; Fig. 2A and B), as has been described previously (55–57). Notably, one pre-CRT KRAS-mutated tumor harbored a TP53 mutation post-CRT that was not detected in the pretreatment tumor despite sufficient power to detect a mutation; this patient was also a NR (Fig. 2C; Supplementary Figs. S4 and S5), suggesting emergence of a radioresistant subclone. Given its association with NR, we next investigated the association between KP genotype and PFS. Patients with the KP genotype experienced reduced 5-year PFS (38%) compared with those without (90%, log-rank P = 0.04; Fig. 2D).

Figure 2.

Comutation of KRAS and TP53 predicts resistance to chemoradiation. A, NR tumors were enriched for concurrent mutations in KRAS and TP53 genes compared with R (P = 0.05; Fisher exact test). B, Cancer cell fraction pre-/post-CRT for the KRAS and TP53 genes among the one R and six NR samples, respectively. C, Cancer cell fraction cluster plot for RC009 demonstrates the TP53 mutation in the posttreatment clones. D, Patients harboring the co-KP genotype had poorer 5-year PFS (38%, log-rank P = 0.04).

Figure 2.

Comutation of KRAS and TP53 predicts resistance to chemoradiation. A, NR tumors were enriched for concurrent mutations in KRAS and TP53 genes compared with R (P = 0.05; Fisher exact test). B, Cancer cell fraction pre-/post-CRT for the KRAS and TP53 genes among the one R and six NR samples, respectively. C, Cancer cell fraction cluster plot for RC009 demonstrates the TP53 mutation in the posttreatment clones. D, Patients harboring the co-KP genotype had poorer 5-year PFS (38%, log-rank P = 0.04).

Close modal

Immune microenvironmental properties in rectal cancers treated with chemoradiation

To complement our investigation of tumor-intrinsic genomic properties discriminating response to CRT, we examined how transcriptional programs in the tumor or microenvironment were impacted by exposure to these therapies. Among the responders, there were 14 unique transcriptional programs significantly enriched in the pre-CRT samples and one unique transcriptional program significantly enriched in the post-CRT samples, with IFNα response genes enriched in both pre-/post-CRT samples (FWER P = 0.00; Fig. 3A). Among the NR, there were no unique significantly enriched transcriptional programs in the pre-CRT samples and there were five unique transcriptional programs significantly enriched in the post-CRT samples, with the angiogenesis and epithelial–mesenchymal transition (EMT) transcriptional programs enriched among both pre- and post-CRT samples (FWER P = 0.00; Fig. 3A).

Figure 3.

Transcriptome and gene expression profiling identifies unique mechanisms behind radiation resistance among pathologic NRs. A, Gene set enrichment analysis demonstrated IFNα response genes enriched in both pre-/post-CRT samples in the R (FWER P = 0.00), while angiogenesis and epithelial–mesenchymal transition genes were enriched in both pre-/post-CRT samples in the NR (FWER P = 0.00). B, The y-axis is an absolute quantification. Dotted lines represent individual paired patients (R vs. NR). Immune cell infiltrate significantly increased between pre-/post-CRT samples (t test; P = 0.04). C, The y-axis is an absolute quantification, x-axis denotes immune cell subset populations with pre/post next to each other for each individual subset. Increased M2 macrophages were observed in post-CRT specimens among NR (Mann–Whitney U test; P = 0.005; Benjamini–Hochberg FDR q = 0.1), along with increased naïve B cells (Mann–Whitney U test; P = 0.03), monocytes (Mann–Whitney U test; P = 0.03), and resting mast cells (Mann–Whitney U test; P = 0.03). ***, significance per t test; *, significance per Mann–Whitney U test; **, significance per Benjamini–Hochberg FDR.

Figure 3.

Transcriptome and gene expression profiling identifies unique mechanisms behind radiation resistance among pathologic NRs. A, Gene set enrichment analysis demonstrated IFNα response genes enriched in both pre-/post-CRT samples in the R (FWER P = 0.00), while angiogenesis and epithelial–mesenchymal transition genes were enriched in both pre-/post-CRT samples in the NR (FWER P = 0.00). B, The y-axis is an absolute quantification. Dotted lines represent individual paired patients (R vs. NR). Immune cell infiltrate significantly increased between pre-/post-CRT samples (t test; P = 0.04). C, The y-axis is an absolute quantification, x-axis denotes immune cell subset populations with pre/post next to each other for each individual subset. Increased M2 macrophages were observed in post-CRT specimens among NR (Mann–Whitney U test; P = 0.005; Benjamini–Hochberg FDR q = 0.1), along with increased naïve B cells (Mann–Whitney U test; P = 0.03), monocytes (Mann–Whitney U test; P = 0.03), and resting mast cells (Mann–Whitney U test; P = 0.03). ***, significance per t test; *, significance per Mann–Whitney U test; **, significance per Benjamini–Hochberg FDR.

Close modal

Given the immune-related transcriptional programs enriched pre-/post-CRT, we next examined immune cell infiltrates inferred from bulk transcriptome data (Materials and Methods). Total immune infiltrate levels were significantly higher in post-CRT specimens relative to their pre-CRT counterparts (P = 0.04; Fig. 3B). Overall, we observed significantly more naïve B cells (P = 0.044), CD8 T cells (P = 0.002), monocytes (P = 0.01), M2 macrophages (P = 0.002), and resting mast cells (P = 0.0007) in the post-CRT tumor specimens. In contrast, there were significantly more memory B cells (P = 0.04) and activated mast cells (P = 0.006) in the pre-CRT tumor specimens (Supplementary Fig. S6).

Interestingly, when limiting the analysis to NR pre-/post-CRT, we observed significantly more M2 macrophages (P = 0.005; FDR q = 0.1) in the post-CRT tumor specimens, as well as naïve B cells (P = 0.03), monocytes (P = 0.03), and resting mast cells (P = 0.03), with significantly more activated mast cells in the pre-CRT specimens (P = 0.04; Fig. 3C).

To complement bulk transcriptome analysis, we also evaluated immune infiltrate using IHC for CD4 and CD8 T cells (Supplementary Table S4). The number of CD8 T cells trended toward a global increase between pre-CRT and post-CRT samples (P = 0.47; Supplementary Fig. S7). In the pre-CRT samples, there were more CD8 T cells in R compared with NR (P = 0.14; Fig. 4A) and complete responders (CR) samples had significantly more CD8 immune infiltration compared with NR (P = 0.04; Fig. 4A and B; Supplementary Figs. S8 and S9).

Figure 4.

Immune infiltrate analysis by IHC demonstrates shifts in immune cell composition after chemoradiation. A, Among the pre-CRT samples, there were more CD8 cells among R compared with NR, and CR samples had significantly more CD8 cells compared with NR (Mann–Whitney U test; P = 0.04). B, Representative images of a R with high CD8 infiltrate and NR with low CD8 infiltrate in pre-CRT specimens. C, Among the pre-CRT samples, CR had increased CD4 immune infiltrate compared with NR (Mann–Whitney U test; P = 0.37). D, Representative images of a R with high CD4 infiltrate and NR with low CD4 infiltrate in pre-CRT specimens. *, significant P value; ns, nonsignificant; each individual dot represents the average of four independent regions of interest per patient.

Figure 4.

Immune infiltrate analysis by IHC demonstrates shifts in immune cell composition after chemoradiation. A, Among the pre-CRT samples, there were more CD8 cells among R compared with NR, and CR samples had significantly more CD8 cells compared with NR (Mann–Whitney U test; P = 0.04). B, Representative images of a R with high CD8 infiltrate and NR with low CD8 infiltrate in pre-CRT specimens. C, Among the pre-CRT samples, CR had increased CD4 immune infiltrate compared with NR (Mann–Whitney U test; P = 0.37). D, Representative images of a R with high CD4 infiltrate and NR with low CD4 infiltrate in pre-CRT specimens. *, significant P value; ns, nonsignificant; each individual dot represents the average of four independent regions of interest per patient.

Close modal

Globally, CD4 infiltrate decreased between pre- and post-CRT, but this trend was not statistically significant (P = 0.89; Supplementary Fig. S10). Similar to CD8 T cells, NR trended toward having less CD4 immune infiltration compared with R (P = 0.32; Fig. 4C). When further breaking down response into CR versus partial responders (PR), CR appeared to have more CD4 immune infiltrate compared with NR (P = 0.37; Fig. 4C and D; Supplementary Figs. S11 and S12). In summary, while IHC demonstrated significant differences in T-cell infiltrate pre-CRT between R versus NR, clear shifts in immune infiltrate composition were observed after CRT in NR patients based on bulk transcriptome analysis.

To our knowledge, this is the first study to evaluate both genomic and microenvironmental changes at a primary rectal cancer tumor site exposed to preoperative CRT. Our data provide an opportunity to understand treatment-associated genomic changes between pre- and post-CRT specimens directly in patients. Tumor evolution has been previously studied primarily in the context of systemic cancer therapeutics in solid tumors (23, 36, 58–61), while most radiotherapy-based studies have examined candidate germline features or leveraged microarray data (11, 12, 62–75). Here we performed integrative comprehensive molecular characterization to dissect tumor and immune properties that track with CRT resistance.

Tumor mutation burden has been extensively studied and is suggested to be a marker of tumor responsiveness to immune checkpoint blockade (18, 19). It has been hypothesized that radiation may be able to increase TMB through its DNA-damaging mechanisms. Our data did not demonstrate an increase in overall mutational or neoantigen load after exposure to CRT. This finding is consistent with other pre- and postmatched tumor evolutionary assessments in the context of systemic therapy, particularly with cisplatin-based chemotherapy (23). Our data support the notion that chemotherapy or radiation is generally insufficient to prime the immune system by creating appropriate mutations or neoantigens (76, 77).

While global genomic tumor properties were not clearly different between response groups, NR were more likely to harbor co-KRAS/TP53 mutations compared with R. The KP genotype has been previously suggested to be associated with radioresistance but the underlying mechanisms are poorly understood (55–57). Our observations in KP/NR cases suggest a previously unrecognized mechanism of immune suppression (Fig. 3). We demonstrated that NRs were more likely to express a M2 macrophage phenotype, as well as enrichment for an EMT transcriptional program in the post-CRT specimens. The M2 phenotype is known to be anti-inflammatory, proangiogenic, and metastasis promoting (78–80), while EMT plays a role in cancer metastasis and treatment resistance (81–85). Thus, KP/NR status may be associated with local immune escape during or after CRT. Of note, in our cohort without local recurrences, we found that NR/KP was associated with metastatic progression. Taken together, this suggests that KP/NR-associated local immune escape leads to distant metastatic disease and reduced PFS (Figs. 1B and 2D). Thus, these tumors may benefit from novel neoadjuvant treatment approaches to reduce the risk of immune escape and metastatic seeding.

There are several limitations to this study. Small patient numbers make additional in-depth analyses and conclusions difficult, hence our findings need validation in larger, independent cohorts in diverse clinical settings. Many of our associations may be dependent on one another, as we do not have enough events to appropriately determine whether KRAS/TP53 genotype or pCR rate is more predictive of PFS through a multivariable regression. We rely on pCR as a biomarker of response, which has been called into question after preoperative CRT for rectal cancer (86) as pCR can vary and may be a function of time between end of CRT and surgical resection, although it has been used as a robust endpoint when evaluating novel systemic agents in other solid tumors (4). Some of our findings may be attributable to samples having higher or lower initial tumor burden; to overcome this issue, we performed purity/ploidy corrected molecular analysis through the ABSOLUTE algorithm (35) to account for differences in stromally admixed tumor specimens. We did not evaluate the impact of short-course preoperative radiotherapy nor other high-dose ablative radiotherapy schedules, which may elicit more mutagenesis and an immune response within the tumor microenvironment due to the higher dose per fraction during treatment (16, 17, 87, 88). We also acknowledge that interpretation of in silico–derived neoantigens from the mutations for each sample requires significant validation for improved interpretation. In addition, tumor spatial heterogeneity cannot be ruled out in this study as we do not have data from multiple areas of each tissue sample.

Overall, our study creates a path forward by leveraging molecular profiling for consideration of preoperative CRT in patients with locally advanced tumors. This study also highlights the larger opportunity for additional investigations to elucidate novel mechanisms behind radioresistance across solid tumors.

R.B. Corcoran has ownership interests at Avidity Biosciences, nRichDx, and Fount Therapeutics, is a consultant/advisory board member for Array Biopharma, Astex Pharmaceuticals, Avidity Biosciences, Merrimack, Novartis, Shire, nRichDx, Bristol-Myers Squibb, FOG Pharma, Fount Therapeutics, N-of-One, Revolution Medicines, Taiho, Amgen, Chugai, Genentech, LOXO Oncology, Roivant, Roche, Spectrum Pharmaceuticals, Symphogen, and Warp Drive Bio, and reports receiving commercial research support from Asana, Sanofi, and AstraZeneca. T.S. Hong is a consultant/advisory board member for EMD Serono and Merck, and reports receiving commercial research support from Taiho, AstraZeneca, IntraOp, Bristol Myers Squibb and Ipsen. E.M. Van Allen has ownership interests (including patents) at Syapse, Microsoft, Genome Medical, and Tango Therapeutics, is a consultant/advisory board member for Genome Medical, Invitae, Tango Therapeutics, Dynamo, Illumina, and Foresite Capital, and reports receiving commercial research grants from Novartis and Bristol-Myers Squibb IION. No potential conflicts of interest were disclosed by the other authors.

The content is solely the responsibility of the authors and does not necessarily reflect the official views of the NIH.

Conception and design: S.C. Kamran, J.K. Lennerz, R.B. Corcoran, T.S. Hong, E.M. Van Allen

Development of methodology: S.C. Kamran, J.K. Lennerz, C.A. Margolis, D. Liu, B. Reardon, S.L. Carter, E.M. Van Allen

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): S.C. Kamran, J.K. Lennerz, E.E. Van Seventer, J.Y. Wo, T.S. Hong, E.M. Van Allen

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): S.C. Kamran, J.K. Lennerz, C.A. Margolis, D. Liu, B. Reardon, S.A. Wankowicz, J.Y. Wo, H. Willers, R.B. Corcoran, T.S. Hong, E.M. Van Allen

Writing, review, and/or revision of the manuscript: S.C. Kamran, J.K. Lennerz, C.A. Margolis, B. Reardon, S.A. Wankowicz, J.Y. Wo, H. Willers, R.B. Corcoran, T.S. Hong, E.M. Van Allen

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): S.C. Kamran, J.K. Lennerz, A. Tracy, E.M. Van Allen

Study supervision: S.C. Kamran, T.S. Hong, E.M. Van Allen

This work is funded by grants from Damon Runyon Foundation (to E.M. Van Allen), NCI R01CA227388, K08 CA188615, R37 CA222574, U01 CA233100 (E.M. Van Allen) NCI U01CA220714 (to H. Willers). This work was funded in part by NIH grant no. R01 CA225655 (to J.K. Lennerz).

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.
Miller
KD
,
Siegel
RL
,
Lin
CC
,
Mariotto
AB
,
Kramer
JL
,
Rowland
JH
, et al
Cancer treatment and survivorship statistics, 2016
.
CA Cancer J Clin
2016
;
66
:
271
89
.
2.
Brown
JM
,
Carlson
DJ
,
Brenner
DJ
. 
The tumor radiobiology of SRS and SBRT: are more than the 5 Rs involved?
Int J Radiat Oncol Biol Phys
2014
;
88
:
254
62
.
3.
Kim
NK
,
Baik
SH
,
Seong
JS
,
Kim
H
,
Roh
JK
,
Lee
KY
, et al
Oncologic outcomes after neoadjuvant chemoradiation followed by curative resection with tumor-specific mesorectal excision for fixed locally advanced rectal cancer: Impact of postirradiated pathologic downstaging on local recurrence and survival
.
Ann Surg
2006
;
244
:
1024
30
.
4.
Das
S
,
Lo
AW
. 
Re-inventing drug development: a case study of the I-SPY 2 breast cancer clinical trials program
.
Contemp Clin Trials
2017
;
62
:
168
74
.
5.
Blum Murphy
M
,
Xiao
L
,
Patel
VR
,
Maru
DM
,
Correa
AM
,
F
GA
, et al
Pathological complete response in patients with esophageal cancer after the trimodality approach: the association with baseline variables and survival-The University of Texas MD Anderson Cancer Center experience
.
Cancer
2017
;
123
:
4106
13
.
6.
Hellmann
MD
,
Chaft
JE
,
William
WN
 Jr
,
Rusch
V
,
Pisters
KM
,
Kalhor
N
, et al
Pathological response after neoadjuvant chemotherapy in resectable non-small-cell lung cancers: proposal for the use of major pathological response as a surrogate endpoint
.
Lancet Oncol
2014
;
15
:
e42
50
.
7.
Spring
L
,
Greenup
R
,
Niemierko
A
,
Schapira
L
,
Haddad
S
,
Jimenez
R
, et al
Pathologic complete response after neoadjuvant chemotherapy and long-term outcomes among young women with breast cancer
.
J Natl Compr Cancer Netw
2017
;
15
:
1216
23
.
8.
Roh
MS
,
Colangelo
LH
,
O'Connell
MJ
,
Yothers
G
,
Deutsch
M
,
Allegra
CJ
, et al
Preoperative multimodality therapy improves disease-free survival in patients with carcinoma of the rectum: NSABP R-03
.
J Clin Oncol
2009
;
27
:
5124
30
.
9.
Minsky
BD
,
Cohen
AM
,
Kemeny
N
,
Enker
WE
,
Kelsen
DP
,
Reichman
B
, et al
Enhancement of radiation-induced downstaging of rectal cancer by fluorouracil and high-dose leucovorin chemotherapy
.
J Clin Oncol
1992
;
10
:
79
84
.
10.
Mohiuddin
M
,
Hayne
M
,
Regine
WF
,
Hanna
N
,
Hagihara
PF
,
McGrath
P
, et al
Prognostic significance of postchemoradiation stage following preoperative chemotherapy and radiation for advanced/recurrent rectal cancers
.
Int J Radiat Oncol Biol Phys
2000
;
48
:
1075
80
.
11.
Kamran
SC
,
Mouw
KW
. 
Applying precision oncology principles in radiation oncology
.
JCO Precision Oncol
2018
;
2
:
1
23
.
12.
Hall
WA
,
Bergom
C
,
Thompson
RF
,
Baschnagel
AM
,
Vijayakumar
S
,
Willers
H
, et al
Precision oncology and genomically guided radiation therapy: a report from the American Society for radiation oncology/American association of physicists in medicine/national cancer institute precision medicine conference
.
Int J Radiat Oncol Biol Phys
2018
;
101
:
274
84
.
13.
Bristow
RG
,
Alexander
B
,
Baumann
M
,
Bratman
SV
,
Brown
JM
,
Camphausen
K
, et al
Combining precision radiotherapy with molecular targeting and immunomodulatory agents: a guideline by the American Society for Radiation Oncology
.
Lancet Oncol
2018
;
19
:
e240
e51
.
14.
Demaria
S
,
Coleman
CN
,
Formenti
SC
. 
Radiotherapy: changing the game in immunotherapy
.
Trends Cancer
2016
;
2
:
286
94
.
15.
Kirsch
DG
,
Diehn
M
,
Kesarwala
AH
,
Maity
A
,
Morgan
MA
,
Schwarz
JK
, et al
The future of radiobiology
.
J Natl Cancer Inst
2018
;
110
:
329
40
.
16.
Burnette
B
,
Fu
YX
,
Weichselbaum
RR
. 
The confluence of radiotherapy and immunotherapy
.
Front Oncol
2012
;
2
:
143
.
17.
Kwilas
AR
,
Donahue
RN
,
Bernstein
MB
,
Hodge
JW
. 
In the field: exploiting the untapped potential of immunogenic modulation by radiation in combination with immunotherapy for the treatment of cancer
.
Front Oncol
2012
;
2
:
104
.
18.
Rizvi
NA
,
Hellmann
MD
,
Snyder
A
,
Kvistborg
P
,
Makarov
V
,
Havel
JJ
, et al
Cancer immunology. Mutational landscape determines sensitivity to PD-1 blockade in non-small cell lung cancer
.
Science
2015
;
348
:
124
8
.
19.
Samstein
RM
,
Lee
CH
,
Shoushtari
AN
,
Hellmann
MD
,
Shen
R
,
Janjigian
YY
, et al
Tumor mutational load predicts survival after immunotherapy across multiple cancer types
.
Nat Genet
2019
;
51
:
202
6
.
20.
Sauer
R
,
Becker
H
,
Hohenberger
W
,
Rodel
C
,
Wittekind
C
,
Fietkau
R
, et al
Preoperative versus postoperative chemoradiotherapy for rectal cancer
.
N Engl J Med
2004
;
351
:
1731
40
.
21.
Andre
T
,
Boni
C
,
Mounedji-Boudiaf
L
,
Navarro
M
,
Tabernero
J
,
Hickish
T
, et al
Oxaliplatin, fluorouracil, and leucovorin as adjuvant treatment for colon cancer
.
N Engl J Med
2004
;
350
:
2343
51
.
22.
Van Allen
EM
,
Mouw
KW
,
Kim
P
,
Iyer
G
,
Wagle
N
,
Al-Ahmadie
H
, et al
Somatic ERCC2 mutations correlate with cisplatin sensitivity in muscle-invasive urothelial carcinoma
.
Cancer Discov
2014
;
4
:
1140
53
.
23.
Liu
D
,
Abbosh
P
,
Keliher
D
,
Reardon
B
,
Miao
D
,
Mouw
K
, et al
Mutational patterns in chemotherapy resistant muscle-invasive bladder cancer
.
Nat Commun
2017
;
8
:
2193
.
25.
Cibulskis
K
,
McKenna
A
,
Fennell
T
,
Banks
E
,
DePristo
M
,
Getz
G
. 
ContEst: estimating cross-contamination of human samples in next-generation sequencing data
.
Bioinformatics
2011
;
27
:
2601
2
.
26.
Lawrence
MS
,
Stojanov
P
,
Mermel
CH
,
Robinson
JT
,
Garraway
LA
,
Golub
TR
, et al
Discovery and saturation analysis of cancer genes across 21 tumour types
.
Nature
2014
;
505
:
495
501
.
27.
Cibulskis
K
,
Lawrence
MS
,
Carter
SL
,
Sivachenko
A
,
Jaffe
D
,
Sougnez
C
, et al
Sensitive detection of somatic point mutations in impure and heterogeneous cancer samples
.
Nat Biotechnol
2013
;
31
:
213
9
.
28.
Saunders
CT
,
Wong
WS
,
Swamy
S
,
Becq
J
,
Murray
LJ
,
Cheetham
RK
. 
Strelka: accurate somatic small-variant calling from sequenced tumor-normal sample pairs
.
Bioinformatics
2012
;
28
:
1811
7
.
29.
Ramos
AH
,
Lichtenstein
L
,
Gupta
M
,
Lawrence
MS
,
Pugh
TJ
,
Saksena
G
, et al
Oncotator: cancer variant annotation tool
.
Hum Mutat
2015
;
36
:
E2423
9
.
30.
Robinson
JT
,
Thorvaldsdottir
H
,
Winckler
W
,
Guttman
M
,
Lander
ES
,
Getz
G
, et al
Integrative genomics viewer
.
Nat Biotechnol
2011
;
29
:
24
6
.
31.
Thorvaldsdottir
H
,
Robinson
JT
,
Mesirov
JP
. 
Integrative Genomics Viewer (IGV): high-performance genomics data visualization and exploration
.
Brief Bioinform
2013
;
14
:
178
92
.
32.
Le
X
,
Antony
R
,
Razavi
P
,
Treacy
DJ
,
Luo
F
,
Ghandi
M
, et al
Systematic functional characterization of resistance to PI3K inhibition in breast cancer
.
Cancer Discov
2016
;
6
:
1134
47
.
33.
Rooney
MS
,
Shukla
SA
,
Wu
CJ
,
Getz
G
,
Hacohen
N
. 
Molecular and genetic properties of tumors associated with local immune cytolytic activity
.
Cell
2015
;
160
:
48
61
.
34.
Nielsen
M
,
Lundegaard
C
,
Blicher
T
,
Lamberth
K
,
Harndahl
M
,
Justesen
S
, et al
NetMHCpan, a method for quantitative predictions of peptide binding to any HLA-A and -B locus protein of known sequence
.
PLoS One
2007
;
2
:
e796
.
35.
Carter
SL
,
Cibulskis
K
,
Helman
E
,
McKenna
A
,
Shen
H
,
Zack
T
, et al
Absolute quantification of somatic DNA alterations in human cancer
.
Nat Biotechnol
2012
;
30
:
413
21
.
36.
Brastianos
PK
,
Carter
SL
,
Santagata
S
,
Cahill
DP
,
Taylor-Weiner
A
,
Jones
RT
, et al
Genomic characterization of brain metastases reveals branched evolution and potential therapeutic targets
.
Cancer Discov
2015
;
5
:
1164
77
.
37.
Rodrigues
DN
,
Rescigno
P
,
Liu
D
,
Yuan
W
,
Carreira
S
,
Lambros
MB
, et al
Immunogenomic analyses associate immunological alterations with mismatch repair defects in prostate cancer
.
J Clin Invest
2018
;
128
:
5185
.
38.
Johnson
WE
,
Li
C
,
Rabinovic
A
. 
Adjusting batch effects in microarray expression data using empirical Bayes methods
.
Biostatistics
2007
;
8
:
118
27
.
39.
Chakraborty
S
,
Datta
S
,
Datta
S
. 
Surrogate variable analysis using partial least squares (SVA-PLS) in gene expression studies
.
Bioinformatics
2012
;
28
:
799
806
.
40.
Subramanian
A
,
Tamayo
P
,
Mootha
VK
,
Mukherjee
S
,
Ebert
BL
,
Gillette
MA
, et al
Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles
.
Proc Natl Acad Sci U S A
2005
;
102
:
15545
50
.
41.
Newman
AM
,
Liu
CL
,
Green
MR
,
Gentles
AJ
,
Feng
W
,
Xu
Y
, et al
Robust enumeration of cell subsets from tissue expression profiles
.
Nat Methods
2015
;
12
:
453
7
.
42.
Lennerz
JK
,
Kim
SH
,
Oates
EL
,
Huh
WJ
,
Doherty
JM
,
Tian
X
, et al
The transcription factor MIST1 is a novel human gastric chief cell marker whose expression is lost in metaplasia, dysplasia, and carcinoma
.
Am J Pathol
2010
;
177
:
1514
33
.
43.
Lehe
C
,
Ghebeh
H
,
Al-Sulaiman
A
,
Al Qudaihi
G
,
Al-Hussein
K
,
Almohareb
F
, et al
The Wilms' tumor antigen is a novel target for human CD4+ regulatory T cells: implications for immunotherapy
.
Cancer Res
2008
;
68
:
6350
9
.
44.
Maruse
Y
,
Kawano
S
,
Jinno
T
,
Matsubara
R
,
Goto
Y
,
Kaneko
N
, et al
Significant association of increased PD-L1 and PD-1 expression with nodal metastasis and a poor prognosis in oral squamous cell carcinoma
.
Int J Oral Maxillofac Surg
2018
;
47
:
836
45
.
45.
Pardoll
DM
. 
The blockade of immune checkpoints in cancer immunotherapy
.
Nat Rev Cancer
2012
;
12
:
252
64
.
46.
Steele
KE
,
Tan
TH
,
Korn
R
,
Dacosta
K
,
Brown
C
,
Kuziora
M
, et al
Measuring multiple parameters of CD8+ tumor-infiltrating lymphocytes in human cancers by image analysis
.
J Immunother Cancer
2018
;
6
:
20
.
47.
Zheng
B
,
Ren
T
,
Huang
Y
,
Sun
K
,
Wang
S
,
Bao
X
, et al
PD-1 axis expression in musculoskeletal tumors and antitumor effect of nivolumab in osteosarcoma model of humanized mouse
.
J Hematol Oncol
2018
;
11
:
16
.
48.
Anagnostou
V
,
Smith
KN
,
Forde
PM
,
Niknafs
N
,
Bhattacharya
R
,
White
J
, et al
Evolution of neoantigen landscape during immune checkpoint blockade in non-small cell lung cancer
.
Cancer Discov
2017
;
7
:
264
76
.
49.
Chen
PL
,
Roh
W
,
Reuben
A
,
Cooper
ZA
,
Spencer
CN
,
Prieto
PA
, et al
Analysis of immune signatures in longitudinal tumor samples yields insight into biomarkers of response and mechanisms of resistance to immune checkpoint blockade
.
Cancer Discov
2016
;
6
:
827
37
.
50.
Reuben
A
,
Gittelman
R
,
Gao
J
,
Zhang
J
,
Yusko
EC
,
Wu
CJ
, et al
TCR repertoire intratumor heterogeneity in localized lung adenocarcinomas: an association with predicted neoantigen heterogeneity and postsurgical recurrence
.
Cancer Discov
2017
;
7
:
1088
97
.
51.
Baker
SJ
,
Preisinger
AC
,
Jessup
JM
,
Paraskeva
C
,
Markowitz
S
,
Willson
JK
, et al
p53 gene mutations occur in combination with 17p allelic deletions as late events in colorectal tumorigenesis
.
Cancer Res
1990
;
50
:
7717
22
.
52.
Soussi
T
. 
The p53 tumor suppressor gene: from molecular biology to clinical investigation
.
Ann
N Y
Acad Sci
2000
;
910
:
121
37
.
53.
Takayama
T
,
Ohi
M
,
Hayashi
T
,
Miyanishi
K
,
Nobuoka
A
,
Nakajima
T
, et al
Analysis of K-ras, APC, and beta-catenin in aberrant crypt foci in sporadic adenoma, cancer, and familial adenomatous polyposis
.
Gastroenterology
2001
;
121
:
599
611
.
54.
Vogelstein
B
,
Fearon
ER
,
Hamilton
SR
,
Kern
SE
,
Preisinger
AC
,
Leppert
M
, et al
Genetic alterations during colorectal-tumor development
.
N Engl J Med
1988
;
319
:
525
32
.
55.
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
:
djx031
.
56.
Wang
M
,
Han
J
,
Marcar
L
,
Black
J
,
Liu
Q
,
Li
X
, et al
Radiation resistance in KRAS-mutated lung cancer is enabled by stem-like properties mediated by an osteopontin-EGFR pathway
.
Cancer Res
2017
;
77
:
2018
28
.
57.
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
.
58.
Faltas
BM
,
Prandi
D
,
Tagawa
ST
,
Molina
AM
,
Nanus
DM
,
Sternberg
C
, et al
Clonal evolution of chemotherapy-resistant urothelial carcinoma
.
Nat Genet
2016
;
48
:
1490
9
.
59.
Gerlinger
M
,
Rowan
AJ
,
Horswell
S
,
Math
M
,
Larkin
J
,
Endesfelder
D
, et al
Intratumor heterogeneity and branched evolution revealed by multiregion sequencing
.
N Engl J Med
2012
;
366
:
883
92
.
60.
Johnson
BE
,
Mazor
T
,
Hong
C
,
Barnes
M
,
Aihara
K
,
McLean
CY
, et al
Mutational analysis reveals the origin and therapy-driven evolution of recurrent glioma
.
Science
2014
;
343
:
189
93
.
61.
Patch
AM
,
Christie
EL
,
Etemadmoghadam
D
,
Garsed
DW
,
George
J
,
Fereday
S
, et al
Whole-genome characterization of chemoresistant ovarian cancer
.
Nature
2015
;
521
:
489
94
.
62.
Agostini
M
,
Zangrando
A
,
Pastrello
C
,
D'Angelo
E
,
Romano
G
,
Giovannoni
R
, et al
A functional biological network centered on XRCC3: a new possible marker of chemoradiotherapy resistance in rectal cancer patients
.
Cancer Biol Ther
2015
;
16
:
1160
71
.
63.
Kim
IJ
,
Lim
SB
,
Kang
HC
,
Chang
HJ
,
Ahn
SA
,
Park
HW
, et al
Microarray gene expression profiling for predicting complete response to preoperative chemoradiotherapy in patients with advanced rectal cancer
.
Dis Colon Rectum
2007
;
50
:
1342
53
.
64.
Rimkus
C
,
Friederichs
J
,
Boulesteix
AL
,
Theisen
J
,
Mages
J
,
Becker
K
, et al
Microarray-based prediction of tumor response to neoadjuvant radiochemotherapy of patients with locally advanced rectal cancer
.
Clin Gastroenterol Hepatol
2008
;
6
:
53
61
.
65.
Akiyoshi
T
,
Kobunai
T
,
Watanabe
T
. 
Predicting the response to preoperative radiation or chemoradiation by a microarray analysis of the gene expression profiles in rectal cancer
.
Surg Today
2012
;
42
:
713
9
.
66.
Cecchin
E
,
Agostini
M
,
Pucciarelli
S
,
De Paoli
A
,
Canzonieri
V
,
Sigon
R
, et al
Tumor response is predicted by patient genetic profile in rectal cancer patients treated with neo-adjuvant chemo-radiotherapy
.
Pharmacogenomics J
2011
;
11
:
214
26
.
67.
Chen
Z
,
Liu
Z
,
Li
W
,
Qu
K
,
Deng
X
,
Varma
MG
, et al
Chromosomal copy number alterations are associated with tumor response to chemoradiation in locally advanced rectal cancer
.
Genes Chromosomes Cancer
2011
;
50
:
689
99
.
68.
Gantt
GA
,
Chen
Y
,
Dejulius
K
,
Mace
AG
,
Barnholtz-Sloan
J
,
Kalady
MF
. 
Gene expression profile is associated with chemoradiation resistance in rectal cancer
.
Colorectal Dis
2014
;
16
:
57
66
.
69.
Grade
M
,
Gaedcke
J
,
Wangsa
D
,
Varma
S
,
Beckmann
J
,
Liersch
T
, et al
Chromosomal copy number changes of locally advanced rectal cancers treated with preoperative chemoradiotherapy
.
Cancer Genet Cytogenet
2009
;
193
:
19
28
.
70.
Ho-Pun-Cheung
A
,
Assenat
E
,
Bascoul-Mollevi
C
,
Bibeau
F
,
Boissiere-Michot
F
,
Thezenas
S
, et al
A large-scale candidate gene approach identifies SNPs in SOD2 and IL13 as predictive markers of response to preoperative chemoradiation in rectal cancer
.
Pharmacogenomics J
2011
;
11
:
437
43
.
71.
Hur
H
,
Kang
J
,
Kim
NK
,
Min
BS
,
Lee
KY
,
Shin
SJ
, et al
Thymidylate synthase gene polymorphism affects the response to preoperative 5-fluorouracil chemoradiation therapy in patients with rectal cancer
.
Int J Radiat Oncol Biol Phys
2011
;
81
:
669
76
.
72.
Kim
JC
,
Ha
YJ
,
Roh
SA
,
Cho
DH
,
Choi
EY
,
Kim
TW
, et al
Novel single-nucleotide polymorphism markers predictive of pathologic response to preoperative chemoradiation therapy in rectal cancer patients
.
Int J Radiat Oncol Biol Phys
2013
;
86
:
350
7
.
73.
Nishioka
M
,
Shimada
M
,
Kurita
N
,
Iwata
T
,
Morimoto
S
,
Yoshikawa
K
, et al
Gene expression profile can predict pathological response to preoperative chemoradiotherapy in rectal cancer
.
Cancer Genomics and Proteomics
2011
;
8
:
87
92
.
74.
Watanabe
T
,
Kobunai
T
,
Akiyoshi
T
,
Matsuda
K
,
Ishihara
S
,
Nozawa
K
. 
Prediction of response to preoperative chemoradiotherapy in rectal cancer by using reverse transcriptase polymerase chain reaction analysis of four genes
.
Dis Colon Rectum
2014
;
57
:
23
31
.
75.
Watanabe
T
,
Komuro
Y
,
Kiyomatsu
T
,
Kanazawa
T
,
Kazama
Y
,
Tanaka
J
, et al
Prediction of sensitivity of rectal cancer cells in response to preoperative radiotherapy by DNA microarray analysis of gene expression profiles
.
Cancer Res
2006
;
66
:
3370
4
.
76.
Giordano
FA
,
Veldwijk
MR
,
Herskind
C
,
Wenz
F
. 
Radiotherapy, tumor mutational burden, and immune checkpoint inhibitors: time to do the math
.
Strahlenther Onkol
2018
;
194
:
873
5
.
77.
Helleday
T
. 
Making immunotherapy ‘cold’ tumours ‘hot’ by chemotherapy-induced mutations - a misconception
.
Ann Oncol
2019
;
30
:
360
1
.
78.
Hanada
T
,
Nakagawa
M
,
Emoto
A
,
Nomura
T
,
Nasu
N
,
Nomura
Y
. 
Prognostic value of tumor-associated macrophage count in human bladder cancer
.
Int J Urol
2000
;
7
:
263
9
.
79.
Leek
RD
,
Lewis
CE
,
Whitehouse
R
,
Greenall
M
,
Clarke
J
,
Harris
AL
. 
Association of macrophage infiltration with angiogenesis and prognosis in invasive breast carcinoma
.
Cancer Res
1996
;
56
:
4625
9
.
80.
Lin
EY
,
Li
JF
,
Gnatovskiy
L
,
Deng
Y
,
Zhu
L
,
Grzesik
DA
, et al
Macrophages regulate the angiogenic switch in a mouse model of breast cancer
.
Cancer Res
2006
;
66
:
11238
46
.
81.
Hugo
H
,
Ackland
ML
,
Blick
T
,
Lawrence
MG
,
Clements
JA
,
Williams
ED
, et al
Epithelial–mesenchymal and mesenchymal–epithelial transitions in carcinoma progression
.
J Cell Physiol
2007
;
213
:
374
83
.
82.
Theys
J
,
Jutten
B
,
Habets
R
,
Paesmans
K
,
Groot
AJ
,
Lambin
P
, et al
E-Cadherin loss associated with EMT promotes radioresistance in human tumor cells
.
Radiother Oncol
2011
;
99
:
392
7
.
83.
Valastyan
S
,
Weinberg
RA
. 
Tumor metastasis: molecular insights and evolving paradigms
.
Cell
2011
;
147
:
275
92
.
84.
Zhang
H
,
Luo
H
,
Jiang
Z
,
Yue
J
,
Hou
Q
,
Xie
R
, et al
Fractionated irradiation-induced EMT-like phenotype conferred radioresistance in esophageal squamous cell carcinoma
.
J Radiat Res
2016
;
57
:
370
80
.
85.
Zhou
P
,
Li
B
,
Liu
F
,
Zhang
M
,
Wang
Q
,
Liu
Y
, et al
The epithelial to mesenchymal transition (EMT) and cancer stem cells: implication for treatment resistance in pancreatic cancer
.
Mol Cancer
2017
;
16
:
52
.
86.
Rose
BS
,
Winer
EP
,
Mamon
HJ
. 
Perils of the pathologic complete response
.
J Clin Oncol
2016
;
34
:
3959
62
.
87.
Formenti
SC
,
Rudqvist
NP
,
Golden
E
,
Cooper
B
,
Wennerberg
E
,
Lhuillier
C
, et al
Radiotherapy induces responses of lung cancer to CTLA-4 blockade
.
Nat Med
2018
;
24
:
1845
51
.
88.
van Gijn
W
,
Marijnen
CA
,
Nagtegaal
ID
,
Kranenbarg
EM
,
Putter
H
,
Wiggers
T
, et al
Preoperative radiotherapy combined with total mesorectal excision for resectable rectal cancer: 12-year follow-up of the multicentre, randomised controlled TME trial
.
Lancet Oncol
2011
;
12
:
575
82
.