Purpose: We have previously demonstrated that patients with metastatic colorectal cancer who exhibit immune responses to a dendritic cell (DC) vaccine have superior recurrence-free survival following surgery, compared with patients in whom responses do not occur. We sought to characterize the patterns of T-lymphocyte infiltration and somatic mutations in metastases that are associated with and predictive of response to the DC vaccine.

Experimental Design: Cytotoxic, memory, and regulatory T cells in resected metastases and surrounding normal liver tissue from 22 patients (11 responders and 11 nonresponders) were enumerated by immunohistochemistry prior to vaccine administration. In conjunction with tumor sequencing, the combined multivariate and collapsing method was used to identify gene mutations that are associated with vaccine response. We also derived a response prediction score for each patient using his/her tumor genotype data and variant association effect sizes computed from the other 21 patients; greater weighting was placed on gene products with cell membrane–related functions.

Results: There was no correlation between vaccine response and intratumor, peritumor, or hepatic densities of T-cell subpopulations. Associated genes were found to be enriched in the PI3K/Akt/mTOR signaling axis (P < 0.001). Applying a consistent prediction score cutoff over 22 rounds of leave-one-out cross-validation correctly inferred vaccine response in 21 of 22 patients (95%).

Conclusions: Adjuvant DC vaccination has shown promise as a form of immunotherapy for patients with metastatic colorectal cancer. Its efficacy may be influenced by somatic mutations that affect pathways involving PI3K, Akt, and mTOR, as well as tumor surface proteins. Clin Cancer Res; 23(2); 399–406. ©2016 AACR.

Translational Relevance

It has been well established that the immune system can effectively and specifically destroy cancer cells. Immunotherapies are intended to augment this natural aptitude for targeting tumor antigens. Dendritic cells serve as one of the body's most important antigen presenters that stimulate the adaptive immune system. Among patients with metastatic colorectal cancer, approximately half respond to a dendritic cell vaccine in the adjuvant setting following surgical resection of metastases. These patients have better recurrence-free survival compared with patients who do not respond. In the present study, resected colorectal cancer liver metastases were sequenced to identify likely mechanisms driven by somatic mutations that influence response to the vaccine. Our bioinformatic implication of PI3K/Akt/mTOR signaling and various plasma membrane proteins provides both a useful model for vaccine response inference in precision medicine (95% accurate) and new experimental directions for exploring cancer resistance against immunologic elimination.

Colorectal cancer is the fourth leading cause of cancer death in the world (1). About one in three patients diagnosed with colorectal cancer dies from metastatic disease (2). The majority of metastases develop in the liver and lung. Although metastases can be completely resected in many patients, 60% to 80% of these patients eventually die due to the growth of small metastases that were undetectable at the time of surgery (3, 4). Currently, the addition of adjuvant chemotherapy has limited impact on patient survival (5, 6). The pressing need for better treatments and growing recognition of the immune system's role in cancer progression have prompted tremendous efforts to develop immunotherapies for colorectal cancer (7).

Immunotherapies aim to either enhance the antitumor immune response or prevent immunosurveillance suppression by cancer cells. Whole tumor vaccines for colorectal cancer have not been shown to confer survival benefit. Since only a tiny fraction of proteins in an autologous tumor vaccine is specific to cancer cells, dilution by self-antigens is believed to make the vaccine poorly immunogenic (8). Specific colorectal cancer tumor-associated antigens have subsequently been identified and used as peptide vaccines (9–14). However, the extent of antigen presentation varies widely depending on patients' HLA type (15). Dendritic cell (DC) vaccines overcome the limitations of these two earlier vaccine approaches. Following ex vivo stimulation by tumor lysate, DCs are not confined to process any specific antigen chosen in advance and can also activate a tumor-specific T-lymphocyte response (16).

We have previously demonstrated that patients with metastatic colorectal cancer who exhibit immune responses to a DC vaccine have superior 5-year recurrence-free survival rate following surgical resection of liver metastases compared with counterparts in whom immune responses do not occur (63% vs. 18%, P = 0.037; ref. 17). Since T-cell densities in the tumor microenvironment are well-known to correlate with survival of patients who have either primary or metastatic colorectal cancer (18, 19), we now investigated potential relationships between DC vaccine response and densities of cytotoxic, memory, and regulatory T cells in the context of metastatic colorectal cancer. Furthermore, we pursued genomic analyses. A recent phase II study of pembrolizumab, an anti-programmed death 1 immune checkpoint inhibitor, for colorectal cancer showed that clinical benefit is more likely to be observed in patients with tumors carrying higher mutation burdens due to deficiency in DNA mismatch repair (MMR; ref. 20); however, the prevalence of MMR deficiency among metastatic colorectal cancer cases is only 3.5% (21). Therefore, DC vaccination may be an effective therapy for a larger portion (17) of such patients. To evaluate the feasibility of precision immunotherapy using this approach, we examined mutation patterns and constructed a vaccine response prediction model on the basis of colorectal cancer metastasis sequencing.

Details on the preparation of DC vaccines, detection of immune response, and approval for the study of human subjects have been previously reported (17). Briefly, 22 patients with colorectal cancer underwent surgical resection of liver metastases (Table 1) within 1 to 2 months of diagnosis at the Norris Cotton Cancer Center. Tumor lysates were derived from resected samples. These lysates were then added to immature DC cultures differentiated from autologous leukopheresed monocytes that had been supplemented with recombinant human granulocyte macrophage colony-stimulating factor (GM-CSF) and recombinant human IL4. At 4, 7, and 10 weeks after surgery, tumor lysate-pulsed DCs (5 × 106 cells in 0.5 mL) were injected into two inguinal lymph nodes of each patient. All patients were confirmed to have no detectable residual disease by computed tomography (CT) scan prior to the initial vaccine injection. Development of immune response was defined as observing significantly greater secretion of IFNγ (22) or induced proliferation of T lymphocytes (23, 24) by peripheral blood mononuclear cells 1 week following the third autologous tumor lysate-pulsed DC vaccination, compared with unpulsed DC stimulation.

Table 1.

Characteristics of DC vaccine responders and nonresponders

Responders (n = 11)Nonresponders (n = 11)P
Age 61.6 (7.9) 62.5 (11.2) 0.835 
Male 64% 82% 0.346 
Carcinoembryonic antigen level, ng/mL 32.1 (57.4) 18.3 (28.5) 0.483 
Size of largest metastasis, cm 3.1 (1.8) 4.6 (3.4) 0.220 
Number of metastases 1.5 (0.7) 1.6 (0.7) 0.519 
Lymph node–positive primary tumor 55% 45% 0.670 
Synchronous metastasis detection 45% 73% 0.200 
Fong clinical risk score 1.7 (1.0) 2.3 (1.2) 0.252 
Cytotoxic (CD8) T cells, per mm2 
 Tumor interior 13.3 (12.4) 9.0 (6.8) 0.323 
 Tumor periphery 44.1 (13.0) 37.4 (16.2) 0.289 
 Surrounding nonadjacent liver 10.5 (4.4) 8.5 (2.9) 0.201 
Memory (CD45RO) T cells, per mm2 
 Tumor interior 24.8 (11.5) 17.5 (11.9) 0.159 
 Tumor periphery 80.4 (19.6) 70.9 (24.3) 0.311 
 Surrounding nonadjacent liver 15.9 (9.1) 12.9 (4.8) 0.337 
Regulatory (FoxP3) T cells, per mm2 
 Tumor interior 6.1 (4.3) 5.5 (3.2) 0.693 
 Tumor periphery 8.1 (3.3) 7.6 (5.3) 0.777 
 Surrounding nonadjacent liver 0.3 (0.5) 0.2 (0.4) 0.613 
Mutations per megabase 39.1 (2.3) 40.6 (2.1) 0.135 
Responders (n = 11)Nonresponders (n = 11)P
Age 61.6 (7.9) 62.5 (11.2) 0.835 
Male 64% 82% 0.346 
Carcinoembryonic antigen level, ng/mL 32.1 (57.4) 18.3 (28.5) 0.483 
Size of largest metastasis, cm 3.1 (1.8) 4.6 (3.4) 0.220 
Number of metastases 1.5 (0.7) 1.6 (0.7) 0.519 
Lymph node–positive primary tumor 55% 45% 0.670 
Synchronous metastasis detection 45% 73% 0.200 
Fong clinical risk score 1.7 (1.0) 2.3 (1.2) 0.252 
Cytotoxic (CD8) T cells, per mm2 
 Tumor interior 13.3 (12.4) 9.0 (6.8) 0.323 
 Tumor periphery 44.1 (13.0) 37.4 (16.2) 0.289 
 Surrounding nonadjacent liver 10.5 (4.4) 8.5 (2.9) 0.201 
Memory (CD45RO) T cells, per mm2 
 Tumor interior 24.8 (11.5) 17.5 (11.9) 0.159 
 Tumor periphery 80.4 (19.6) 70.9 (24.3) 0.311 
 Surrounding nonadjacent liver 15.9 (9.1) 12.9 (4.8) 0.337 
Regulatory (FoxP3) T cells, per mm2 
 Tumor interior 6.1 (4.3) 5.5 (3.2) 0.693 
 Tumor periphery 8.1 (3.3) 7.6 (5.3) 0.777 
 Surrounding nonadjacent liver 0.3 (0.5) 0.2 (0.4) 0.613 
Mutations per megabase 39.1 (2.3) 40.6 (2.1) 0.135 

NOTE: Continuous and categorical patient characteristics are displayed as means (SE) and percentages, respectively. “Mutations per megabase” refers to the mutation burden across all targeted sequencing gene regions. P values indicate the predictive utility of each characteristic for dendritic cell vaccine response, as evaluated by univariate logistic regression.

Inspection of T-cell infiltration

The resected specimens were formalin-fixed, paraffin-preserved, serially sectioned, and confirmed to contain metastatic tumor by hematoxylin and eosin staining. We performed immunohistochemical analysis on tumor-containing slides using the Leica BOND-MAX automated IHC system (Leica Microsystems) along with primary antibodies to human cytotoxic T cells (CD8, Novacastra #NCL-CD8-4B11), human memory T cells (CD45RO, Novacastra #NCL-L-UCHL1), and human regulatory T cells (FoxP3, BioLegend #623802). These subpopulations were specifically chosen to examine both immune-promoting (cytotoxic and memory) and immunosuppressive (regulatory) T-cell roles in the tumor microenvironment. Blinded to the clinical characteristics of patients, an experienced gastrointestinal pathologist (A.A. Suriawinata) enumerated the positively stained CD8, CD45RO, and FoxP3 T cells within the tumor, at the tumor periphery, and in surrounding normal liver tissue. Five high-power fields with a field area of 0.24 mm2 were counted at each location, and average counts were recorded as cells/mm2. We tested for associations between response to the DC vaccine and densities of T-cell subpopulations as well as other clinical metrics using logistic regression (Table 1).

Targeted gene sequencing

The 22 metastatic tumor samples were submitted to the Genomics & Molecular Biology Shared Resource at Dartmouth to sequence 541 genes (Supplementary Table S1) from the HaloPlex Cancer Research Panel (Agilent Technologies). DNA was isolated from samples using the QiaAmp DNA FFPE Tissue Kit (Qiagen) and digested with restriction enzymes to create a library of DNA fragments. Digests were hybridized to HaloPlex probes for target enrichment and sample barcoding. Targeted DNA fragments were then captured by magnetic beads, amplified, and purified using AMPure XP beads (Beckman Coulter). The final library sizes were 150 to 550 bp, as determined by Qubit (Thermo Fisher Scientific) after quality control by Fragment Analyzer (Advanced Analytical). Sequencing was performed using the Ion Proton System (Thermo Fisher Scientific) with the Ion PI Template OT2 200 v2 Kit, the Ion PI Sequencing 200 v2 Kit, and a PI v2 chip. Sequenced reads in FASTQ file format (25) were aligned to the hg19 reference genome using TMAP (https://github.com/iontorrent/TMAP). The Genome Atlas Toolkit (GATK; ref. 26) was used to call variants in VCF format (27), filtering for quality score > 100 and read depth > 30 in all 22 patients. Likely germline variants were removed from consideration by matching their position, reference allele, and alternative allele with records in the National Center for Biotechnology Information (NCBI) Short Genetic Variations database (28).

Gene associations and pathway enrichment

Sequencing genotype data facilitate the calling of both common and rare variants. To identify associations between genes containing these variants and a given outcome, collapsing approaches that test for the collective presence of variants within genes have been developed to optimize the detection of rare variant effects (29, 30). We identified associated genes using the Combined Multivariate and Collapsing (CMC) method, which has been shown to be more powerful than previous single-marker tests and more robust to misclassification of relevant variants than previous multiple-marker tests (30). The method ranks genes by computing Hotelling's t2 statistics and corresponding P values (Supplementary Table S2). Given the small number of subjects in this study, we set a liberal cutoff for choosing genes (41 genes with P < 0.1) to evaluate statistical enrichment of biologic pathways. For every curated BioCarta, KEGG, PID, and Reactome pathway (set C2) in the Molecular Signatures Database (MSigDB; ref. 31), the hypergeometric test was used to assess the probability that 41 random genes from 8,384 genes in 1,291 pathways (in MSigDB at the time of this analysis) would better represent the pathway than our top 41 genes. These probabilities can be interpreted as P values and have been adjusted for Benjamini–Hochberg false discovery rate in multiple hypothesis testing (32). In addition, we accounted for spurious enrichment due to pathway size bias and the fact that most subsets of genes on the HaloPlex Cancer Research Panel should by default be overrepresented in cancer-related pathways. Null distributions of pathway enrichment were generated by randomly sampling 41 genes from the original panel gene list and applying the hypergeometric test over 1,000 permutations. We retained only pathways (Table 2) whose observed enrichment values surpassed the 95th percentile of their respective null distributions (33).

Table 2.

Pathway enrichment results

Pathway nameSizePRandomization rankImplicated components
1. KEGG: mTOR signaling pathway 52 6.25 × 10–5 0.964 MTOR, AKT3, RPS6KA2, TSC2 
2. REACTOME: PI3K cascade 56 7.31 × 10–5 0.977 MTOR, AKT3, FGFR2, TSC2 
3. REACTOME: Signaling by the B-cell receptor 126 1.04 × 10–4 0.967 MTOR, AKT3, MALT1, CD79B, TSC2 
4. REACTOME: PIP3 activates AKT signaling 29 1.34 × 10–4 0.972 MTOR, AKT3, TSC2 
5. BIOCARTA: PITX2 pathway 15 6.31 × 10–4 0.968 TRRAP, CTNNB1 
6. PID: AR pathway 61 1.04 × 10–3 0.985 NCOA4, CTNNB1, AR 
7. REACTOME: PKB-mediated events 29 2.41 × 10–3 0.966 MTOR, TSC2 
Pathway nameSizePRandomization rankImplicated components
1. KEGG: mTOR signaling pathway 52 6.25 × 10–5 0.964 MTOR, AKT3, RPS6KA2, TSC2 
2. REACTOME: PI3K cascade 56 7.31 × 10–5 0.977 MTOR, AKT3, FGFR2, TSC2 
3. REACTOME: Signaling by the B-cell receptor 126 1.04 × 10–4 0.967 MTOR, AKT3, MALT1, CD79B, TSC2 
4. REACTOME: PIP3 activates AKT signaling 29 1.34 × 10–4 0.972 MTOR, AKT3, TSC2 
5. BIOCARTA: PITX2 pathway 15 6.31 × 10–4 0.968 TRRAP, CTNNB1 
6. PID: AR pathway 61 1.04 × 10–3 0.985 NCOA4, CTNNB1, AR 
7. REACTOME: PKB-mediated events 29 2.41 × 10–3 0.966 MTOR, TSC2 

NOTE: Genes identified to be strongly associated with DC vaccine response by the CMC method were assessed for enrichment (“Implicated Components”) of curated pathways in MSigDB using the hypergeometric test. “P values” have been adjusted using the Benjamini–Hochberg false discovery rate method. “Size” describes the total number of genes comprising each pathway. “Randomization Rank” denotes the fraction of 1,000 null enrichment values, computed using randomly sampled genes from the sequencing panel, that are less than the observed enrichment. Results have been filtered for Randomization Rank > 95th percentile.

Abbreviations: AR, androgen receptor; PIP3, phosphatidylinositol (3,4,5)-trisphosphate; PITX2, paired-like homeodomain transcription factor 2; PKB, protein kinase B which is also synonymous with AKT.

Prediction of DC vaccine response

Although the CMC method is superior to single-marker tests at detecting gene-level associations, the former masks association direction by combining variants that both positively and negatively influence the outcome of interest (34). Therefore, examining individual markers is more informative for predicting vaccine response. We derived a response prediction score for each patient using his/her tumor genotype data and variant association effect sizes computed from the other 21 patients:

Si is the prediction score for individual i, tk is the t statistic for difference in allele frequency of variant k between vaccine responders and nonresponders excluding individual i, all n variants chosen for consideration have tk with corresponding P < 0.005, wk is an assigned weight for variant k, and xk,i is the allele count (0, 1, or 2) of variant k carried by individual i. Under this construction, Si is more positive for individuals with tumors that have an abundance of variants associated with vaccine response; conversely, Si is more negative for individuals with tumors that have an abundance of variants associated with lack of vaccine response. We then performed 22 rounds of leave-one-out cross-validation. Inference of response to the DC vaccine for each individual i was determined by whether Si > C, a consistent cutoff applied to every round of cross-validation. Vaccine response prediction accuracy (percentage correct out of 22 predictions) was recorded as the highest achieved accuracy through incrementing C along the range of 22 Si values. We repeated this procedure for various weighting schemes of wk. First, we set wk = 1 for all variants. With respect to each curated MSigDB pathway, we then differentially weighted variants according to pathway membership of their spanning genes (wk = 8 for inclusion, wk = 1 otherwise). The choice of 8 was arbitrary, as results do not change for integer weights between 4 and 15. Finally, pathways were ranked on the basis of the prediction accuracy of their annotation-guided Si calculations, with top findings outlined in Table 3. Since leave-one-out cross-validation requires determining variant associations using only 21 of the 22 total individuals, effect sizes for some variants fluctuate between rounds of cross-validation. The upweighted components of each pathway listed in Table 3 have association effect sizes that are significant (P < 0.005) and carry the same sign in all 22 rounds of cross-validation.

Table 3.

Top pathway-guided predictions of DC vaccine response

Pathway nameAccuracyUpweighted pathway components
No pathway weighting 73% All weights in Eq. A were set to 1 
1. KEGG: Extracellular matrix receptor interactions 77% CD36 (7:80299307/G>GT/+), ITGA10 (1:145527997/AG>A/−), ITGA10 (1:145534180/A>AT/+), ITGB3 (17:45367575/AC>A/−), ITGB3 (17:45367575/AC>A/−), COL1A1 (17:48271806/AC>A/−), COL1A1 (17:48275531/AG>A/−), FN1 (2:216288075/GC>G/−) 
2. KEGG: Natural killer cell–mediated cytotoxicity 77% LCK (1:32739904/GC>G/−), PIK3R1 (5:67522514/A>G/+), PIK3CD (1:9775549/TC>T/−), PIK3CD (1:9775564/TG>T/+), ITGB2 (21:46309974/CG>C/−), ITGB2 (21:46323312/G>C/+) 
3. PID: αVβ3 integrin pathway 77% VEGFR2 (4:55971036/G>C/+), PIK3R1 (5:67522514/A>G/+), GPR124 (8:37693120/TC>T/−), IGF1R (15:99459983/TG>T/−), IGF1R (15:99478596/GA>G/+), ITGB3 (17:45367575/AC>A/−), ITGB3 (17:45367575/AC>A/−), COL1A1 (17:48271806/AC>A/−), COL1A1 (17:48275531/AG>A/−), FN1 (2:216288075/GC>G/−), TGFBR2 (3:30715604/C>CG/+) 
4. PID: Caspase pathway 77% MAP3K1 (5:56179436/G>C/−), NUMA1 (11:71726363/AG>A/−), SREBF1 (17:17720826/TG>T/−) 
5. PID: SHP2 pathway 77% VEGFR2 (4:55971036/G>C/+), LCK (1:32739904/GC>G/−), PIK3R1 (5:67522514/A>G/+), PDGFRB (5:149506102/AG>A/−), PDGFRB (5:149515110/GC>G/−), BDNF (11:27722583/TG>T/−), IGF1R (15:99459983/TG>T/−), IGF1R (15:99478596/GA>G/+) 
6. REACTOME: Cell surface interactions at the vascular wall 77% LCK (1:32739904/GC>G/−), PIK3R1 (5:67522514/A>G/+), ITGB3 (17:45367575/AC>A/−), ITGB3 (17:45367575/AC>A/−), COL1A1 (17:48271806/AC>A/−), COL1A1 (17:48275531/AG>A/−), ITGB2 (21:46309974/CG>C/−), ITGB2 (21:46323312/G>C/+), FN1 (2:216288075/GC>G/−) 
7. REACTOME: PPARα activates gene expression 77% CD36 (7:80299307/G>GT/+), NCOA2 (8:71069295/GC>G/−), CREBBP (16:3842089/TG>T/−), SREBF1 (17:17720826/TG>T/−), ANGPTL4 (19:8434086/A>AC/+) 
8. PID: αMβ2 integrin pathway 82% LCK (1:32739904/GC>G/−), ITGB2 (21:46309974/CG>C/−), ITGB2 (21:46323312/G>C/+) 
9. PID: Integrin 1 pathway 82% ITGA10 (1:145527997/AG>A/−), ITGA10 (1:145534180/A>AT/+), COL1A1 (17:48271806/AC>A/−), COL1A1 (17:48275531/AG>A/−), FN1 (2:216288075/GC>G/−) 
10. REACTOME: Metabolism of amino acids and derivatives 86% GLS2 (12:56868601/TC>T/−), MTR (1:237025593/GA>G/−) 
Union of above pathways 95% Union of above components 
Pathway nameAccuracyUpweighted pathway components
No pathway weighting 73% All weights in Eq. A were set to 1 
1. KEGG: Extracellular matrix receptor interactions 77% CD36 (7:80299307/G>GT/+), ITGA10 (1:145527997/AG>A/−), ITGA10 (1:145534180/A>AT/+), ITGB3 (17:45367575/AC>A/−), ITGB3 (17:45367575/AC>A/−), COL1A1 (17:48271806/AC>A/−), COL1A1 (17:48275531/AG>A/−), FN1 (2:216288075/GC>G/−) 
2. KEGG: Natural killer cell–mediated cytotoxicity 77% LCK (1:32739904/GC>G/−), PIK3R1 (5:67522514/A>G/+), PIK3CD (1:9775549/TC>T/−), PIK3CD (1:9775564/TG>T/+), ITGB2 (21:46309974/CG>C/−), ITGB2 (21:46323312/G>C/+) 
3. PID: αVβ3 integrin pathway 77% VEGFR2 (4:55971036/G>C/+), PIK3R1 (5:67522514/A>G/+), GPR124 (8:37693120/TC>T/−), IGF1R (15:99459983/TG>T/−), IGF1R (15:99478596/GA>G/+), ITGB3 (17:45367575/AC>A/−), ITGB3 (17:45367575/AC>A/−), COL1A1 (17:48271806/AC>A/−), COL1A1 (17:48275531/AG>A/−), FN1 (2:216288075/GC>G/−), TGFBR2 (3:30715604/C>CG/+) 
4. PID: Caspase pathway 77% MAP3K1 (5:56179436/G>C/−), NUMA1 (11:71726363/AG>A/−), SREBF1 (17:17720826/TG>T/−) 
5. PID: SHP2 pathway 77% VEGFR2 (4:55971036/G>C/+), LCK (1:32739904/GC>G/−), PIK3R1 (5:67522514/A>G/+), PDGFRB (5:149506102/AG>A/−), PDGFRB (5:149515110/GC>G/−), BDNF (11:27722583/TG>T/−), IGF1R (15:99459983/TG>T/−), IGF1R (15:99478596/GA>G/+) 
6. REACTOME: Cell surface interactions at the vascular wall 77% LCK (1:32739904/GC>G/−), PIK3R1 (5:67522514/A>G/+), ITGB3 (17:45367575/AC>A/−), ITGB3 (17:45367575/AC>A/−), COL1A1 (17:48271806/AC>A/−), COL1A1 (17:48275531/AG>A/−), ITGB2 (21:46309974/CG>C/−), ITGB2 (21:46323312/G>C/+), FN1 (2:216288075/GC>G/−) 
7. REACTOME: PPARα activates gene expression 77% CD36 (7:80299307/G>GT/+), NCOA2 (8:71069295/GC>G/−), CREBBP (16:3842089/TG>T/−), SREBF1 (17:17720826/TG>T/−), ANGPTL4 (19:8434086/A>AC/+) 
8. PID: αMβ2 integrin pathway 82% LCK (1:32739904/GC>G/−), ITGB2 (21:46309974/CG>C/−), ITGB2 (21:46323312/G>C/+) 
9. PID: Integrin 1 pathway 82% ITGA10 (1:145527997/AG>A/−), ITGA10 (1:145534180/A>AT/+), COL1A1 (17:48271806/AC>A/−), COL1A1 (17:48275531/AG>A/−), FN1 (2:216288075/GC>G/−) 
10. REACTOME: Metabolism of amino acids and derivatives 86% GLS2 (12:56868601/TC>T/−), MTR (1:237025593/GA>G/−) 
Union of above pathways 95% Union of above components 

NOTE: Of the variants found to be associated with vaccine response among training set individuals (P < 0.005), those within genes that participate in the pathways above were upweighted in the prediction model. The variants' chromosomal position, reference allele, alternative allele, and direction of association with vaccine response are shown in parentheses. Responses of the 22 test set individuals in cross-validation were inferred on the basis of whether their prediction scores exceed the cutoffs displayed in Supplementary Fig. S1 and Fig. 1. Corresponding prediction accuracies are also presented.

Patient characteristics of DC vaccine responders and nonresponders are presented in Table 1. Of the 11 patients who had immune responses, eight patients displayed greater IFNγ secretion, five patients displayed greater T-cell proliferation, and two patients displayed both. None of the measured attributes was associated with vaccine response. The Fong clinical risk score (3), a popular prognostic tool for patients with colorectal cancer undergoing liver metastasis resection (constructed from indicators for preoperative carcinoembryonic antigen blood level > 200 ng/mL, size of largest metastasis ≥ 5 cm, >1 metastasis, lymph node–positive primary tumor, and < 12 months between primary tumor resection and liver metastasis diagnosis), was not useful for vaccine response inference. Recruitment of cytotoxic and memory T cells to metastases was significantly elevated relative to surrounding liver abundances (P < 0.001). However, vaccine response among patients with metastatic colorectal cancer did not correlate with these tumor T-cell densities. The mutation burden within targeted sequencing regions of tumor samples used to form the DC vaccine also did not differ between vaccine responders and nonresponders. Nevertheless, sequencing of the metastatic tumors revealed other important insights.

Genes identified to be associated with DC vaccine response by the CMC method demonstrate statistical enrichment of pathways that primarily involve the PI3K/Akt/mTOR signaling axis (pathway Nos. 1, 2, 4, and 7 in Table 2). In brief, PI3K is induced by extracellular signals through receptor tyrosine kinases to phosphorylate and activate Akt, which then promotes mTOR to localize to the nucleus and alter gene transcription (35). A diversity of transcription repertoire can then be affected, influencing many mechanisms such as cell survival, proliferation, motility, and metabolism. These also intersect with functions of the other highlighted transcription factor pathways (pathway Nos. 5 and 6 in Table 2). Although it is intriguing that disturbance to B-lymphocyte receptor signaling is implicated as well (pathway No. 3 in Table 2), this finding may be a statistical artifact due to the pathway's considerable overlap with the actions of Akt and mTOR, including another shared downstream effector of Akt, tuberous sclerosis complex 2 (TSC2; ref. 35). The antitumor response provoked by antigen presentation has been characterized to be predominantly T-cell–mediated, whereas the roles of B cells are still being clarified (16).

Table 3 presents the pathways that optimally guide variant weights for prediction of DC vaccine response using Eq. (A). Prediction scores of the 21 training set individuals and leave-one-out test set individual across all rounds of cross-validation for these top pathways are plotted in Supplementary. S1. Not surprisingly, training set scores cluster well into distinguishable groups of vaccine responders and nonresponders, as their differences in allele frequencies were used to determine standardized variant association effects tk (P < 0.005) in Eq. (A). Among test set individuals, applying equal weights to associated variants correctly predicted 73% of vaccine responses (16 of 22). Differential weighting guided by top pathways increased accuracy up to 86% (19 of 22). In the absence of restrictions for pathway definitions, placing greater weight on variants within genes that comprise any top pathway further improved accuracy to 95% (21 of 22; Fig. 1); the only misclassified DC vaccine response was that of Individual 10. Even so, the corresponding prediction score became more positive with pathway-based variant weighting compared with equal weighting, which indicates adjustment in the correct direction as Individual 10 was a vaccine responder.

Figure 1.

Pathway-guided vaccine response prediction. Response prediction scores for 22 rounds of leave-one-out cross-validation are presented. The test set individual in each round is denoted by a black asterisk, whereas training set responders and nonresponders are denoted by blue dots and red dots, respectively. Test set individuals with scores above/below the dashed line were predicted to be vaccine responders/nonresponders. Of the variants found to be associated with vaccine response among training set individuals (P < 0.005), those within genes that are listed in Table 3 were upweighted in the prediction model.

Figure 1.

Pathway-guided vaccine response prediction. Response prediction scores for 22 rounds of leave-one-out cross-validation are presented. The test set individual in each round is denoted by a black asterisk, whereas training set responders and nonresponders are denoted by blue dots and red dots, respectively. Test set individuals with scores above/below the dashed line were predicted to be vaccine responders/nonresponders. Of the variants found to be associated with vaccine response among training set individuals (P < 0.005), those within genes that are listed in Table 3 were upweighted in the prediction model.

Close modal

There is a conspicuous theme underlying upweighted variants found to improve the accuracy of vaccine response prediction (Table 3). They tend to reside within genes that encode plasma membrane–related proteins. Many are proteins that interact with the extracellular matrix (integrins, collagens, fibronectin, and cluster of differentiation; ref. 36). Some are receptor tyrosine kinases that relay signals from outside of the cell to inside (VEGF receptor, platelet-derived growth factor receptor, TGFβ receptor, and IGF receptor). Other proteins are downstream effectors (PI3K and mitogen-activated protein kinases) of cascades initiated by the aforementioned classes of transmembrane proteins. Two important contributors to prediction using Eq. (A) and distinct outliers to this trend are variants within genes for glutaminase (GLS2) and methionine synthase (MTR). Normally, enzymes involved in amino acid metabolism (pathway No. 10 in Table 3), GLS2 and MTR are suspected to promote tumorigenesis when their underexpression or impaired function compromises regulation of oxidative stress (36, 37). Furthermore, the majority of association directions across highlighted variants is negative (Table 3 and asymmetry of prediction scores with respect to zero seen in Fig. 1). In other words, response to adjuvant DC vaccination is more likely to be observed in a patient with colorectal cancer who has liver metastases that carry the reference genome allele instead of the alternative allele, for most somatic mutations with association P < 0.005 identified by our tumor sequencing.

Through histologic enumeration of T-lymphocyte subpopulations, targeted gene sequencing of colorectal cancer liver metastases, and subsequent pathway-based genomic analyses, we explored the potential biologic mechanisms that confer greater likelihood of response to an adjuvant DC vaccine among patients with colorectal cancer following metastasis resection. Identification of biomarkers that predict response to immunotherapies is critical for optimizing their use in treating patients (38). Previous investigations have shown that cytotoxic and memory T-cell infiltration of primary and metastatic colorectal cancer tumors is positively associated with patient survival after surgical resection (18, 19). Conversely, depletion of regulatory T cells enhances the tumor antigen–specific immune response (39). However, in the present study, none of these microenvironment T-cell parameters that characterize the tumor interior, periphery, and surrounding of colorectal cancer liver metastases trended with DC vaccine response or survival.

Our pathway analyses implicate signaling by PI3K/Akt/mTOR and a variety of plasma membrane–related proteins as factors that may influence vaccine response among patients with metastatic colorectal cancer. Although these inferences seemingly differ on the basis of whether we performed multiple-variant joint association testing (the CMC method) or single-variant association testing, respectively, the findings are not contradictory but rather complementary. The CMC method is more statistically robust and conservative in identifying gene associations, but the generated results suffer from absence of association direction. And while single-variant association testing is more liable to identify false positives, it is able to produce association magnitudes and directions that can be directly used in prediction modeling. Biologically, the PI3K/Akt/mTOR axis (Table 2) is in fact a convergence point for upstream signaling from integrins, receptor tyrosine kinases, and apoptosis regulators Shp2 and the caspase family (Table 3; refs. 35, 40).

These cellular mechanisms can plausibly account for tumor attributes that promote DC vaccine resistance and poorer survival. First, impaired sensitivity to molecular cues for cell death maintains tumor survival (pathway Nos. 4, 5, and 10 in Table 3). Second, tumor surface proteins interact with the extracellular matrix to repurpose the microenvironment for optimal growth (pathway Nos. 1, 3, 6, 8, and 9 in Table 3). Reciprocal signaling from the extracellular matrix to transmembrane proteins that is improperly relayed to the rest of the cell can stimulate unwarranted growth as well (10, 41). Specifically, integrins on the cell membrane regulate migration, invasion, and proliferation, especially through endothelium interactions during metastasis and tumor-initiated angiogenesis (pathway No. 7 in Table 3; refs. 40, 42). Integrin αVβ3 expression in colorectal cancer liver metastases (pathway No. 3 in Table 3) has been shown to be nearly double the level in nonmetastatic primary colorectal cancer tumors (43). Tumors can also hijack the vascular migration apparatus of innate immune cells (pathway No. 2 in Table 3) by inducing them to release membrane vesicles that contain integrin αMβ2 (pathway No. 8 in Table 3) for fusion with tumor cells, ultimately facilitating metastasis (44). Moreover, integrin αMβ2 is involved in monocyte development and differentiation (45). Substantial loss of integrin αMβ2 from monocytes may compromise their ability to secrete IFNγ or prime T lymphocytes following antigen presentation, the principal antitumor action of DC vaccines.

Third, the directions of variant association effects in Table 3 offer additional clues. As mentioned earlier, the abundance of negative effects suggest that collective loss of function among some pathway components leads to pathway dysregulation and may give rise to a more aggressive cancer phenotype. Every other variant with a positive effect is contained within a gene that encodes a plasma membrane–related or secreted protein. It has been shown that although an intracellular tumor-specific protein can trigger both cell-mediated and humoral immune responses, no appreciable tumor eradication is observed, likely a consequence of the protein's intracellular expression (46). In contrast, considerable exposed changes in peptide sequence would be expected for membrane proteins linked to the vaccine response-predisposing variants in Table 3; most are frameshift indels. Even the remaining variants are still missense mutations that cause amino acid substitutions. Despite their potential deleterious effects on pathway functions, the positive associations between these variants and DC vaccine response may be due to their gene products' cell surface immunogenicity, especially in the face of proactive tumor efforts to hide foreign antigens through underexpressing major histocompability complex class I (MHC-I) molecules or displaying MHC-I surrogates (47).

In constructing an accurate prediction model for DC vaccine response among patients with metastatic colorectal cancer following surgery (95% correct, 21 of 22 patients), we implicated several mechanisms of cancer proliferation and immune system evasion that are likely to impact immunotherapy success in this disease context. A weakness of our study is its small sample size. Substantially larger sample sizes were used to identify the survival T lymphocyte relationships reported in previous studies (18, 19) that could not be replicated here for DC vaccine response. Having more than 22 patients with colorectal cancer with liver metastases can allow the derivation of stronger and more reliable variant association effects. It would be interesting to assess whether the prediction score cutoff shown in Fig. 1 can maintain the current prediction accuracy when applied to somatic genotype data from an independent cohort of patients.

Given the discovery nature of this study, targeted gene sequencing was performed in favor of whole-genome and whole-exome sequencing. With the latter two approaches in future studies, new pathway suspicions may be raised. Furthermore, whole-genome sequencing would be able to reveal outliers in total mutation burden which limited gene panel sequencing cannot (Table 1). Perhaps uncommon patients with metastatic colorectal cancer with orders of magnitude more tumor mutations due to deficient MMR (21) will have greater tendency to benefit from DC vaccination, as they do for immune checkpoint inhibition (20). Interestingly, this within-cancer phenomenon has been shown to be untrue for DC vaccination across cancers, as clinical benefit relative to placebo is lower among patients with melanoma patients (more mutations) than patients with glioma and renal cell carcinoma (fewer mutations; ref. 48). Another important difference compared with checkpoint inhibitors is that no toxicities were noted (17), so it is conceivable that even higher doses of DC vaccine may be indicated. Sequenced variants were also called relative to the publically available hg19 reference genome instead of paired normal colorectal tissue genomes. Although we removed germline variants on the basis of annotations in the NCBI database (28), we may have still captured noise signals that are not relevant to colorectal cancer given their undiscerned presence in normal tissue.

In addition to more comprehensive bioinformatics studies, companion experimental pursuits are warranted. Our genomic analyses serve to not only construct a meaningful DC vaccine response prediction model but also suggest novel directions for molecular investigations that may lead to an increase in the response prevalence or absolute efficacy of this immunotherapy. If metastatic colorectal cancer resistance to DC vaccination is truly influenced by the display of tumor surface neoantigens and aberrant transmembrane signaling through integrins and the PI3K/Akt/mTOR axis, combination immunotherapy with pharmacologic therapy may synergistically enhance tumor-specific immune responses and also lower the required dose, plus corresponding adverse effects, of each regimen. Among patients with colorectal cancer, PI3K inhibitors and integrin inhibitors have already been separately shown in clinical trials to be well-tolerated anticancer agents, in particular with superior effects for metastatic disease (49, 50). Findings in the present study merit further work to evaluate the potential benefits of DC vaccine co-administration.

No potential conflicts of interest were disclosed.

Conception and design: D.C. Qian, R.J. Barth Jr.

Development of methodology: D.C. Qian, C.I. Amos, R.J. Barth Jr.

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): A.A. Suriawinata, R.J. Barth Jr.

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): D.C. Qian, X. Xiao, J. Byun, S. Her, C.I. Amos, R.J. Barth Jr.

Writing, review, and/or revision of the manuscript: D.C. Qian, A.A. Suriawinata, S. Her, C.I. Amos, R.J. Barth Jr.

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): C.I. Amos, R.J. Barth Jr.

Study supervision: R.J. Barth Jr.

We would like to thank Heidi Trask and Dr. Craig Tomlinson in the Genomics & Molecular Biology Shared Resource at Dartmouth for sequencing the tumor samples.

This research was financially supported by the NIH (grants P30CA0123108 and P20GM103534) and a generous contribution from Ted and Rae Bachelder to R.J. Barth Jr.

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

1.
Stewart
BW
,
Wild
CP
.
World cancer report. Lyon, France: International Agency for Research on Cancer; 2014
.
2.
Jemal
A
,
Siegel
R
,
Ward
E
,
Hao
Y
,
Xu
J
,
Thun
MJ
. 
Cancer statistics, 2009
.
CA Cancer J Clin
2009
;
59
:
225
49
.
3.
Fong
Y
,
Fortner
J
,
Sun
RL
,
Brennan
MF
,
Blumgart
LH
. 
Clinical score for predicting recurrence after hepatic resection for metastatic colorectal cancer: analysis of 1001 consecutive cases
.
Ann Surg
1999
;
230
:
309
18
.
4.
Nordlinger
B
,
Guiguet
M
,
Vaillant
JC
,
Balladur
P
,
Boudjema
K
,
Bachellier
P
, et al
Surgical resection of colorectal carcinoma metastases to the liver. A prognostic scoring system to improve case selection, based on 1568 patients
.
Cancer
1996
;
77
:
1254
62
.
5.
Portier
G
,
Elias
D
,
Bouche
O
,
Rougier
P
,
Bosset
JF
,
Saric
J
, et al
Multicenter randomized trial of adjuvant fluorouracil and folinic acid compared with surgery alone after resection of colorectal liver metastases: FFCD ACHBTH AURC 9002 trial
.
J Clin Oncol
2006
;
24
:
4976
82
.
6.
Nordlinger
B
,
Sorbye
H
,
Glimelius
B
,
Poston
GJ
,
Schlag
PM
,
Rougier
P
, et al
Perioperative chemotherapy with FOLFOX4 and surgery versus surgery alone for resectable liver metastases from colorectal cancer (EORTC Intergroup trial 40983): a randomised controlled trial
.
Lancet
2008
;
371
:
1007
16
.
7.
Markman
JL
,
Shiao
SL
. 
Impact of the immune system and immunotherapy in colorectal cancer
.
J Gastrointest Oncol
2015
;
6
:
208
23
.
8.
Harris
JE
,
Ryan
L
,
Hoover
HC
 Jr
,
Stuart
RK
,
Oken
MM
,
Benson
AB
 III
, et al
Adjuvant active specific immunotherapy for stage II and III colon cancer with an autologous tumor cell vaccine: Eastern Cooperative Oncology Group Study E5283
.
J Clin Oncol
2000
;
18
:
148
57
.
9.
Bilusic
M
,
Heery
CR
,
Arlen
PM
,
Rauckhorst
M
,
Apelian
D
,
Tsang
KY
, et al
Phase I trial of a recombinant yeast-CEA vaccine (GI-6207) in adults with metastatic CEA-expressing carcinoma
.
Cancer Immunol Immunother
2014
;
63
:
225
34
.
10.
Kimura
T
,
McKolanis
JR
,
Dzubinski
LA
,
Islam
K
,
Potter
DM
,
Salazar
AM
, et al
MUC1 vaccine for individuals with advanced adenoma of the colon: a cancer immunoprevention feasibility study
.
Cancer Prev Res
2013
;
6
:
18
26
.
11.
Miyagi
Y
,
Imai
N
,
Sasatomi
T
,
Yamada
A
,
Mine
T
,
Katagiri
K
, et al
Induction of cellular immune responses to tumor cells and peptides in colorectal cancer patients by vaccination with SART3 peptides
.
Clin Cancer Res
2001
;
7
:
3950
62
.
12.
Moulton
HM
,
Yoshihara
PH
,
Mason
DH
,
Iversen
PL
,
Triozzi
PL
. 
Active specific immunotherapy with a beta-human chorionic gonadotropin peptide vaccine in patients with metastatic colorectal cancer: antibody response is associated with improved survival
.
Clin Cancer Res
2002
;
8
:
2044
51
.
13.
Idenoue
S
,
Hirohashi
Y
,
Torigoe
T
,
Sato
Y
,
Tamura
Y
,
Hariu
H
, et al
A potent immunogenic general cancer vaccine that targets survivin, an inhibitor of apoptosis proteins
.
Clin Cancer Res
2005
;
11
:
1474
82
.
14.
Speetjens
FM
,
Kuppen
PJ
,
Welters
MJ
,
Essahsah
F
,
Voet van den Brink
AM
,
Lantrua
MG
, et al
Induction of p53-specific immunity by a p53 synthetic long peptide vaccine in patients treated for metastatic colorectal cancer
.
Clin Cancer Res
2009
;
15
:
1086
95
.
15.
Yamada
A
,
Sasada
T
,
Noguchi
M
,
Itoh
K
. 
Next-generation peptide vaccines for advanced cancer
.
Cancer Sci
2013
;
104
:
15
21
.
16.
Palucka
K
,
Banchereau
J
. 
Dendritic-cell-based therapeutic cancer vaccines
.
Immunity
2013
;
39
:
38
48
.
17.
Barth
RJ
 Jr
,
Fisher
DA
,
Wallace
PK
,
Channon
JY
,
Noelle
RJ
,
Gui
J
, et al
A randomized trial of ex vivo CD40L activation of a dendritic cell vaccine in colorectal cancer patients: tumor-specific immune responses are associated with improved survival
.
Clin Cancer Res
2010
;
16
:
5548
56
.
18.
Nosho
K
,
Baba
Y
,
Tanaka
N
,
Shima
K
,
Hayashi
M
,
Meyerhardt
JA
, et al
Tumour-infiltrating T-cell subsets, molecular changes in colorectal cancer, and prognosis: cohort study and literature review
.
J Pathol
2010
;
222
:
350
66
.
19.
Katz
SC
,
Pillarisetty
V
,
Bamboat
ZM
,
Shia
J
,
Hedvat
C
,
Gonen
M
, et al
T cell infiltrate predicts long-term survival following resection of colorectal cancer liver metastases
.
Ann Surg Oncol
2009
;
16
:
2524
30
.
20.
Le
DT
,
Uram
JN
,
Wang
H
,
Bartlett
BR
,
Kemberling
H
,
Eyring
AD
, et al
PD-1 blockade in tumors with mismatch-repair deficiency
.
N Engl J Med
2015
;
372
:
2509
20
.
21.
Venderbosch
S
,
Nagtegaal
ID
,
Maughan
TS
,
Smith
CG
,
Cheadle
JP
,
Fisher
D
, et al
Mismatch repair status and BRAF mutation status in metastatic colorectal cancer patients: a pooled analysis of the CAIRO, CAIRO2, COIN, and FOCUS studies
.
Clin Cancer Res
2014
;
20
:
5322
30
.
22.
Czerkinsky
CC
,
Nilsson
LA
,
Nygren
H
,
Ouchterlony
O
,
Tarkowski
A
. 
A solid-phase enzyme-linked immunospot (ELISPOT) assay for enumeration of specific antibody-secreting cells
.
J Immunol Methods
1983
;
65
:
109
21
.
23.
Givan
AL
,
Fisher
JL
,
Waugh
M
,
Ernstoff
MS
,
Wallace
PK
. 
A flow cytometric method to estimate the precursor frequencies of cells proliferating in response to specific antigens
.
J Immunol Methods
1999
;
230
:
99
112
.
24.
Bercovici
N
,
Givan
AL
,
Waugh
MG
,
Fisher
JL
,
Vernel-Pauillac
F
,
Ernstoff
MS
, et al
Multiparameter precursor analysis of T-cell responses to antigen
.
J Immunol Methods
2003
;
276
:
5
17
.
25.
Cock
PJ
,
Fields
CJ
,
Goto
N
,
Heuer
ML
,
Rice
PM
. 
The Sanger FASTQ file format for sequences with quality scores, and the Solexa/Illumina FASTQ variants
.
Nucleic Acids Res
2010
;
38
:
1767
71
.
26.
McKenna
A
,
Hanna
M
,
Banks
E
,
Sivachenko
A
,
Cibulskis
K
,
Kernytsky
A
, et al
The Genome Analysis Toolkit: a MapReduce framework for analyzing next-generation DNA sequencing data
.
Genome Res
2010
;
20
:
1297
303
.
27.
Danecek
P
,
Auton
A
,
Abecasis
G
,
Albers
CA
,
Banks
E
,
DePristo
MA
, et al
The variant call format and VCFtools
.
Bioinformatics
2011
;
27
:
2156
8
.
28.
Sherry
ST
,
Ward
MH
,
Kholodov
M
,
Baker
J
,
Phan
L
,
Smigielski
EM
, et al
dbSNP: the NCBI database of genetic variation
.
Nucleic Acids Res
2001
;
29
:
308
11
.
29.
Dering
C
,
Hemmelmann
C
,
Pugh
E
,
Ziegler
A
. 
Statistical analysis of rare sequence variants: an overview of collapsing methods
.
Genet Epidemiol
2011
;
35
:
S12
7
.
30.
Li
B
,
Leal
SM
. 
Methods for detecting associations with rare variants for common diseases: application to analysis of sequence data
.
Am J Hum Genet
2008
;
83
:
311
21
.
31.
Subramanian
A
,
Tamayo
P
,
Mootha
VK
,
Mukherjee
S
,
Ebert
BL
,
Gillette
MA
, et al
Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles
.
Proc Natl Acad Sci
2005
;
102
:
15545
50
.
32.
Benjamini
Y
,
Yekutieli
D
. 
The control of the false discovery rate in multiple testing under dependency
.
Ann Statist
2001
;
29
:
1165
88
.
33.
Qian
DC
,
Byun
J
,
Han
Y
,
Greene
CS
,
Field
JK
,
Hung
RJ
, et al
Identification of shared and unique susceptibility pathways among cancers of the lung, breast, and prostate from genome-wide association studies and tissue-specific protein interactions
.
Hum Mol Genet
2015
;
24
:
7406
20
.
34.
Ross
OA
,
Soto-Ortolaza
AI
,
Heckman
MG
,
Aasly
JO
,
Abahuni
N
,
Annesi
G
, et al
Association of LRRK2 exonic variants with susceptibility to Parkinson's disease: a case–control study
.
Lancet Neurol
2011
;
10
:
898
908
.
35.
Dienstmann
R
,
Rodon
J
,
Serra
V
,
Tabernero
J
. 
Picking the point of inhibition: a comparative review of PI3K/AKT/mTOR pathway inhibitors
.
Mol Cancer Ther
2014
;
13
:
1021
31
.
36.
Suzuki
S
,
Tanaka
T
,
Poyurovsky
MV
,
Nagano
H
,
Mayama
T
,
Ohkubo
S
, et al
Phosphate-activated glutaminase (GLS2), a p53-inducible regulator of glutamine metabolism and reactive oxygen species
.
Proc Natl Acad Sci
2010
;
107
:
7461
6
.
37.
Miller
JW
,
Beresford
SA
,
Neuhouser
ML
,
Cheng
TY
,
Song
X
,
Brown
EC
, et al
Homocysteine, cysteine, and risk of incident colorectal cancer in the Women's Health Initiative observational cohort
.
Am J Clin Nutr
2013
;
97
:
827
34
.
38.
Osada
T
,
Nagaoka
K
,
Takahara
M
,
Yang
XY
,
Liu
CX
,
Guo
H
, et al
Precision cancer immunotherapy: optimizing dendritic cell-based strategies to induce tumor antigen-specific T-cell responses against individual patient tumors
.
J Immunother
2015
;
38
:
155
64
.
39.
Morse
MA
,
Hobeika
AC
,
Osada
T
,
Serra
D
,
Niedzwiecki
D
,
Lyerly
HK
, et al
Depletion of human regulatory T cells specifically enhances antigen-specific immune responses to cancer vaccines
.
Blood
2008
;
112
:
610
8
.
40.
Desgrosellier
JS
,
Cheresh
DA
. 
Integrins in cancer: biological implications and therapeutic opportunities
.
Nat Rev Cancer
2010
;
10
:
9
22
.
41.
Lu
P
,
Weaver
VM
,
Werb
Z
. 
The extracellular matrix: a dynamic niche in cancer progression
.
J Cell Biol
2012
;
196
:
395
406
.
42.
Bishop-Bailey
D
. 
PPARs and angiogenesis
.
Biochem Soc Trans
2011
;
39
:
1601
5
.
43.
Vonlaufen
A
,
Wiedle
G
,
Borisch
B
,
Birrer
S
,
Luder
P
,
Imhof
BA
. 
Integrin alpha(v)beta(3) expression in colon carcinoma correlates with survival
.
Mod Pathol
2001
;
14
:
1126
32
.
44.
Ma
J
,
Cai
W
,
Zhang
Y
,
Huang
C
,
Zhang
H
,
Liu
J
, et al
Innate immune cell-derived microparticles facilitate hepatocarcinoma metastasis by transferring integrin alpha(M)beta(2) to tumor cells
.
J Immunol
2013
;
191
:
3453
61
.
45.
Xue
ZH
,
Zhao
CQ
,
Chua
GL
,
Tan
SW
,
Tang
XY
,
Wong
SC
, et al
Integrin alphaMbeta2 clustering triggers phosphorylation and activation of protein kinase C delta that regulates transcription factor Foxp1 expression in monocytes
.
J Immunol
2010
;
184
:
3697
709
.
46.
Noguchi
T
,
Kato
T
,
Wang
L
,
Maeda
Y
,
Ikeda
H
,
Sato
E
, et al
Intracellular tumor-associated antigens represent effective targets for passive immunotherapy
.
Cancer Res
2012
;
72
:
1672
82
.
47.
Igney
FH
,
Krammer
PH
. 
Immune escape of tumors: apoptosis resistance and tumor counterattack
.
J Leukoc Biol
2002
;
71
:
907
20
.
48.
Anguille
S
,
Smits
EL
,
Lion
E
,
van Tendeloo
VF
,
Berneman
ZN
. 
Clinical use of dendritic cells for cancer therapy
.
Lancet Oncol
2014
;
15
:
e257
67
.
49.
Zhu
YF
,
Yu
BH
,
Li
DL
,
Ke
HL
,
Guo
XZ
,
Xiao
XY
. 
PI3K expression and PIK3CA mutations are related to colorectal cancer metastases
.
World J Gastroenterol
2012
;
18
:
3745
51
.
50.
Eskens
FA
,
Dumez
H
,
Hoekstra
R
,
Perschl
A
,
Brindley
C
,
Bottcher
S
, et al
Phase I and pharmacokinetic study of continuous twice weekly intravenous administration of Cilengitide (EMD 121974), a novel inhibitor of the integrins alphavbeta3 and alphavbeta5 in patients with advanced solid tumours
.
Eur J Cancer
2003
;
39
:
917
26
.

Supplementary data