Somatic Mutations and Immune Alternation in Rectal Cancer ......8 Department of Gastrointestinal...

54
1 Somatic Mutations and Immune Alternation in Rectal Cancer Following 1 Neoadjuvant Chemoradiotherapy 2 3 Dengbo Ji 1* , Haizhao Yi 1, 2* , Dakui Zhang 3* , Tiancheng Zhan 1 , Zhaowei Li 1 , Ming Li 1 , 4 Jinying Jia 1 , Meng Qiao 1 , Jinhong Xia 1 , Zhiwei Zhai 4 , Can Song 5, 6 , and Jin Gu 1, 6, 7§ 5 6 1 Key laboratory of Carcinogenesis and Translational ResearchMinistry of Education), 7 Department of Gastrointestinal Surgery III, Peking University Cancer Hospital & 8 Institute, No. 52 Fucheng Rd., Haidian District, Beijing, China, 100142 9 2 Department of General Surgery 1, Affiliated Hospital of Chengde Medical College, No. 10 36 Nanyingzi Rd., Chengde, China, 067000 11 3 Department of colorectal surgery, China-Japan Friendship Hospital, 100029 12 4 Department of Gastrointestinal Surgery, Chaoyang Hospital, Beijing, China, 100020 13 5 School of Life Sciences, Tsinghua University, Beijing, China, 100084 14 6 Peking-Tsinghua Center for Life Sciences 15 7 Peking University S.G. Hospital 16 17 * These authors contributed equally to this study. 18 Running title: nCRT enables rectal cancer checkpoint blockade therapy 19 Keywords: Rectal Cancer; Neoadjuvant Chemoradiotherapy; Mutation Burden; 20 Checkpoint Blockade Therapy; Immune Activation 21 Grant Support: This work was supported by the National Natural Science 22 on April 7, 2020. © 2018 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on October 3, 2018; DOI: 10.1158/2326-6066.CIR-17-0630

Transcript of Somatic Mutations and Immune Alternation in Rectal Cancer ......8 Department of Gastrointestinal...

1  

Somatic Mutations and Immune Alternation in Rectal Cancer Following 1 

Neoadjuvant Chemoradiotherapy 2 

Dengbo Ji1*, Haizhao Yi1, 2*, Dakui Zhang3*, Tiancheng Zhan1, Zhaowei Li1, Ming Li1, 4 

Jinying Jia1, Meng Qiao1, Jinhong Xia1, Zhiwei Zhai4, Can Song5, 6, and Jin Gu1, 6, 7§ 5 

1Key laboratory of Carcinogenesis and Translational Research(Ministry of Education), 7 

Department of Gastrointestinal Surgery III, Peking University Cancer Hospital & 8 

Institute, No. 52 Fucheng Rd., Haidian District, Beijing, China, 100142 9 

2 Department of General Surgery 1, Affiliated Hospital of Chengde Medical College, No. 10 

36 Nanyingzi Rd., Chengde, China, 067000 11 

3 Department of colorectal surgery, China-Japan Friendship Hospital, 100029 12 

4 Department of Gastrointestinal Surgery, Chaoyang Hospital, Beijing, China, 100020 13 

5School of Life Sciences, Tsinghua University, Beijing, China, 100084 14 

6Peking-Tsinghua Center for Life Sciences 15 

7Peking University S.G. Hospital 16 

17 

*These authors contributed equally to this study. 18 

Running title: nCRT enables rectal cancer checkpoint blockade therapy 19 

Keywords: Rectal Cancer; Neoadjuvant Chemoradiotherapy; Mutation Burden; 20 

Checkpoint Blockade Therapy; Immune Activation 21 

Grant Support: This work was supported by the National Natural Science 22 

on April 7, 2020. © 2018 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on October 3, 2018; DOI: 10.1158/2326-6066.CIR-17-0630

2  

Foundation (81772565 to D.B. J, 81372593 to J G, 81201965 to D.B. J), Beijing 23 

Natural Science Foundation (7132052 to J G), and the National High Technology 24 

Research and Development Program of China (863 Program) (No.2012AA02A506 25 

to J G partially , 2014AA020801 to M L partially). 26 

§Correspondence to: 27 

Jin Gu, MD; FACS 28 

Key laboratory of Carcinogenesis and Translational Research (Ministry of 29 

Education), Department of Gastrointestinal Surgery III, Peking University Cancer 30 

Hospital & Institute, No. 52 Fucheng Road, Haidian District, Beijing, China, 100142, 31 

Tel/Fax: +86-10-88196238, e-mail: [email protected] 32 

Disclosure of Potential Conflicts of Interest: 33 

The authors declare no competing financial interests. 34 

Word Counts: Abstract 214; Body 5,484 35 

Figures: 7 Figures; 1 Table; 10 Suppl. Figures; 12 Suppl. Tables 36 

37 

38 

39 

40 

41 

42 

43 

44 

45 

46 

47 

48 

on April 7, 2020. © 2018 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on October 3, 2018; DOI: 10.1158/2326-6066.CIR-17-0630

3  

49 

Abstract 50 

Checkpoint blockade therapy triggers tumor-specific immune responses in a variety 51 

of cancer types. We presumed that rectal cancer patients could have become 52 

sensitive to immunotherapy after receiving neoadjuvant chemoradiotherapy (nCRT). 53 

In this study, we report immune alternation in post-nCRT patients compared to 54 

pretreatment conditions from GEO data. Whole exome sequencing of 14 locally 55 

advanced rectal cancer (LARC) patient samples showed that nCRT induced new 56 

mutations compared to the paired pre-treatment biopsies evidenced by appearance of 57 

a neoantigen landscape. An association was identified between mutation burden and 58 

enrichment of immune activation-related pathways. Animal experiment results 59 

further demonstrated that radiotherapy enhanced the efficacy of anti–PD-1. 60 

Mutation burden and the neoantigens of LARC patients were associated with 61 

response to nCRT. The mRNA expression profiling of 66 pre-treatment biopsy 62 

samples from LARC patients showed that immune activation-related pathways were 63 

enriched in response to nCRT. PD-L1 expression was negatively correlated with 64 

disease-free survival in the CD8-low expression group who received nCRT in a 65 

cohort of 296 samples. Thus, nCRT was able to alter immune function in LARC 66 

patients, which may be associated with the appearance of neoantigens. Neoantigens 67 

could make rectal cancer patients potential candidates to receive checkpoint 68 

blockade immunotherapy, and mutation burden could be a useful biomarker to 69 

stratify patients into responding and non-responding groups for immunotherapy. 70 

71 

on April 7, 2020. © 2018 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on October 3, 2018; DOI: 10.1158/2326-6066.CIR-17-0630

4  

Introduction 72 

The treatment guidelines, published by the National Comprehensive Cancer Network 73 

(NCCN) of the United States, clearly states that pre-operative neoadjuvant 74 

chemoradiotherapy is crucial for rectal cancer treatment, which can improve the rate 75 

of curative resection and significantly reduce local recurrence (1,2). Although local 76 

recurrence and overall survival have improved, distant recurrence rate has not 77 

decreased significantly. About 30% of patients treated with a curative regiment will 78 

eventually develop distant metastases (3-5). Adjuvant drug therapy has been used to 79 

prevent distant metastases by eliminating circulating tumor cells and 80 

micrometastases. However, the use of adjuvant chemotherapy for patients with rectal 81 

cancer treated with pre-operative chemoradiotherapy and surgery is still 82 

controversial (6). Administration of adjuvant chemotherapy to patients with stage II 83 

or III rectal cancer was based on results from phase 3 trials of adjuvant treatment for 84 

colon cancer (7-10) , as well as from trials in patients with rectal cancer who were 85 

treated without pre-operative chemoradiotherapy (11). Fluorouracil-based adjuvant 86 

chemotherapy did not improve overall survival, disease-free survival, or distant 87 

recurrences.(6,12) 88 

Colorectal cancers (CRCs) with a high density of tumor-infiltrating lymphocytes 89 

(TILs), especially CD8+ T lymphocytes, are associated with a better prognosis 90 

(13-16), suggesting that a cytotoxic antitumor immune response is involved in 91 

controlling cancer progression. Checkpoint blockade immunotherapy has improved 92 

cancer treatment. Along with radical surgery, radiation therapy, chemotherapy, and 93 

on April 7, 2020. © 2018 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on October 3, 2018; DOI: 10.1158/2326-6066.CIR-17-0630

5  

targeted oncogene treatment, checkpoint blockade is on the top of the list of 94 

therapeutic options. Checkpoint blockade therapy utilizes monoclonal antibodies 95 

(mAbs) to rescue suppressed T cells through activating and restoring their antitumor 96 

activity (17). Similarly, targeting cytotoxic T lymphocyte–associated antigen 4 97 

(CTLA-4) and programmed cell-death protein-1 (PD-1) pathways in metastatic 98 

melanoma, non–small-cell lung cancer (NSCLC), and other malignancies has 99 

significantly prolonged survival (18-20). 100 

Despite revolutionary achievements, the efficacy of checkpoint blockade 101 

immunotherapy varies among different tumor types, and a few cancer types, such as 102 

CRC, appear to be refractory to this therapy (19,21). A notable exception is that 103 

patients with mismatch repair (MMR)-deficient CRC lesions obtain clinical benefits 104 

from the administration of anti–PD-1 (22). 105 

Increasing evidence indicates that antitumor effects, clinically noted with checkpoint 106 

inhibitors such as ipilimumab, may rely on boosting tumor-specific immune 107 

responses that were pre-existing or newly induced. High somatic mutation loads are 108 

correlated with responsiveness to PD-1 blockade therapy in NSCLC, melanoma, and 109 

MMR-deficient CRCs (22-24). Research has revealed that radiation can not only 110 

reduce tumor burden but also enhance antitumor immune responses to tumor cells 111 

(25). The combination of radiation therapy, anti–CTLA-4, and anti–PD-L1 promotes 112 

clinical responses in melanoma (26). 113 

Therefore, we hypothesized that rectal cancer patients could be potential candidates 114 

for checkpoint blockade immunotherapy after receiving nCRT. To test this 115 

on April 7, 2020. © 2018 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on October 3, 2018; DOI: 10.1158/2326-6066.CIR-17-0630

6  

hypothesis, whole-exome sequencing of rectal cancer pre- and post- nCRT samples 116 

was performed to analyze the mutational landscape differences. We further 117 

performed an integrative analysis of mutational landscape and gene expression using 118 

TCGA and GSE data to evaluate the influence of somatic mutations and its 119 

association with immune response induced by treatment. 120 

121 

Methods and Materials 122 

Patients and samples 123 

Sample collection and usage was approved by the Ethics Review Committees of 124 

Peking University Cancer Hospital & Institute and in accordance with the Declaration 125 

of Helsinki. All patients were informed prior to the study and a consent form was 126 

signed by each participant. 127 

Tumor and paired normal adjacent tissue samples from cohort 1 included 14 LARC 128 

patients who received nCRT and were used for whole-exome sequencing. 9 paired 129 

LARC tumor tissues were obtained pre- and post-nCRT, and the remaining patients 130 

only had pre-therapy biopsies due to complete pathological response (pCR) or near 131 

pCR after nCRT. Pre-treatment blood samples from cohort 2 consisted of 42 LARC 132 

patients who received nCRT and were used for circulating tumor DNA (ctDNA) 133 

extraction and targeted ctDNA sequencing. The normal adjacent biopsies from 134 

patients of cohort 2 were obtained for tissues DNA extraction and used as control. All 135 

the tissue and blood samples were from patients who received nCRT and surgical 136 

resections between 2014 and 2016 at the Peking University Cancer Hospital & 137 

on April 7, 2020. © 2018 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on October 3, 2018; DOI: 10.1158/2326-6066.CIR-17-0630

7  

Institute (Beijing, China). Pre-treatment tumor staging was performed in all patients 138 

using endorectal ultrasonography, pelvic magnetic resonance imaging, or computed 139 

tomography. The radiotherapy regimen consisted of a 50.6 Gy dose delivered in 22 140 

fractions with concurrent capecitabine treatment at a dose of 825 mg/m2 orally twice 141 

per day for 5 weeks. 142 

Inclusion criteria were: 1. diagnosis of rectal adenocarcinoma by biopsy; 2. tumor 143 

staged as T3-4 or any T, N+ by endorectal ultrasonography, pelvic magnetic 144 

resonance imaging, or computed tomography. Exclusion criteria were: 1. previous 145 

chemotherapy or pelvic radiation; 2. presence of any other malignant disorders or 146 

other chronic diseases. For cohort 1, biopsies pre nCRT collected by rectoscopy and 147 

tissue samples collected by surgical resection were stored immediately in frozen in 148 

liquid nitrogen and stored at −80°C until use. For cohort 2, 3.5 mL of venous blood 149 

was collected prior to nCRT from each patient and processed within 1 h based on 150 

protocols of NCI’s Early Detection Research Networks (EDRN). Plasma was 151 

harvested after centrifugation twice at 1,600g for 10min aliquoted immediately for 152 

ctDNA extraction or stored at -80 C. 153 

In order to generate differential mRNA profiling between responders and 154 

non-responders to nRT and evaluate the influence of PD-1 and PD-L1 on prognosis 155 

after neoadjuvant therapy, large-scale cohorts of pre-therapy biopsy and cancerous 156 

tissue samples were selected and categorized into cohorts 3, 4, and 5. Cohort 3 was 157 

composed of biopsy samples from 66 patients before nRT and resection. All patients 158 

were treated with intermediate-fraction nRT (30 Gy/10 fractions) followed by a total 159 

on April 7, 2020. © 2018 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on October 3, 2018; DOI: 10.1158/2326-6066.CIR-17-0630

8  

mesorectal excision (TME) surgery. The patients achieving Grade 0 and 1 evaluated 160 

using tumor regression grade (TRG) system were defined as responders. The patients 161 

with Grade 3 were defined as non-responders. Inclusion criteria were: 1. diagnosis of 162 

rectal adenocarcinoma by biopsy; 2. tumor staged as T3-4 or any T, N+ by endorectal 163 

ultrasonography, pelvic magnetic resonance imaging, or computed tomography; 3. no 164 

evidence of distant metastasis. Patients with the following characteristics were 165 

excluded: 1. previous chemotherapy or pelvic radiation; 2. presence of any other 166 

malignant disorders or other chronic diseases. Biopsies were collected by rectoscopy 167 

and stored immediately in RNAlater (Qiagen) and then stored at −80°C until use. 168 

Cohort 4 consisted of 294 CRC patients who did not receive nRT, and the samples 169 

were collected after surgical resection. Patients with stage I-IV colorectal cancer, and 170 

with clinicopathological characteristics and follow-up information available, were 171 

included. We excluded patients if they had any other malignant disorders or other 172 

chronic diseases, previous treatment with any anticancer therapy, presence of any 173 

tumor type other than adenocarcinoma or mucinous carcinoma, and familial 174 

adenomatous polyposis CRC. 175 

Cohort 5 included 296 samples from patients who received nRT, and the samples 176 

were collected after resection. The radiation dosage was 30 Gy and delivered in 10 177 

fractions over 2 weeks. Inclusion criteria were: 1. diagnosis of rectal 178 

adenocarcinoma by biopsy; 2. tumor staged as T3-4 or any T, N+ by endorectal 179 

ultrasonography, pelvic magnetic resonance imaging, or computed tomography; 3. 180 

no evidence of distant metastasis. Patients with the following characteristics were 181 

on April 7, 2020. © 2018 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on October 3, 2018; DOI: 10.1158/2326-6066.CIR-17-0630

9  

excluded: 1. previous chemotherapy or pelvic radiation; 2. presence of any other 182 

malignant disorders or other chronic diseases. For cohort 4 and 5, tumor tissue 183 

samples were directly collected after surgical resection. All samples were 184 

immediately frozen in liquid nitrogen and stored at −80°C or fixed in 10% formalin 185 

for paraffin embedding. 186 

A summary of the clinical characteristics of these patients is shown in 187 

Supplementary Table S1-5. 188 

189 

Assessment of treatment response and tumor downstaging 190 

The 7th edition of the American Joint Committee on Cancer TNM system was used 191 

for Pathological staging(27). Neoadjuvant radiotherapy effect was evaluated after 192 

surgery by specialized gastrointestinal pathologists using tumor regression grade 193 

(TRG) system as follows: Grade 0: complete regression, no tumor cells; Grade 1: 194 

single or small groups of tumor cells, moderate response; Grade 2: residual cancer 195 

outgrown by fibrosis, minimal response; Grade 3: minimal or no tumor cells killed, 196 

poor response. 197 

198 

Whole-exome capture and sequencing 199 

Sample preparation, library construction, exome capture, next generation sequencing, 200 

and bioinformatics analysis were performed at Shanghai Biotechnology Corporation. 201 

(Shanghai, China). Genomic DNA from cohort 1 tumor samples was randomly 202 

fragmented and used to construct an in vitro shotgun library. For each sample to be 203 

on April 7, 2020. © 2018 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on October 3, 2018; DOI: 10.1158/2326-6066.CIR-17-0630

10  

sequenced, individual library preparations, hybridizations, and captures were 204 

performed following the protocol of SureSelectXT Target Enrichment System for 205 

Illumina Paired-End Sequencing Library (Agilent Technologies, Inc. 5301 Stevens 206 

Creek Rd Santa Clara, CA95051 USA). The library fragments were flanked by index 207 

adaptors following end repair and A-tailing addition. The library was further 208 

enriched with biotinylated probes (SureSelect biotinylated library mix) for sequences 209 

corresponding to exons through aqueous-phase hybridization capture. The 210 

hybridized fragments were captured by streptavidin-based magnetic beads 211 

(Dynabeads MyOne Streptavidin T1, Life Technologies, cat#65603), followed by 212 

amplification, quality inspection, and massive parallel sequencing of the enriched 213 

library. Quantity of library was assessed with Qubit 2.0 Fluoromete. The quality and 214 

size range was assessed using 2100 Bioanalyzer High Sensitivity DNA Assay as 215 

instructed in the reagent kit guide. BaseCalls directory, containing the binary base 216 

call files (BCL files), was generated by Real Time Analysis (RTA, Illumina, 217 

California, USA). The bcl2fastq (v1.8.3, Illumina, California, USA) was used to 218 

combine the per-cycle BCL files in each run and translate them into FASTQ files. 219 

The FASTQ files were aligned to a human reference genome (hg19) by 220 

Burrows-Wheeler Aligner (BWA, v0.7.12,(28,29) (Sanger Institute, Cambridge, 221 

UK). The aligned files (SAM/BAM format files) were initially sorted by SAM tools 222 

(30)(v0.1.19, Sanger Institute, Cambridge, UK). The aligned read duplicating the 223 

start position of another read was flagged as a duplicate (‘Mark duplicate’) by using 224 

Picard Tools (v1.107, Broad Institute, Cambridge, MA, USA). Data was processed 225 

on April 7, 2020. © 2018 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on October 3, 2018; DOI: 10.1158/2326-6066.CIR-17-0630

11  

using the Genome Analysis Toolkit (GATK, v3.1, Broad Institute, Cambridge, MA, 226 

USA). Reads were locally realigned (GATK IndelRealigner), and the base qualities 227 

were recalibrated (GATK BaseRecalibrator). Final mapping statistics, including 228 

coverage and depth, were generated from recalibrated files by BED tools(31) 229 

(v2.16.1, Quinlan Laboratory at the University of Virginia) and perl/python scripts. 230 

The readings were further formatted with SAM tools, duplication was removed with 231 

Picard, local realignment around InDels was processed by GATK InDelRealigner, 232 

and base quality score recalibration was performed by using GATK Base 233 

Recalibrator. Potential somatic substitutions were identified using GATK Unified 234 

Genotyper followed by variant annotation through Annovar/VEP/snpEFF. 235 

Somatic mutations were determined using MuTect2 (32)(Broad Institute, Cambridge, 236 

MA, USA ) to identify mutations in matched tumor and normal samples. A detailed 237 

MuTect2 procedure is available at http://www.broadinstitute.org/cancer/cga/mutect/. 238 

The paired sample data was finally filtered by ANNOVAR(33) (Wed, 17 Jun 2015, 239 

Center for Applied Genomics, Children’s Hospital of Philadelphia, Department of 240 

Biostatistics and Epidemiology and Department of Pediatrics, University of 241 

Pennsylvania, Philadelphia, PA, USA) for further analysis. 242 

243 

Targeted ctDNA sequencing 244 

Sample preparation, library construction, exome capture, next generation sequencing, 245 

and bioinformatics analysis were performed at Genecast Biotechnology Co., Ltd. 246 

(Beijing, China). Plasma from cohort 2 blood samples was harvested after 247 

on April 7, 2020. © 2018 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on October 3, 2018; DOI: 10.1158/2326-6066.CIR-17-0630

12  

centrifugation twice at 1,600 x g for 10 minutes and aliquoted immediately for ctDNA 248 

extraction or stored at -80°C. ctDNA was extracted from 2 mL of plasma using the 249 

MagMAXTM Cell-Free DNA isolation kit (Life Technologies, California, USA) and 250 

quantified by Qubit dsDNA HS Assay kit or Qubit dsDNA BR Assay kit (Life 251 

Technologies, California, USA). 252 

Genomic DNA was sheared into 150-200 bp fragments with Covaris M220 253 

Focused-ultrasonicatorTM Instrument (Covaris, Massachusetts, USA). Fragmented 254 

DNA and the ctDNA library were constructed by KAPA HTP Library Preparation Kit 255 

(Illumina platforms, KAPA Biosystems, Massachusetts, USA), following 256 

manufacturer’s instructions. DNA libraries were captured following NimbleGen 257 

SeqCap EZ Library SR (Roche, Wisconsin, USA) Users’ Guide, with a designed 0.8M 258 

size panel (Genecast Biotech, Beijing, China), which included 325 major 259 

tumor-related genes. The captured samples were subjected to Illumina HiSeq X-Ten 260 

for paired-end sequencing. 261 

Paired-end reads generated from Hiseq X-Ten platform were mapped to the hg19 262 

reference genome with BWA v0.7.12 (default parameters)(28,29)(Sanger Institute, 263 

Cambridge, UK), then sorted, filtered, and indexed with SAM tools(30)(1.3, Sanger 264 

Institute, Cambridge, UK). In order to identify somatic single nucleotide 265 

polymorphisms (SNPs) and indel mutations, the obtained BAM files from both 266 

plasma and matched normal tissues from each patient were processed for pairwise 267 

variant calling using VarScan (34)(v2.3.8, The Genome Institue, Washington 268 

University, St. Louis, Missouri, USA). Minimum coverage for calling somatic 269 

variants in matched normal tissues samples and plasma samples were 5 and 3, 270 

on April 7, 2020. © 2018 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on October 3, 2018; DOI: 10.1158/2326-6066.CIR-17-0630

13  

respectively. P-value threshold to call a somatic site was 0.05, and variants with 271 

<90% strand bias were kept for further study. The generated candidate mutations 272 

were annotated using ANNOVAR software (33)(Wed, 17 Jun 2015, Center for 273 

Applied Genomics, Children’s Hospital of Philadelphia, Department of Biostatistics 274 

and Epidemiology and Department of Pediatrics, University of Pennsylvania, 275 

Philadelphia, PA, USA), the dbNSFP and Exome Aggregation Consortum (ExAC) 276 

database were used to remove either the benign mutations with pp2_hdiv score < 277 

0.452 or the population polymorphic sites. Finally, the resulted nonsynonymous 278 

mutations at the exonic regions were reserved. Tumor mutation burden (TMB) was 279 

defined as the number of non-synonymous mutations per megabase of exonic region. 280 

281 

Mismatch repair status testing 282 

Mismatch repair status was assessed using the MSI Analysis System (Promega, 283 

Madison, WI), consisting of 5 pseudomonomorphic mononucleotide repeats (BAT-25, 284 

BAT-26, NR-21, NR-24, and MONO-27) to detect MSI and 2-pentanucleotide repeat 285 

loci (PentaC and PentaD). According to the manufacturer’s guidelines, 2ng template 286 

DNA for each sample was added into the Amplification Mix including nuclease-free 287 

water, gold ST*R buffer, MSI primer pair mix and AmpliTaq Gold DNA polymerase 288 

(Life Technologies Cat.# N8080242) and mix gently. For the positive amplification 289 

control, 2ng of the diluted K562 High Molecular Weight DNA was added into the 290 

Amplification Mix. For the negative amplification control, nuclease-fee water (instead 291 

of template DNA) was added into Amplification Mix. Amplification was performed 292 

in a GeneAmpR PCR System 9600 (Applied Biosystems). Amplification conditions 293 

were as follows: 95°C for 11 minutes, 96°C for 1 minute, then: 94°C for 30 seconds, 294 

on April 7, 2020. © 2018 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on October 3, 2018; DOI: 10.1158/2326-6066.CIR-17-0630

14  

ramp 68 seconds to 58°C, hold for 30 seconds, ramp 50 seconds to 70°C, hold for 1 295 

minute, for 10 cycles, then: 90°C for 30 seconds, ramp 60 seconds to 58°C, hold for 296 

30 seconds, ramp 50 seconds to 70°C, hold for 1 minute for 20 cycles, then: 60°C for 297 

30 minutes, 4°C soak. The primers used were provided in the MSI Analysis System 298 

(Promega, Madison, WI). Following amplification of DNA, the fluorescent PCR 299 

products were analyzed with the Applied Biosystems 3130xl Gene Analyzer using 300 

GeneScan analysis software (Applied Biosystems, Foster City, CA, USA). The length 301 

of the sequence was determined for each microsatellite locus, and the tumors were 302 

designated as high-frequency MSI (MSI-H) if two or more mononucleotide loci varies 303 

were identified in length compared to the germline DNA. One vary was considered as 304 

low-frequency MSI (MSI-L) and none as microsatellite stable (MSS). 305 

306 

RNA isolation and microarray analyses 307 

Total RNA from cohort 3 biopsy samples was isolated using TRIzol reagent (Life 308 

technologies, Carlsbad, CA, US), according to the manufacturer’s instructions, and 309 

purified by using the RNeasy Mini Kit (Qiagen, GmBH, Germany). RNA samples of 310 

each group were subsequently used to generate fluorescence-labeled cRNA targets 311 

for the Affymetrix Human U133 Plus 2 arrays (Affymetrix, Santa Clara, CA, US;312 

Ca#900467). The labeled cRNA targets were then hybridized on slides. After 313 

hybridization, slides were scanned by GeneChip® Scanner 3000 (Affymetrix, Santa 314 

Clara, CA, US). Data were extracted with Command Console Software 4.0 315 

(Affymetrix, Santa Clara, CA, US). Raw data were normalized by the MAS 5.0 316 

on April 7, 2020. © 2018 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on October 3, 2018; DOI: 10.1158/2326-6066.CIR-17-0630

15  

algorithm and Gene Spring Software v12.6.1 (Agilent technologies, Santa Clara, CA, 317 

US). The microarray experiments were performed following the protocol of 318 

Affymetrix Inc. (Shanghai Biotechnology Corporation, China). 319 

320 

Cell lines 321 

The MC38 cell line was purchased from National Infrastructure of Cell Line Resource 322 

(Beijing, China) and was maintained in RPMI 1640 with 10% fetal bovine serum 323 

(Gibco, Carlsbad, CA), penicillin sodium (100 U/mL), and streptomycin sulfate (100 324 

mg/mL) in humidified 5% CO2 at 37°C. The cell line was tested and authenticated by 325 

(STR) profiling. Cells were routinely tested for mycoplasma infection and used only 326 

when negative. Cells were passaged a maximum of 3-4 times in vitro before they were 327 

used in in vivo experiments. 328 

329 

In vivo mouse studies 330 

Four- to six-week old male C57BL/6 mice were purchased from Beijing 331 

Vitalriver Experimental Animal Technology Co. Ltd. (Beijing, China). Mice were 332 

maintained in a pathogen-free facility and used in accordance with the institutional 333 

guidelines for animal care. All animal experiments were performed following 334 

protocols approved by the Institutional Animal Care and Use Committee (IACUC) at 335 

the Peking University Cancer Hospital. MC38 (1×104) cells were mixed with an equal 336 

volume of Matrigel (BD Biosciences) and subcutaneously injected on the right leg of 337 

the mice on day 0. The tumors were irradiated with 8 Gy on day 15. Blocking 338 

on April 7, 2020. © 2018 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on October 3, 2018; DOI: 10.1158/2326-6066.CIR-17-0630

16  

antibodies (PD-1, clone RMP1-14; PD-L1, clone 10F.9G2 and rat IgG2B isotype, 339 

clone LTF-2, all from BioXCell) were given intraperitoneally on days 16, 19, and 22. 340 

All irradiation was performed using the Edge linear accelerator (Varian Medical 341 

Systems, Inc., Palo Alto, CA). Antibodies used for in vivo immune checkpoint 342 

blockade experiments were given intraperitoneally at a dose of 200 μg/mouse and 343 

include: PD-1 (clone RMP1-14), PD-L1 (clone 10F.9G2), and rat IgG2B isotype 344 

(clone LTF-2) (all from BioXCell). The tumor volumes were measured using CT 345 

scans and MRI once a week. Volume was calculated using the formula L × A × B × 346 

0.52, where L is the longest dimension and A and B are long and short diameters of 347 

the largest coronal section, respectively. The untreated tumor volumes were 348 

determined at day 14 using CT scans and were considered as a baseline tumor volume 349 

(Vcont). Normalized tumor response to treatment was the measured volume (V) 350 

relative to Vcont. The tumor volumes were also measured using calipers every three 351 

days. Differences in survival were determined for each group by the Kaplan-Meier 352 

method. The overall p-value was calculated by the log-rank test. For mouse studies, 353 

an event was defined as death or when tumor burden reached a protocol-specified size 354 

of 1.5 cm in maximum dimension to minimize morbidity (26). 355 

356 

Immunohistochemical analysis on tissue microarrays (TMAs) 357 

Immunohistochemistry was performed on TMAs using rabbit polyclonal anti-human 358 

PD-1 (LS-B540, LifeSpan BioSciences, USA), anti-human PD-L1 (ab58810, Abcam, 359 

USA), anti-human PD-L2 (HPA013411, Sigma, USA), mouse monoclonal 360 

on April 7, 2020. © 2018 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on October 3, 2018; DOI: 10.1158/2326-6066.CIR-17-0630

17  

anti-human CD8 (clone 4B11, Novus Biologicals, USA). All images were examined 361 

by two experienced pathologists independently. For PD-L1 and PD-L2, the 362 

immunoreactivity of the proteins detected was recorded through the intensity of 363 

staining, and the percentage of immunoreactive cells scored as: tissues with no 364 

staining were rated as 0, with a faint or moderate staining to strong staining in <25% 365 

of cells rated as 1, strong staining in 25–50% of cells rated as 2, and strong staining 366 

in >50% of cells rated as 3. The slides were further analyzed to identify PD-1 and 367 

CD8 using an image analysis workstation (Spot Browser, Alphelys). The total 368 

number of PD-1+ or CD8+ cells in each tissue spot was counted, and the density of 369 

PD-1+ or CD8+ TILs was defined as the cell number per square millimeter. 370 

371 

TCGA and GEO datasets 372 

Level 3 TCGA RNA-seq and DNA exome-seq data for rectal cancer patients with 373 

clinical information was downloaded from the TCGA data portal, including 341 rectal 374 

cancer samples on 18 July, 2015. The cases that had clear information about nCRT 375 

were analyzed, including 7 rectal cancers with nCRT and 41 without nCRT. 376 

EdgeR-normalized data of these 48 cases were used for correlative analysis. 377 

Two patient datasets of rectal cancer were downloaded from the Gene Expression 378 

Omnibus (GEO) database. GSE 15781 consisted of 22 patients with resectable 379 

adenocarcinoma of the rectum, in which thirteen patients had surgery only, and nine 380 

patients received nCRT. GSE 45404 included pre-treatment biopsies of 42 rectal 381 

cancer patients who received nCRT with conventional fractionation (≥50 Gy in 28 382 

on April 7, 2020. © 2018 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on October 3, 2018; DOI: 10.1158/2326-6066.CIR-17-0630

18  

fractions, 1.8 Gy/day, 5 sessions per week) and 5-fluorouracil (5-FU)-based 383 

chemotherapy. 384 

385 

Mutant peptide MHC binding prediction 386 

All non-synonymous point mutations identified were translated into strings of 17 387 

amino acids, with the mutant amino acid situated centrally according to previous 388 

research (23,24). Our initial analysis was focused on HLA-A and HLA-B (reference 389 

set of 27 alleles was assembled and covered >97% of population (35), the accession 390 

numbers for the reference alleles were shown in Supplementary Table S6). The 17 391 

mutant amino acid fragments were analyzed by the epitope prediction program 392 

NetMHC v4.0 (http://www.cbs.dtu.dk/services/NetMHC/). Epitopes with a predicted 393 

affinity of <50 nm were considered to be strong potential binders, and epitopes with a 394 

predicted affinity of <500 nm were considered weak potential binders, as suggested 395 

by the NetMHC group. To further refine the total neoantigen burden, we repeated the 396 

same process for the complementary wild-type peptide for each mutant peptide. The 397 

mutant peptides that held strong potential binders were used to compare with the 398 

complementary wild-type peptide, which was predicted to have a weak potential 399 

binder. These mutant peptides were referred to as mutation-associated neoantigens. 400 

These mutant peptides were further evaluated for putative binding to the T-cell 401 

receptor (TCR) using the IEDB immunogenicity predictor with patient-specific HLA 402 

types (http://tools.immuneepitope.org/immunogenicity/) and CTLPred 403 

(http://www.imtech.res.in/raghava/ctlpred/). 404 

on April 7, 2020. © 2018 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on October 3, 2018; DOI: 10.1158/2326-6066.CIR-17-0630

19  

405 

Statistics 406 

The Mann-Whitney test and Student t test were used to compare mutation burdens and 407 

differences in the frequency of nucleotide changes. The log-rank test was used to 408 

compare Kaplan-Meier survival curves. Correlations between the non-synonymous 409 

mutation burden and the neoantigen burden, frequency of the neoantigen 410 

burden/non-synonymous mutation, and immune response were calculated using the 411 

Pearson correlation formula. A mixed effect linear model was used to determine 412 

significance of differences in tumor growth. Statistical analyses were performed using 413 

GraphPad Prism v.6. 414 

Availability of data and material 415 

Whole-exome sequencing data are available under the NCBI Sequence Read 416 

Archive (SRA) study accession no. SRP159539. Microarray data are prepared 417 

according to minimum information about a microarray gene experiment (MIAME) 418 

guidelines and deposited in the GENE Expression Omnibus (GEO) database. The 419 

GEO accession number is GSE119409. 420 

421 

Results 422 

nCRT induces immune activation in LARC 423 

To assess whether nCRT could affect the immune response, we compared rectal 424 

cancer samples with and without treatment from GEO data (GSE 15781) using GSEA. 425 

This data consisted of 22 patients with resectable adenocarcinoma of the rectum. 426 

on April 7, 2020. © 2018 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on October 3, 2018; DOI: 10.1158/2326-6066.CIR-17-0630

20  

Thirteen patients had surgery only, and nine patients received nCRT. The irradiated 427 

and non-irradiated tumor tissues and normal tissues were used to perform 428 

genome-wide gene expression analysis. A supervised method (Significance Analysis 429 

of Microarrays -SAM-) was initially used to identify a statistical significance 430 

(adjusted p<0.05) in differentially expressed genes between samples with and without 431 

nCRT treatment. 3359 differentially expressed genes were identified between these 432 

two subgroups. Of which, 1840 genes revealed significantly higher expression in 433 

post-treatment samples, whereas 1519 genes showed lower expression. A hierarchical 434 

clustering analysis was subsequently performed based on the expression values of the 435 

3359 genes in the 22 samples (Supplementary Fig. S1) to categorize the samples into 436 

2 main subgroups (branches), in which the gene expressions showed the opposite 437 

trend. 438 

To further investigate the mechanism of the immune response alternation after the 439 

chemoradiotherapy, the differentially expressed genes induced by chemoradiotherapy 440 

with the immune-related genes defined by Gene Ontology (1776 genes) were 441 

compared. A total of 342 immune-related genes were altered, with most being 442 

upregulated. Unsupervised hierarchical clustering of this subset of overlapping genes 443 

resulted in a clear separation of the samples by nCRT (Fig. 1A and B). 444 

Reactome pathway analysis showed that immune activation-related pathways were 445 

induced, including interferon signaling, antigen presentation, class I MHC-mediated 446 

antigen processing and presentation, PD-1 signaling, peptide-ligand binding receptors, 447 

and co-stimulation from the CD28 family (Table 1). Amongst the altered 342 448 

on April 7, 2020. © 2018 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on October 3, 2018; DOI: 10.1158/2326-6066.CIR-17-0630

21  

immune-related genes, 17 genes fell into the CD28 costimulatory signal pathway 449 

(defined by Reactome pathway database) (Fig. 1B). CD28, CD86, HLA class II 450 

molecules, PIK3CA, PTPN11, MAPKAP1, GRB2, VAV1, FYN, and MAP3K8 were 451 

upregulated (Fig 1C). In contrast, PAK1, PPP2R1A, and MLST8 were downregulated. 452 

The expression of T-cell differentiation and activation markers were analyzed, and the 453 

results revealed that differentiation markers (KLRG1 and PTPRC) and activation 454 

markers (CD69, IL2RA, CD38) (36,37) were upregulated in LARC samples following 455 

nCRT (Fig. 1D). 456 

We then focused on the differentially expressed genes and performed gene set 457 

enrichment analysis using a prior defined immunological signature set (signature of 458 

gene expression upon perturbation of certain immune-related genes). Post-treatment 459 

LARC was characterized by increased expression of most immune-related genes and 460 

associated with a diffuse immune infiltrate mainly composed of TH1 cells and 461 

cytotoxic T cells expressing checkpoint molecules (Fig. 1E). 462 

To further assess the effect of nCRT on the infiltration of specific immune cell 463 

subsets, we used CIBERSORT (38) as an approach to dissect infiltration of specific 464 

immune cell subsets in the tumors. The higher fractions of monocytes, activated 465 

dendritic cells, and neutrophils cells were found in rectal cancers with nCRT. The 466 

faction of gamma delta T cells slightly increased from 0 to 0.78% following nCRT 467 

(Supplementary Fig. S2). The list of differentially expressed genes, immune genes, 468 

and CD28 co-stimulatory signals are presented in Supplementary Tables S7-9. 469 

on April 7, 2020. © 2018 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on October 3, 2018; DOI: 10.1158/2326-6066.CIR-17-0630

22  

nCRT influences the mutational landscape of LARC 470 

Pre- and post-nCRT tumor samples from 9 of the 14 patients of cohort 1 were 471 

collected and sequenced, and the other 5 cases only had biopsy samples prior to 472 

therapy because of pathologically invisible tumor masses after achieving pCR. The 473 

tumor DNA sequencing generated a mean target coverage of 239X, and a mean of 474 

99.8% of the target sequence was identified as a depth of minimum 10X. A median of 475 

80 non-synonymous mutations per pre-nCRT sample (range 3 to 313) was detected. 476 

The median number of exonic mutations per pre-nCRT sample was 122 (range 3 to 477 

455). The microsatellite instability status of the nine cases was tested, and all cases 478 

were microsatellite stable (MSS). The quantity and range of mutations were similar to 479 

published series of CRCs (22). The nCRT-induced mutations appeared in 480 

post-treatment samples. The median number of exonic mutations induced by nCRT 481 

was 23 compared to pre-treatment samples (range 8 to 89). The median 482 

non-synonymous mutation induced by nCRT was 15 per post-nCRT sample (range 6 483 

to 57). 484 

nCRT influences the neoantigens landscape of LARC 485 

The landscape of neoantigens according to previously described methods (23,24) were 486 

examined. This approach identified mutant nonamers with ≤500 nM binding affinity 487 

for patient-specific class I human lymphocyte antigen (HLA) alleles, which were 488 

considered candidate neoantigens. A median of 58 candidate neoantigens from each 489 

pre-treatment tumor (range 0 to 256) and the quantity of neoantigens per tumor 490 

correlated with mutation burden was identified (Pearson r 0.9491, p<0.0001; Fig. 2A). 491 

on April 7, 2020. © 2018 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on October 3, 2018; DOI: 10.1158/2326-6066.CIR-17-0630

23  

New mutations induced by nCRT from post-treatment tumors also resulted in a 492 

median of 11 candidate neoantigens with a range 3 to 50. These candidate neoantigens 493 

significantly correlated with new mutation numbers (Pearson r=0.9640, p<0.0001; Fig. 494 

2B). 495 

The mutational landscape from TCGA data analysis of rectal cancer with and without 496 

nCRT showed that the median non-synonymous mutation burden was 163 in tumors 497 

from patients with nCRT compared to 120 in patients without nCRT (Student t test, 498 

p=0.0387; Fig. 2C). The median candidate neoantigens per tumor was 22 (range 0 to 499 

55), and the quantity of neoantigens per tumor correlated with mutation burden 500 

(Pearson r=0.9988, p<0.0001; Fig. 2D). The median putative neoantigens binding to 501 

the TCR was 18 in with nCRT patients compared to 14 in without nCRT. 502 

(Mann-Whitney test, p=0.01, Fig. 2E) 503 

Immune activation is related to neoantigens arising from mutation burden 504 

To further confirm that immune activation following nCRT was related to 505 

neoantigens arising from mutation burden, the correlation between the number of 506 

mutations and the expression of all genes in rectal cancer from the TCGA DNA 507 

exome-seq and RNA-seq data was studied. An ordered list of 354 significant related 508 

genes were generated (Pearson r>0.8 or r<-0.8, p<0.01) for the Reactome pathway 509 

database analysis. The enriched pathways focused on the immune system, signal 510 

transduction, and developmental biology. An association was identified between 511 

mutation burden and enrichment of immune activation-related pathways, including 512 

on April 7, 2020. © 2018 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on October 3, 2018; DOI: 10.1158/2326-6066.CIR-17-0630

24  

alpha-defensins, IL2 family signaling, and other several interleukin-related pathways 513 

(interleukin receptor SHC signaling, interleukin-19, 20, 22, 24, 26, 28, and 29 514 

signaling, interleukin-3, 5 and GM-CSF signaling, interleukin-17 signaling) (Table 515 

1). 516 

RT combined with anti–PD-1 enhances control of tumor growth 517 

To understand the contribution of RT to immune checkpoint blockade, we utilized 518 

the MC38 CRC mouse model. Mice with right leg tumors received irradiation (RT), 519 

anti–PD-1/PD-L1 (PD1/PDL1, anti-PD-1/PD-L1 are abbreviated as PD-1/PDL1), or 520 

both treatments delivered sequentially (RT+PD1 or RT+PDL1) (Fig. 3A). Tumor 521 

growth was controlled by RT, PD1, RT+PD1, and RT+PDL1 (p=0.0163, p=0.02, 522 

p<0.0001, p=0.0035, respectively), and tumor growth was well-controlled when 523 

RT+PD1 was administered compared to either treatment alone (p=0.0037, p=0.0021). 524 

No significant differences among PDL1, RT, and RT+PDL1 treatments were seen 525 

(Fig. 3B-D). 526 

Kaplan-Meier analysis showed that PD1, RT, RT+PD1, PDL1, and RT+PDL1 527 

improved the survival of mice (p=0.001, p=0.018 for comparisons of PD1 and PDL1 528 

treatment groups, respectively). The largest improvement in survival was observed in 529 

the RT+PD1 group. When compared with RT or PD1 alone, RT combined with PD1 530 

still improved the survival significantly (p=0.046, p=0.042, respectively). No 531 

significant difference among PDL1, RT, and RT+PDL1 groups was found (Fig. 3E). 532 

Mutation burden and neoantigens correlated with the response to nCRT 533 

on April 7, 2020. © 2018 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on October 3, 2018; DOI: 10.1158/2326-6066.CIR-17-0630

25  

The analysis of whole-exome sequencing indicated that rectal cancers with higher 534 

mutation burden were more sensitive to nCRT (Fig.4 and 5A). The results showed 535 

high numbers of somatic mutations in pre-nCRT biopsies, whcih were associated with 536 

tumor regression grade (TRG; Pearson r=-0.5418, p=0.0454; Fig.5A). The potential 537 

mutation-associated neoantigen numbers were negatively correlated with TRG but 538 

was not statistically significant (Pearson r=-0.3684, p=0.1949; Fig. 5B). 539 

Target-capture sequencing of plasma ctDNA and matched normal tissue DNA (cohort 540 

2) was performed to detect somatic mutations in each sample, achieving a mean 541 

sequencing coverage of 2313×(914~3792×) for plasma ctDNA and 1675×(765~2649×) 542 

for normal tissue DNA. The correlation between the tumor mutation burden (TMB) of 543 

ctDNA and the tumor regression grade varied among the patients (mean 9.76 544 

mutation/Mb, range 4–16 mutations/Mb; Fig. 5C). TMB was associated with the 545 

tumor regression grade (Pearson r=-0.3636, p=0.0179; Fig. 5D). The mean TMB was 546 

higher in pCR patients than in non-pCR patients (13 vs 9.22, respectively; p=0.0014; 547 

Fig. 5E). The patients were divided into two groups by assessing the percentage of 548 

viable residual tumor cells less or more than 30% in the resected specimens. The 549 

mean TMB was higher in the <30% group than in the >30% group (10.5 vs 8.78, 550 

respectively; p=0.0472; Fig. 5F). 551 

Immune activity correlated with the response to nCRT 552 

To further investigate the correlation between gene expression and response to nRT, 553 

mRNA expression in pre-therapy biopsies of cohort 3 was profiled into responding 554 

(n=19) and non-responding (n=47) groups. A supervised method (Significance 555 

on April 7, 2020. © 2018 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on October 3, 2018; DOI: 10.1158/2326-6066.CIR-17-0630

26  

Analysis of Microarrays -SAM-) was used to find statistical significance (adjusted 556 

p<0.05) in differentially expressed genes between responding and non-responding 557 

groups. A total of 59 genes were differentially expressed between these two 558 

subgroups (p<0.05, fold change >1.5), with most genes presenting significantly 559 

higher expression in the responding group and 4 genes with lower expression 560 

(Supplementary Fig. S3). Reactome pathway analysis showed that the immune 561 

activation-related pathways were enriched, including interleukin-10 signaling, 562 

chemokine receptors bind chemokines, cytokine signaling in immune system, 563 

peptide ligand-binding receptors, immune system, PD-1 signaling, interferon 564 

signaling, and co-stimulation factors within the CD28 family (Supplementary Table 565 

S10). 566 

We also compared responding and non-responding rectal cancer pre-treatment 567 

biopsies from GEO data (GSE 45404) consisting of 42 patients who received nCRT 568 

with conventional fractionation (≥50 Gy in 28 fractions, 1.8 Gy/day, 5 sessions per 569 

week) and 5-fluorouracil (5-FU)-based chemotherapy. The readings from the 570 

pre-treatment patients responding (n=19) and non-responding (n=23) to 571 

chemoradiotherapy were compared. Reactome pathway analysis showed similar 572 

results to cohort 3(Supplementary Table S10). 573 

The correlation between infiltrating immune cells and response to nCRT was further 574 

assessed using CIBERSORT. The results of cohort 3 showed that the fractions of 575 

CD8+ T cells (19% vs. 12%) and activated CD4+ memory T cells (0.47% vs. 0) in the 576 

responding group were higher than the non-responding group. In the non-responding 577 

on April 7, 2020. © 2018 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on October 3, 2018; DOI: 10.1158/2326-6066.CIR-17-0630

27  

group, the fraction of regulatory T cells (Tregs) was higher than the responding 578 

group (2.29% vs. 0.59%). GSE 45404 data also showed a significant increase in the 579 

fractions of CD8+ T cells (1.99% vs. 0%) and activated CD4+ memory T cells 580 

(7.67% vs. 2.97%) in the responding group compared to the non-responding group 581 

(Supplementary Fig. S4). 582 

PD-L1 expression negatively correlates with prognosis 583 

The expression pattern of PD-L1, PD-1, and PDL2 in tumors and corresponding 584 

non-tumor tissue of cohort 4 was studied using IHC. PD-L1 was expressed not only 585 

on tumor cells, but also on TILs. The majority of PD-L1 expression was in the tumor 586 

cells, whereas weak expression was observed on TILs (Fig. 6A). A high expression 587 

of PD-L1 was detected in epithelial cells with elevated expression in tumor tissue 588 

compared with normal tissue (Z=-5.538, p<0.0001). 589 

Expression of PD-L1 was correlated with M stage (Spearman r=0.13, p=0.03), 590 

differentiation (Spearman r=0.131, p=0.029), and histological type (U=3.8158, 591 

p=0.000136). PD-L1 expression in liver metastasis tissues was lower than expression 592 

in paired primary tumor tissues (Z=-2.346, p=0.019, Supplementary Table S11). 593 

Positive PD-L1 expression in CRC tissues was associated with a shorter overall 594 

survival (OS, p=0.065) and disease-free survival (DFS, p=0.06) (Supplementary Fig. 595 

S5A and B), although the tendency was not statistically significant. For stage I–III 596 

CRC patients, positive PD-L1 expression was significantly associated with poor OS 597 

(p=0.019) (Fig. 6B), whereas for stage IV CRC patients, positive PD-L1 expression 598 

on April 7, 2020. © 2018 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on October 3, 2018; DOI: 10.1158/2326-6066.CIR-17-0630

28  

was significantly associated with favorable prognosis of CRC (p=0.001) 599 

(Supplementary Fig. S5C). 600 

The association between PD-L1 expression and prognosis of rectal cancer indicated 601 

that positive PD-L1 expression was significantly associated with poor prognosis of 602 

rectal cancer (for DFS, p=0.031; for OS, p=0.043) (Fig. 6C and D). No significant 603 

correlation between PD-L1 and prognosis of colon cancer was found 604 

(Supplementary Fig. S5D and E). We further performed subgroup analysis in 605 

different stages of colon and rectal cancers to ascertain the effect of primary tumor 606 

location on the correlation. For stage I–III rectal cancer patients, positive P-DL1 607 

expression was significantly associated with poor OS (p=0.023; Supplementary Fig. 608 

S6A). In stage IV rectal cancer, PD-L1 expression was associated with better OS 609 

(p=0.011; Supplementary Fig. S6B). However, no significant correlation between 610 

PD-L1 and prognosis of colon cancer, whether in stage I-III (p=0.069, 611 

Supplementary Fig.6C) or stage IV (p=0.169; Supplementary Fig. S6D), was found. 612 

PD-1 was also expressed in tumor cells and TILs, with TILs having the highest 613 

expression. PD-1 expression was negatively correlated with tumor cell 614 

differentiation (Spearman r=-0.135, p=0.022), but no significant correlation between 615 

PD-1 and other clinical-pathological findings was found (Supplementary Table S11, 616 

Supplementary Fig. S7A-D). 617 

PD-L2 was also expressed not only in tumor cells, but also in TILs. However, the 618 

majority of PD-L2 expression was in tumor cells (Supplementary Fig. S8A). High 619 

on April 7, 2020. © 2018 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on October 3, 2018; DOI: 10.1158/2326-6066.CIR-17-0630

29  

PD-L2 expression was significantly associated with better OS (p=0.001; 620 

Supplementary Fig. S8B) in all stage CRC patients. Specifically, for stage I–III CRC 621 

patients, high PD-L2 expression was significantly associated with better OS (p=0.042; 622 

Supplementary Fig. S8C), whereas for stage IV CRC patients, high PD-L2 expression 623 

was associated with poor prognosis (p=0.067; Supplementary Fig. S8D). We also 624 

performed subgroup analysis in different stages of colon and rectal cancers. For colon 625 

cancer patients, a significant positive correlation between PD-L2 and prognosis in all 626 

stages was seen (p<0.0001; Supplementary Fig. S8E). However, no significant 627 

correlation between PD-L2 and prognosis was found in stage I-III or stage IV alone. 628 

For all stage rectal cancer patients, no significant correlation between PD-L2 and 629 

prognosis was seen. For stage I–III rectal cancer patients, there was no significant 630 

correlation between PD-L2 and prognosis, but in stage IV rectal cancer, PD-L2 631 

expression was associated with poor OS (p<0.0001, Supplementary Fig. S8F). 632 

PD-L1 and PD-1 after nCRT correlated with CD45RO, CD8 and outcomes 633 

To determine whether PD-L1 and PD-1 were associated with clinical and pathologic 634 

features in LARC patients, tissue microarray analysis of 296 LARC patients from 635 

cohort 5 was performed. The patients’ profile is listed in Supplementary Table S5 and 636 

the tumor-infiltrating cytotoxic lymphocytes are shown in Fig. 7A and B. No 637 

significant correlation was found between PD-L1, PD-1, and clinical-pathological 638 

parameters observed (Supplementary Table S12). 639 

To understand the relationship between PD-1, PDL-1, CD45RO, and CD8, the 640 

on April 7, 2020. © 2018 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on October 3, 2018; DOI: 10.1158/2326-6066.CIR-17-0630

30  

correlation coefficients of PD-1, PD-L1, CD45RO, and CD8 expression among all 641 

patient samples were calculated. PD-L1 was significantly correlated with CD45RO 642 

(r=0.157, p=0.009, Spearman’s test) and CD8 (r=0.278, p=0.000, Spearman’s test). 643 

No correlation was found between PD-1 and CD8 or CD45RO (Supplementary Table 644 

S12). 645 

Our previous work demonstrated that CD45RO expression significantly correlates 646 

with prognosis of LARC patients with neoadjuvant radiotherapy (39). To determine if 647 

prognostic associations existed between the expression of PD-L1, PD-1, CD8, and 648 

patient survival, Kaplan–Meier survival curves were plotted. Again, no significant 649 

correlation between DFS or OS and the expression of the proteins was found 650 

(Supplementary Fig. S9A-F). 651 

These results suggested that PD-1, PD-L1, and CD8 were not independent prognostic 652 

factors for patient survival. However, if the patient survival was plotted against a 653 

combination of PD-L1 with CD8, in the CD8-low expression group, a significant 654 

negative correlation was observed in DFS (p=0.042; Fig. 7C). It seemed that 655 

CD8-high expression was not associated with DFS and OS (Supplementary Fig. S10A 656 

and B). The association of patient survival with the combination of PDL1 with 657 

CD45RO, in the CD45RO-low expression group, a negative correlation was observed 658 

in overall patient survival (p=0.081) or DFS (p=0.099), although it was not 659 

statistically significant (Supplementary Fig. S10C and D). 660 

661 

Discussion 662 

on April 7, 2020. © 2018 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on October 3, 2018; DOI: 10.1158/2326-6066.CIR-17-0630

31  

We analyzed somatic gene expression in irradiated and non-irradiated rectal cancer 663 

samples from GEO data to assess the influence of nCRT on the immune condition. 664 

Our study indicated that chemoradiotherapy could be able to induce an immune 665 

response in rectal cancer patients. Post-treatment LARC is characterized by increased 666 

expression of genes involved in immune response pathways, mainly composed of 667 

cytotoxic T and TH cells, along with strong activation of antigen presentation, peptide 668 

ligand-binding receptors, CD28 co-stimulatory signals, interferon signaling, and other 669 

cytokine signaling in the immune system. 670 

T-cell activation requires a two-step process that includes engagement of the TCR to 671 

an antigen presented by an antigen-presenting cell (APC), and a second costimulatory 672 

signal delivered by the engagement of CD28 to its ligands CD80 and CD86 (40). 673 

Tumor cells expressing HLA class I present tumor-associated antigens on their cell 674 

surface and are recognized by CD8+ cytotoxic T cells. Our analysis showed that nCRT 675 

led to a significant increase in CD28, CD86, and HLA class I and II molecule 676 

expression in rectal cancer cells. The class II molecule is expressed on the surface of 677 

professional APCs and to some degree on cancer cells (41) and plays a central role in 678 

the immune system by presenting peptides derived from extracellular proteins. Studies 679 

have shown the direct requirement for competent HLA class II pathway stimulation in 680 

the reduction of HLA class I-mediated response for an effective immunotherapy 681 

approach (42). HLA class II antigen expression in CRC tumors is a favorable 682 

prognostic marker (43). We noticed that T cell activation in post-treatment LARC was 683 

mainly through IFNγ signaling activation and increased PI3K signaling. We further 684 

on April 7, 2020. © 2018 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on October 3, 2018; DOI: 10.1158/2326-6066.CIR-17-0630

32  

analyzed the expression of T-cell differentiation and activation markers. 685 

Differentiation markers (KLRG-1 and CD45 isotypes) and activation markers (CD69, 686 

CD25 and CD38) were increased in LARC following nCRT compared to pre-nCRT 687 

conditions. It was clear that nCRT could induce immune activation in LARC patients. 688 

The mechanism by which radiation induces adaptive immunity remains unclear. 689 

Twyman-Saint et al. report major tumor regressions in a subset of patients with 690 

metastatic melanoma treated with an anti-CTLA4 and radiation, which was 691 

reproduced in mouse models (26). Their study demonstrates that radiation can 692 

diversify the TCR repertoire of TILs and shape the repertoire of expanded clones. 693 

Reits et al. demonstrates that radiation can enhance MHC class I expression by 694 

modulating the peptide repertoire (44). 695 

Interest in the relationship between somatic mutational burden and antitumor immune 696 

response motivated us to examine the differences in the mutational landscape between 697 

the pre- and post-treatment rectal cancers by sequencing the exomes of LARCs. Our 698 

analysis revealed that nCRT influenced the mutational landscape of LARC and 699 

induced novel somatic mutations, and we further validated the results in TCGA 700 

datasets. The most impactful finding from this study was the correlation between 701 

immune activation and mutation burden in post-nCRT treated rectal cancers. 702 

Although the exact mechanism of the enhanced immune response needs to be further 703 

clarified, our observations clearly showed that neoantigens are associated with 704 

non-synonymous mutation burden, which is consistent with the hypothesis that 705 

on April 7, 2020. © 2018 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on October 3, 2018; DOI: 10.1158/2326-6066.CIR-17-0630

33  

recognition of neoantigens derived from somatic mutations is important for the 706 

activity of the immune response, regardless of nCRT treatment. The tumors with 707 

higher mutational load in melanoma, lung cancer, and mismatch repair–deficient 708 

CRCs have a higher rate of response to checkpoint blockade therapy (22,23,45). 709 

These data provide further insight for the idea that mutation-associated neoantigen 710 

recognition is an important component of the endogenous antitumor immune 711 

response. 712 

Numerous studies have shown that chemoradiotherapy induces local immune 713 

reactions that contribute to tumor regression through inflammatory infiltration. Our 714 

previous work also demonstrates that the density of CD45RO+ TILs can predict 715 

tumor downstaging and long-term outcomes for rectal cancer following neoadjuvant 716 

radiotherapy (39). Evidence shows that a high rate of response to checkpoint therapy 717 

is based on boosting tumor-specific immune activity. Therefore, we hypothesized 718 

that rectal cancer patients who responded to nCRT could be good candidates for 719 

checkpoint blockade immunotherapy, especially for the patients without pCR. Our 720 

preclinical mouse experiments demonstrated that radiotherapy could enhance the 721 

efficiency of anti–PD-1, which further supported our hypothesis. 722 

In this work, we demonstrated that a higher tumor mutation burden in pre-treatment 723 

tumors is correlated with a lower tumor regression grade. We also found that the 724 

immune activation-related genes and pathways were enriched in the patients who 725 

were responding to the nCRT. Heterogeneous immune cell infiltration was present in 726 

responding and non-responding patients, and the responding patients displayed a 727 

on April 7, 2020. © 2018 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on October 3, 2018; DOI: 10.1158/2326-6066.CIR-17-0630

34  

higher infiltration of CD8+ and activated CD4+ memory T cells, whereas the 728 

non-responding patients displayed a significantly higher Treg infiltration. Our results 729 

indicated that patients’ immune activity is related to the somatic mutation burden and 730 

associated with nCRT. Mutation burden could be a useful biomarker to stratify 731 

patients into sensitive and resistant categories to nCRT. 732 

Several studies have addressed that tumor shrinkage induced by chemoradiotherapy 733 

is not simply dependent on direct damage to tumor cells but is also affected by the 734 

host immune activity. Immune infiltration could be a biomarker to predict the 735 

response to nCRT in rectal cancer. Yasuda et al. previously analyzed the density of 736 

CD4+ and CD8+ T lymphocytes in rectal cancer patients before nCRT and 737 

demonstrated that a higher density of lymphocytes was correlated with a better 738 

response to nCRT (46). Anitei et al. analyzed immunoscores, which score the 739 

presence of T lymphocytes, in rectal cancer patients and show low immunoscores for 740 

patients who did not respond to nCRT (47). Tumors with higher immune activity are 741 

considered to be immunogenic and tend to further evoke antitumor immune 742 

responses by neoantigens due to chemoradiotherapy, resulting in a better response to 743 

CRT. Studies demonstrate that irradiation can promote remodeling of the 744 

extracellular matrix (ECM) and tumor vasculature by increasing intratumoral 745 

oxygenation and pH and upregulating the expression of cell adhesion molecules. 746 

This leads to increased recruitment of immune effector cells into the tumor (48). 747 

We further investigated the prognostic value of the checkpoint molecules PD-1, 748 

PD-L1, and PD-L2 in CRC. PD-L1 expression was correlated with poor prognosis of 749 

on April 7, 2020. © 2018 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on October 3, 2018; DOI: 10.1158/2326-6066.CIR-17-0630

35  

rectal cancer but not colon cancer. Increasing evidence suggests that CRC should be 750 

considered as a heterogeneous disease, with colon and rectal cancers showing 751 

multiple clinicopathological and molecular distinctions, including the immune 752 

microenvironment (49,50). The TILs in CRC and its microenvironment are 753 

associated with survival, and this prognostic correlation differs according to tumor 754 

location (51). 755 

In rectal patients without nCRT, PD-L1 expression negatively correlated with 756 

prognosis in stage I-III rectal cancer patients and positively correlated in stage IV 757 

rectal cancers. The expression pattern and correlation with prognosis of PD-L2, 758 

another PD-1 ligand, was different from that of PD-L1. PD-L2 expression was 759 

correlated with better prognosis of colon cancer but poor prognosis of stage IV rectal 760 

cancer, indicating that the biological roles of PD-L1 and PD-L2 were different 761 

between colon and rectal cancer. Our results are consistent with the survival analysis 762 

of CRC RNA-seq data from TCGA presented in the Human Protein Atlas 763 

(www.proteinatlas.org). However, our results are inconsistent with the study by 764 

Wang et al (52), which shows that PD-L2 overexpression in CRC tumor cells 765 

associates with poor OS of patients. It was shown that in both early CRC (AJCC 766 

stage I–II) and advanced CRC (stage III–IV), higher PD-L2 expression associated 767 

with worse OS. However, it should be noted that their study cohort was mostly 768 

composed of colon cancer and only had two rectal cancer samples. For stages of 769 

samples, the cohort only had four stage IV samples, and therefore, their results 770 

mainly reflect the expression of PD-L2 in stage I-III colon cancer and its prognostic 771 

on April 7, 2020. © 2018 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on October 3, 2018; DOI: 10.1158/2326-6066.CIR-17-0630

36  

significance. We think that the difference between their results and ours might be 772 

due to the different composition of the study cohort and a different antibody used in 773 

the study. 774 

In patients with neoadjuvant radiotherapy, no significant correlation between PD-L1 775 

expression and prognosis was found. CRC is considered as an immune cold tumor, 776 

except in high mutation CRC with MSI-H or POLE mutation (53). Multiple factors 777 

produced by tumor and stromal cells contribute to the inhibition of antitumor immune 778 

response. Interaction of PD-L1 with PD-1 inhibits T-cell activation and cytokine 779 

production (54,55). In tumor microenvironments, elevated PD-L1 can result in T-cell 780 

exhaustion (56). T cells, thus, fail to maintain an energetic status to fight tumor cells 781 

and are rendered tolerant to tumor antigens or exhausted. Chemoradiotherapy could 782 

reprogram the immune-suppressive TME towards an immune-stimulating one. 783 

Following neoadjuvant radiotherapy, T-cell activation was initiated by the 784 

tumor-specific neoantigens resulting from nCRT, which could partially counteract the 785 

effect of PD-L1. Following neoadjuvant radiotherapy, PD-L1 was significantly 786 

correlated with CD45RO and CD8. In the CD8-low expression group, in which 787 

antitumor immune was poorly activated, PD-L1 expression was negatively correlated 788 

with DFS. Therefore, we propose that the addition of checkpoint blockade to nCRT 789 

may show significant efficacy in improving prognosis of rectal cancer. T-cell 790 

infiltration, such as CD8+ T cells, could be a potential biomarker to further stratify 791 

post-nCRT patients into future immunotherapy groups (CD8-high expression) or 792 

surgery alone group (CD8 low expression). 793 

on April 7, 2020. © 2018 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on October 3, 2018; DOI: 10.1158/2326-6066.CIR-17-0630

37  

It is plausible that the immune features associated with nCRT could influence the 794 

response to immunotherapeutic strategies. The mutational landscape could provide a 795 

simple selection or stratification factor to identify populations of interest for such 796 

treatments, and its exploration as a predictive biomarker is warranted. 797 

Altogether, this work establishes the link between somatic mutations and immune 798 

activity in rectal cancer patients following nCRT and supports the hypothesis that 799 

rectal cancer patients with nCRT could become potential candidates for checkpoint 800 

blockade immunotherapy. Further studies to identify the specific antigenic epitopes 801 

are expected. Therefore, we prudently assume that LARC patients’ mutation burden 802 

and immune activity are correlated with the response to nCRT. This would help to 803 

develop personalized cellular adoptive immunotherapy strategies in the clinical 804 

settings to optimally combine radiation and checkpoint blockade with PD-1 and 805 

CTLA-4 antibodies to achieve the best therapeutic benefits. 806 

807 

Acknowledgments 808 

The authors would like to thank Dr. Bin Dong, the Department of Pathology, Peking 809 

University Cancer Hospital & Institute for her technical assistance. 810 

Author contributions 811 

Dengbo Ji and Haizhao Yi designed and performed the experiments, prepared the 812 

figures and draft the manuscript; Dakui Zhang, Tiancheng Zhan and Ming Li assisted 813 

with analysis of the whole- exome sequencing and RNA-seq data. Zhaowei Li, 814 

on April 7, 2020. © 2018 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on October 3, 2018; DOI: 10.1158/2326-6066.CIR-17-0630

38  

Jinying Jia, Meng Qiao, Jinhong Xia, Zhiwei Zhai and Can Song contributed to the 815 

performance of the experiments; Jin Gu supervised the work and wrote the 816 

manuscript. 817 

References 818 

1.  Braendengen  M,  Tveit  KM,  Berglund  A,  Birkemeyer  E,  Frykholm  G,  Pahlman  L,  et  al. 819 

Randomized phase  III study comparing preoperative radiotherapy with chemoradiotherapy 820 

in nonresectable rectal cancer. Journal of clinical oncology : official journal of the American 821 

Society of Clinical Oncology 2008;26:3687‐94 822 

2.  Colorectal  Cancer  Collaborative  G.  Adjuvant  radiotherapy  for  rectal  cancer:  a  systematic 823 

overview of 8,507 patients from 22 randomised trials. Lancet 2001;358:1291‐304 824 

3.  van  Gijn  W,  Marijnen  CA,  Nagtegaal  ID,  Kranenbarg  EM,  Putter  H,  Wiggers  T,  et  al. 825 

Preoperative  radiotherapy  combined with  total mesorectal  excision  for  resectable  rectal 826 

cancer: 12‐year  follow‐up of the multicentre, randomised controlled TME trial. The Lancet 827 

Oncology 2011;12:575‐82 828 

4.  Sauer R, Liersch T, Merkel S, Fietkau R, Hohenberger W, Hess C, et al. Preoperative versus 829 

postoperative chemoradiotherapy for locally advanced rectal cancer: results of the German 830 

CAO/ARO/AIO‐94 randomized phase III trial after a median follow‐up of 11 years. Journal of 831 

clinical  oncology  :  official  journal  of  the  American  Society  of  Clinical  Oncology 832 

2012;30:1926‐33 833 

5.  Engelen  SM, Maas M,  Lahaye MJ,  Leijtens  JW,  van  Berlo  CL,  Jansen  RL,  et  al. Modern 834 

multidisciplinary  treatment  of  rectal  cancer  based  on  staging  with  magnetic  resonance 835 

imaging  leads  to  excellent  local  control,  but  distant  control  remains  a  challenge.  Eur  J 836 

Cancer 2013;49:2311‐20 837 

6.  Bujko  K,  Glynne‐Jones  R,  Bujko M.  Does  adjuvant  fluoropyrimidine‐based  chemotherapy 838 

provide  a  benefit  for  patients  with  resected  rectal  cancer  who  have  already  received 839 

neoadjuvant  radiochemotherapy?  A  systematic  review  of  randomised  trials.  Ann  Oncol 840 

2010;21:1743‐50 841 

7.  Moertel CG, Fleming TR, Macdonald JS, Haller DG, Laurie JA, Goodman PJ, et al. Levamisole 842 

and fluorouracil for adjuvant therapy of resected colon carcinoma. The New England journal 843 

of medicine 1990;322:352‐8 844 

8.  Taal BG, Van Tinteren H, Zoetmulder FA, group N. Adjuvant 5FU plus levamisole in colonic or 845 

rectal cancer: improved survival in stage II and III. Br J Cancer 2001;85:1437‐43 846 

9.  Twelves C, Wong A, Nowacki MP, Abt M, Burris H, 3rd, Carrato A,  et al. Capecitabine  as 847 

adjuvant  treatment  for  stage  III  colon  cancer.  The  New  England  journal  of  medicine 848 

2005;352:2696‐704 849 

10.  Andre  T,  Boni  C,  Navarro M,  Tabernero  J,  Hickish  T,  Topham  C,  et  al.  Improved  overall 850 

survival with oxaliplatin, fluorouracil, and leucovorin as adjuvant treatment in stage II or III 851 

colon  cancer  in  the  MOSAIC  trial.  Journal  of  clinical  oncology  :  official  journal  of  the 852 

American Society of Clinical Oncology 2009;27:3109‐16 853 

11.  Petersen  SH, Harling H,  Kirkeby  LT, Wille‐Jorgensen  P, Mocellin  S. Postoperative  adjuvant 854 

on April 7, 2020. © 2018 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on October 3, 2018; DOI: 10.1158/2326-6066.CIR-17-0630

39  

chemotherapy  in  rectal  cancer  operated  for  cure.  Cochrane  Database  Syst  Rev 855 

2012:CD004078 856 

12.  Breugom  AJ,  Swets  M,  Bosset  JF,  Collette  L,  Sainato  A,  Cionini  L,  et  al.  Adjuvant 857 

chemotherapy after preoperative (chemo)radiotherapy and surgery for patients with rectal 858 

cancer:  a  systematic  review  and  meta‐analysis  of  individual  patient  data.  The  Lancet 859 

Oncology 2015;16:200‐7 860 

13.  Galon  J,  Costes A,  Sanchez‐Cabo  F,  Kirilovsky A, Mlecnik B,  Lagorce‐Pages  C,  et  al.  Type, 861 

density,  and  location  of  immune  cells  within  human  colorectal  tumors  predict  clinical 862 

outcome. Science 2006;313:1960‐4 863 

14.  Mlecnik  B,  Tosolini  M,  Kirilovsky  A,  Berger  A,  Bindea  G,  Meatchi  T,  et  al. 864 

Histopathologic‐based prognostic factors of colorectal cancers are associated with the state 865 

of the  local  immune reaction. Journal of clinical oncology : official  journal of the American 866 

Society of Clinical Oncology 2011;29:610‐8 867 

15.  Bindea G, Mlecnik B, Tosolini M, Kirilovsky A, Waldner M, Obenauf AC, et al. Spatiotemporal 868 

dynamics  of  intratumoral  immune  cells  reveal  the  immune  landscape  in  human  cancer. 869 

Immunity 2013;39:782‐95 870 

16.  Galon  J,  Angell  HK,  Bedognetti  D,  Marincola  FM.  The  continuum  of  cancer 871 

immunosurveillance:  prognostic,  predictive,  and  mechanistic  signatures.  Immunity 872 

2013;39:11‐26 873 

17.  Ribas  A.  Releasing  the  Brakes  on  Cancer  Immunotherapy.  The  New  England  journal  of 874 

medicine 2015;373:1490‐2 875 

18.  Hamid O, Robert C, Daud A, Hodi FS, Hwu WJ, Kefford R, et al. Safety and tumor responses 876 

with  lambrolizumab  (anti‐PD‐1)  in  melanoma.  The  New  England  journal  of  medicine 877 

2013;369:134‐44 878 

19.  Topalian  SL, Hodi  FS,  Brahmer  JR, Gettinger  SN,  Smith DC, McDermott DF,  et  al.  Safety, 879 

activity, and immune correlates of anti‐PD‐1 antibody in cancer. The New England journal of 880 

medicine 2012;366:2443‐54 881 

20.  Hodi  FS,  O'Day  SJ, McDermott  DF, Weber  RW,  Sosman  JA,  Haanen  JB,  et  al.  Improved 882 

survival with ipilimumab in patients with metastatic melanoma. The New England journal of 883 

medicine 2010;363:711‐23 884 

21.  Brahmer JR, Drake CG, Wollner I, Powderly JD, Picus J, Sharfman WH, et al. Phase I study of 885 

single‐agent  anti‐programmed  death‐1  (MDX‐1106)  in  refractory  solid  tumors:  safety, 886 

clinical  activity,  pharmacodynamics,  and  immunologic  correlates.  Journal  of  clinical 887 

oncology : official journal of the American Society of Clinical Oncology 2010;28:3167‐75 888 

22.  Le  DT, Uram  JN, Wang  H,  Bartlett  BR,  Kemberling H,  Eyring  AD,  et  al.  PD‐1  Blockade  in 889 

Tumors  with  Mismatch‐Repair  Deficiency.  The  New  England  journal  of  medicine 890 

2015;372:2509‐20 891 

23.  Rizvi  NA,  Hellmann  MD,  Snyder  A,  Kvistborg  P,  Makarov  V,  Havel  JJ,  et  al.  Cancer 892 

immunology. Mutational  landscape  determines  sensitivity  to  PD‐1  blockade  in  non‐small 893 

cell lung cancer. Science 2015;348:124‐8 894 

24.  Chan TA, Wolchok  JD, Snyder A. Genetic Basis  for Clinical Response  to CTLA‐4 Blockade  in 895 

Melanoma. The New England journal of medicine 2015;373:1984 896 

25.  Lee Y, Auh SL, Wang Y, Burnette B, Wang Y, Meng Y, et al. Therapeutic effects of ablative 897 

radiation  on  local  tumor  require  CD8+  T  cells:  changing  strategies  for  cancer  treatment. 898 

on April 7, 2020. © 2018 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on October 3, 2018; DOI: 10.1158/2326-6066.CIR-17-0630

40  

Blood 2009;114:589‐95 899 

26.  Twyman‐Saint Victor C, Rech AJ, Maity A, Rengan R, Pauken KE, Stelekati E, et al. Radiation 900 

and  dual  checkpoint  blockade  activate  non‐redundant  immune  mechanisms  in  cancer. 901 

Nature 2015;520:373‐7 902 

27.  Edge SB BD CC. AJCC Cancer Staging Manual. 7th Ed. Chicago: Springer‐Verlag 2010. 903 

28.  Li H, Durbin R.  Fast and accurate  short  read alignment with Burrows‐Wheeler  transform. 904 

Bioinformatics 2009;25:1754‐60 905 

29.  Li H, Durbin  R.  Fast  and  accurate  long‐read  alignment with  Burrows‐Wheeler  transform. 906 

Bioinformatics 2010;26:589‐95 907 

30.  Li  H,  Handsaker  B,  Wysoker  A,  Fennell  T,  Ruan  J,  Homer  N,  et  al.  The  Sequence 908 

Alignment/Map format and SAMtools. Bioinformatics 2009;25:2078‐9 909 

31.  Quinlan AR, Hall  IM. BEDTools: a  flexible suite of utilities  for comparing genomic features. 910 

Bioinformatics 2010;26:841‐2 911 

32.  Cibulskis  K,  Lawrence MS,  Carter  SL,  Sivachenko  A,  Jaffe  D,  Sougnez  C,  et  al.  Sensitive 912 

detection of somatic point mutations in impure and heterogeneous cancer samples. Nature 913 

biotechnology 2013;31:213‐9 914 

33.  Wang  K,  Li M, Hakonarson H.  ANNOVAR:  functional  annotation  of  genetic  variants  from 915 

high‐throughput sequencing data. Nucleic acids research 2010;38:e164 916 

34.  Koboldt DC,  Zhang Q,  Larson DE,  Shen D, McLellan MD,  Lin  L,  et  al. VarScan  2:  somatic 917 

mutation and copy number alteration discovery  in cancer by exome sequencing. Genome 918 

research 2012;22:568‐76 919 

35.  Weiskopf  D,  Angelo MA,  de  Azeredo  EL,  Sidney  J,  Greenbaum  JA,  Fernando  AN,  et  al. 920 

Comprehensive  analysis  of  dengue  virus‐specific  responses  supports  an  HLA‐linked 921 

protective  role  for CD8+ T  cells. Proceedings of  the National Academy of  Sciences of  the 922 

United States of America 2013;110:E2046‐53 923 

36.  Fuertes Marraco SA, Neubert NJ, Verdeil G, Speiser DE.  Inhibitory Receptors Beyond T Cell 924 

Exhaustion. Frontiers in immunology 2015;6:310 925 

37.  Sharma  P, Wagner  K, Wolchok  JD,  Allison  JP.  Novel  cancer  immunotherapy  agents with 926 

survival benefit: recent successes and next steps. Nat Rev Cancer 2011;11:805‐12 927 

38.  Newman AM, Liu CL, Green MR, Gentles AJ, Feng W, Xu Y, et al. Robust enumeration of cell 928 

subsets from tissue expression profiles. Nature methods 2015;12:453‐7 929 

39.  Wang  L,  Zhai  ZW,  Ji  DB,  Li  ZW,  Gu  J.  Prognostic  value  of  CD45RO(+)  tumor‐infiltrating 930 

lymphocytes  for  locally  advanced  rectal  cancer  following  30  Gy/10f  neoadjuvant 931 

radiotherapy. Int J Colorectal Dis 2015;30:753‐60 932 

40.  Esensten  JH,  Helou  YA,  Chopra  G,  Weiss  A,  Bluestone  JA.  CD28  Costimulation:  From 933 

Mechanism to Therapy. Immunity 2016;44:973‐88 934 

41.  Nanda  NK,  Birch  L,  Greenberg  NM,  Prins  GS.  MHC  class  I  and  class  II  molecules  are 935 

expressed  in  both  human  and  mouse  prostate  tumor  microenvironment.  Prostate 936 

2006;66:1275‐84 937 

42.  Klyushnenkova EN, Kouiavskaia DV, Berard CA, Alexander RB. Cutting edge: permissive MHC 938 

class  II  allele  changes  the  pattern  of  antitumor  immune  response  resulting  in  failure  of 939 

tumor rejection. J Immunol 2009;182:1242‐6 940 

43.  Sconocchia G, Eppenberger‐Castori S, Zlobec I, Karamitopoulou E, Arriga R, Coppola A, et al. 941 

HLA  class  II  antigen  expression  in  colorectal  carcinoma  tumors  as  a  favorable  prognostic 942 

on April 7, 2020. © 2018 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on October 3, 2018; DOI: 10.1158/2326-6066.CIR-17-0630

41  

marker. Neoplasia 2014;16:31‐42 943 

44.  Reits EA, Hodge JW, Herberts CA, Groothuis TA, Chakraborty M, Wansley EK, et al. Radiation 944 

modulates the peptide repertoire, enhances MHC class I expression, and induces successful 945 

antitumor immunotherapy. The Journal of experimental medicine 2006;203:1259‐71 946 

45.  Snyder A, Makarov V, Merghoub T, Yuan J, Zaretsky JM, Desrichard A, et al. Genetic basis for 947 

clinical  response  to CTLA‐4 blockade  in melanoma. The New England  journal of medicine 948 

2014;371:2189‐99 949 

46.  Yasuda  K,  Nirei  T,  Sunami  E,  Nagawa  H,  Kitayama  J.  Density  of  CD4(+)  and  CD8(+)  T 950 

lymphocytes  in  biopsy  samples  can  be  a  predictor  of  pathological  response  to 951 

chemoradiotherapy (CRT) for rectal cancer. Radiation oncology 2011;6:49 952 

47.  Anitei MG, Zeitoun G, Mlecnik B, Marliot F, Haicheur N, Todosi AM, et al. Prognostic and 953 

predictive values of the immunoscore in patients with rectal cancer. Clinical cancer research : 954 

an official journal of the American Association for Cancer Research 2014;20:1891‐9 955 

48.  Jiang  W,  Chan  CK,  Weissman  IL,  Kim  BYS,  Hahn  SM.  Immune  Priming  of  the  Tumor 956 

Microenvironment by Radiation. Trends in cancer 2016;2:638‐45 957 

49.  Minoo P, Zlobec  I, Peterson M, Terracciano  L,  Lugli A. Characterization of  rectal, proximal 958 

and  distal  colon  cancers  based  on  clinicopathological,  molecular  and  protein  profiles. 959 

International journal of oncology 2010;37:707‐18 960 

50.  Perez‐Ruiz  E,  Berraondo  P.  Immunological  Landscape  and  Clinical Management  of  Rectal 961 

Cancer. Frontiers in immunology 2016;7:61 962 

51.  Berntsson J, Svensson MC, Leandersson K, Nodin B, Micke P, Larsson AH, et al. The clinical 963 

impact of tumour‐infiltrating lymphocytes in colorectal cancer differs by anatomical subsite: 964 

A cohort study. International journal of cancer 2017;141:1654‐66 965 

52.  Wang H, Yao H, Li C, Liang L, Zhang Y, Shi H, et al. PD‐L2 expression  in colorectal cancer: 966 

Independent  prognostic  effect  and  targetability  by  deglycosylation.  Oncoimmunology 967 

2017;6:e1327494 968 

53.  Cancer Genome Atlas N.  Comprehensive molecular  characterization of human  colon  and 969 

rectal cancer. Nature 2012;487:330‐7 970 

54.  Blank C, Brown  I, Peterson AC, Spiotto M,  Iwai Y, Honjo T, et al. PD‐L1/B7H‐1  inhibits  the 971 

effector phase of  tumor  rejection by T cell  receptor  (TCR)  transgenic CD8+ T cells. Cancer 972 

research 2004;64:1140‐5 973 

55.  Freeman GJ, Long AJ, Iwai Y, Bourque K, Chernova T, Nishimura H, et al. Engagement of the 974 

PD‐1 immunoinhibitory receptor by a novel B7 family member leads to negative regulation 975 

of lymphocyte activation. The Journal of experimental medicine 2000;192:1027‐34 976 

56.  Curran  MA,  Montalvo  W,  Yagita  H,  Allison  JP.  PD‐1  and  CTLA‐4  combination  blockade 977 

expands infiltrating T cells and reduces regulatory T and myeloid cells within B16 melanoma 978 

tumors. Proceedings of the National Academy of Sciences of the United States of America 979 

2010;107:4275‐80 980 

981 

on April 7, 2020. © 2018 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on October 3, 2018; DOI: 10.1158/2326-6066.CIR-17-0630

42  

Table 1. Reactome pathway analysis of with nRT vs. without nRT (GSE15781) and Mutation related pathways (TCGA) 982 

983 

With nRT vs. without nRT(GSE15781) Mutation related pathways(TCGA)

Pathway name Entities pValue Entities FDR Pathway name Entities pValue Entities FDR

Interferon gamma signaling 1.11E-16

1.21E-14

Developmental Biology

5.67E-11 2.34E-08

Antigen Presentation: Folding, assembly

and peptide loading of class I MHC

1.11E-16

1.21E-14

Alpha-defensins

8.46E-08 2.61E-05

Immunoregulatory interactions between

a Lymphoid and a non-Lymphoid cell

1.11E-16

1.21E-14

NCAM signaling for neurite out-growth

2.39E-06 5.89E-04

Class I MHC mediated antigen

processing & presentation

1.11E-16

1.21E-14

Interleukin-2 family signaling

2.17E-05 0.002010602

Cytokine Signaling in Immune system 1.11E-16

1.21E-14

Signalling to RAS

3.58E-05 0.002010602

Receptor binding chemokines 1.34E-13

1.34E-11

RAF/MAP kinase cascade 5.59E-05 0.002010602

PD-1 signaling 7.64E-07

6.65E-05

Interleukin receptor SHC signaling 9.43E-05 0.002545513

Peptide ligand-binding receptors 1.71E-06

1.39E-04

Interleukin-19,20,22,24,26,28 and 29

signaling

1.47E-04 0.003831806

Costimulation by the CD28 family 2.71E-05

0.00195

Interleukin-3, 5 and GM-CSF signaling

2.05E-04 0.005137273

984 

985 

986 

987 

on April 7, 2020. ©

2018 Am

erican Association for C

ancer Research.

cancerimm

unolres.aacrjournals.org D

ownloaded from

Author m

anuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Author M

anuscript Published O

nlineFirst on O

ctober 3, 2018; DO

I: 10.1158/2326-6066.CIR

-17-0630

43  

Figure Legends 988 

Figure 1. nCRT induces immune activation of LARC. (A) Unsupervised clustering 989 

highlighting genome-wide changes in gene expression. Rectal cancer samples were 990 

clustered according to the expression of 342 differentially expressed immune genes 991 

(p<0.05) between 13 pre- and 9 post-nCRT samples (GSE 15781). Rectal cancer 992 

samples are across the horizontal axis, with 1 sample expression pattern shown in 993 

each column. Gene expression values range from green (low expression) to red (high 994 

expression). (B) Venn diagram showing significant overlaps of core genes from the 995 

differentially expressed genes (red circle), immune related genes (blue circle), and 996 

CD28 costimulatory related genes (green circle). (C) Unsupervised clustering for 17 997 

differentially expressed immune genes for CD28 co-stimulatory signals (p<0.05) 998 

between pre- and post-nCRT. (D) Unsupervised clustering for T-cell differentiation 999 

and activation markers between pre- and post-nCRT. (E) Enriched pathways for 1000 

differentially expressed immune genes after nCRT using GSEA. The twenty most 1001 

significant pathways and corresponding normalized enrichment scores (NES) are 1002 

shown. p ≤ 0.01, FDR q< 0.25. using Enrichment Score Calculation, Permutation test 1003 

and Multiple hypothesis test. 1004 

1005 

Figure 2. nCRT influences the neoantigen landscape of LARC. (A) The quantity of 1006 

neoantigens and mutations per tumor prior to nCRT (Cohort 1; n=14), using Pearson 1007 

correlation. (B) The quantity of neoantigens and mutations per tumor post-nCRT 1008 

(Cohort 1, n=9), using Pearson correlation. (C) The mutation burden in patients post 1009 

nCRT (n=7) and patients without nCRT (n=41) (from TCGA data), using Student t 1010 

test. (D) The candidate neoantigens and mutation burden (from TCGA data), using 1011 

Pearson correlation. (E) The neoantigens binding to the T cell receptor in patients post 1012 

on April 7, 2020. © 2018 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on October 3, 2018; DOI: 10.1158/2326-6066.CIR-17-0630

44  

nCRT and patients without nCRT (from TCGA data), using Mann-Whitney test. 1013 

Statistical significance was set at p < 0.05. 1014 

1015 

Figure 3. RT combined with anti–PD-1 enhances tumor growth control in a 1016 

mouse model. (A). Schematic of the experimental setup. Yellow arrow: location of 1017 

transplantation. Timeline starts from original tumor implantation (day 0). Black 1018 

arrows: treatments given. (B) MRI images of combination RT+PD1 (RT combined 1019 

with anti–PD1)-treated tumors and baseline/control tumors. (C-D) Total tumor growth 1020 

for MC38 tumors after the indicated treatment. Radiotherapy: RT (n=9); Anti–PD-1: 1021 

PD1 (n=8); Anti–PD-L1: PDL1 (n=9); RT combined with anti–PD-1 or anti–PD-L1: 1022 

RT+PD1 (n=8) and RT+PDL1 (n=9), respectively. C: normalized values; D: raw 1023 

values. For normalization, volumes were divided by average of untreated controls 1024 

(V/Vcont) to account for differences in growth between different treated groups. 1025 

Using A mixed effect linear model. (E) Survival after RT and/or anti–PD-1 (left) or 1026 

anti–PD-L1 (right). Shown are overall p-values. The p-values for RT+PD1 and RT or 1027 

anti–PD-1 alone are p=0.046 and p=0.042, respectively. Control is an 1028 

isotype-matched antibody. Using the log-rank test, statistical significance was set at p 1029 

< 0.05. 1030 

1031 

Figure 4. Mutation burden correlated with patients’ response to nCRT. Paired 1032 

pre- and post-nCRT tumors from two LARC patients. Tumor regression grade (TRG) 1033 

of case 4 with higher mutation number is 0, complete regression, better response. 1034 

TRG of case 3 with lower mutation number is 3, poor response. The H&E staining 1035 

shows a typical rectal adenocarcinoma. The MRI images show the pre-nCRT and 1036 

post-nCRT tumors. 1037 

on April 7, 2020. © 2018 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on October 3, 2018; DOI: 10.1158/2326-6066.CIR-17-0630

45  

1038 

Figure 5. Mutation burden and neoantigens of LARC correlated with the 1039 

response to nCRT. (A-B) Cohort 1 (n=14). (A) Mutation numbers in pre-nCRT 1040 

biopsies with the different tumor regression grade, using Pearson correlation. (B) The 1041 

mutation-associated neoantigens in pre-nCRT biopsies with the different tumor 1042 

regression grade, using Pearson correlation. (C-F) Cohort 2 (n=42). (C) Tumor 1043 

mutation burden (TMB) in pre-nCRT ctDNA. (D) The correlation between TMB in 1044 

pre-nCRT ctDNA and the tumor regression grade, using Pearson correlation. (E) The 1045 

TMB in pCR patients (n=6) and non-pCR patients (n=36), using Student t test. (F) 1046 

The TMB in residual tumor cells <30% group (n=24) and in >30% group (n=18), 1047 

using Student t test. Statistical significance was set at p < 0.05. 1048 

1049 

Figure 6. Association between PD-L1 and PD-1 expression and prognosis with 1050 

CRC using IHC (Cohort 4). (A) Expression of PD-1 and PD-L1 in the indicated 1051 

CRC tissues using IHC analysis. Positive staining shows as brown. (B) Overall 1052 

survival. Kaplan–Meier analysis of TNM stage I-III CRC patients in cohort 4. 1053 

Number of patients per group indicated on graph. (C) Kaplan–Meier analysis of the 1054 

correlation between PD-L1 expression and disease-free survival in patients with rectal 1055 

cancer in cohort 4. Number of patients per group indicated on graph. (D) 1056 

Kaplan–Meier analysis of the correlation between PDL1 expression and OS in 1057 

patients with rectal cancer in cohort 4. Number of patients per group indicated on 1058 

graph. Using the log-rank test, statistical significance was set at p < 0.05. 1059 

1060 

Figure 7. Expression of PD-L1 and PD-1 in post-nRT rectal cancer tissue samples 1061 

and therapeutic outcome for nRT using IHC (Cohort 5). (A) Representative 1062 

on April 7, 2020. © 2018 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on October 3, 2018; DOI: 10.1158/2326-6066.CIR-17-0630

46  

images of PD-L1 expression on tumor cells and TILs (B) Representative images of 1063 

PD-1 expression on tumor cells and TILs, as well as CD8 expression on TILs (C) 1064 

Kaplan–Meier analysis of the correlation between PD-L1 expression and disease-free 1065 

survival in patients with low CD8 expression in cohort 5. PD-L1–negative n=67; 1066 

PD-L1–positive n=45. Using the log-rank test, statistical significance was set at p < 1067 

0.05. 1068 

1069 

1070 

on April 7, 2020. © 2018 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on October 3, 2018; DOI: 10.1158/2326-6066.CIR-17-0630

on April 7, 2020. © 2018 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on October 3, 2018; DOI: 10.1158/2326-6066.CIR-17-0630

on April 7, 2020. © 2018 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on October 3, 2018; DOI: 10.1158/2326-6066.CIR-17-0630

on April 7, 2020. © 2018 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on October 3, 2018; DOI: 10.1158/2326-6066.CIR-17-0630

on April 7, 2020. © 2018 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on October 3, 2018; DOI: 10.1158/2326-6066.CIR-17-0630

on April 7, 2020. © 2018 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on October 3, 2018; DOI: 10.1158/2326-6066.CIR-17-0630

on April 7, 2020. © 2018 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on October 3, 2018; DOI: 10.1158/2326-6066.CIR-17-0630

on April 7, 2020. © 2018 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on October 3, 2018; DOI: 10.1158/2326-6066.CIR-17-0630

Published OnlineFirst October 3, 2018.Cancer Immunol Res   Dengbo Ji, Haizhao Yi, Dakui Zhang, et al.   Following Neoadjuvant ChemoradiotherapySomatic Mutations and Immune Alternation in Rectal Cancer

  Updated version

  10.1158/2326-6066.CIR-17-0630doi:

Access the most recent version of this article at:

  Material

Supplementary

  C1

http://cancerimmunolres.aacrjournals.org/content/suppl/2018/10/03/2326-6066.CIR-17-0630.DAccess the most recent supplemental material at:

  Manuscript

Authorbeen edited. Author manuscripts have been peer reviewed and accepted for publication but have not yet

   

   

   

  E-mail alerts related to this article or journal.Sign up to receive free email-alerts

  Subscriptions

Reprints and

  [email protected] at

To order reprints of this article or to subscribe to the journal, contact the AACR Publications

  Permissions

  Rightslink site. Click on "Request Permissions" which will take you to the Copyright Clearance Center's (CCC)

.http://cancerimmunolres.aacrjournals.org/content/early/2018/10/03/2326-6066.CIR-17-0630To request permission to re-use all or part of this article, use this link

on April 7, 2020. © 2018 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on October 3, 2018; DOI: 10.1158/2326-6066.CIR-17-0630