Skip to main content
Erschienen in: BMC Cancer 1/2019

Open Access 01.12.2019 | Research article

The neoepitope landscape of breast cancer: implications for immunotherapy

Erschienen in: BMC Cancer | Ausgabe 1/2019

Abstract

Background

Cancer immunotherapy with immune checkpoint blockade (CKB) is now standard of care for multiple cancers. The clinical response to CKB is associated with T cell immunity targeting cancer-induced mutations that generate novel HLA-binding epitopes (neoepitopes).

Methods

Here, we developed a rapid bioinformatics pipeline and filtering strategy, EpitopeHunter, to identify and prioritize clinically relevant neoepitopes from the landscape of somatic mutations. We used the pipeline to determine the frequency of neoepitopes from the TCGA dataset of invasive breast cancers. We predicted HLA class I-binding neoepitopes for 870 breast cancer samples and filtered the neoepitopes based on tumor transcript abundance.

Results

We found that the total mutational burden (TMB) was highest for triple-negative breast cancer, TNBC, (median = 63 mutations, range: 2–765); followed by HER-2(+) (median = 39 mutations, range: 1–1206); and lowest for ER/PR(+)HER-2(−) (median = 32 mutations, range: 1–2860). 40% of the nonsynonymous mutations led to the generation of predicted neoepitopes. The neoepitope load (NEL) is highly correlated with the mutational burden (R2 = 0.86).

Conclusions

Only half (51%) of the predicted neoepitopes are expressed at the RNA level (FPKM≥2), indicating the importance of assessing whether neoepitopes are transcribed. However, of all patients, 93% have at least one expressed predicted neoepitope, indicating that most breast cancer patients have the potential for neo-epitope targeted immunotherapy.
Begleitmaterial
Additional file 4: Figure S2. Comparison of mutations called by the current protocol versus cBioportal (https://​www.​cbioportal.​org). The total number of mutations called by the current protocol (in blue) is comparable to the mutations called from cBioportal (in orange) across all three classifications of breast cancer subtype (PDF 425 kb)
Hinweise

Electronic supplementary material

The online version of this article (https://​doi.​org/​10.​1186/​s12885-019-5402-1) contains supplementary material, which is available to authorized users.
Pooja Narang and Meixuan Chen contributed equally to this work.
Abkürzungen
FPKM
Fragments per Kilobase per transcript per Million mapped reads
NEL
Neoepitope load
SNV
Single Nucleotide Variant
TCGA
The Cancer Genome Atlas
TMB
Tumor mutational burden
TNBC
Triple Negative Breast Cancer

Background

Immune evasion is a hallmark of cancer [1]. Immune checkpoint pathways contribute to cancer-induced immunosuppression, and immune checkpoint blockade (CKB) therapy has been successful in multiple cancers [2, 3]. Blockade of the immune regulatory molecules PD-1/PD-L1 and CTLA-4 reactivates T cell immunity and overcomes underlying T cell exhaustion [4, 5]. Sustained clinical responses and improved survival have been observed in the treatment arms of checkpoint blockade clinical trials, especially in tumors with high mutational burdens, such as melanoma and non-small cell lung cancer (NSCLC) [57]. Immune CKB is now FDA proved in melanoma [8], NSCLC [9], head and neck cancer [9] and bladder cancer [10], with ongoing clinical trials in multiple other cancer types.
Even though checkpoint blockade has achieved significant clinical success, it is not universally successful across patients [6, 10, 11]. There are two predictive biomarkers of response to CKB therapies. First, expression of PD-L1 in tumor samples correlates with checkpoint blockade response [11, 12]. Second, there is a strong association between the total mutational burden and clinical response [1315]. In colorectal cancer, the objective response rate is 40% in tumors with defects in mismatch repair, which have mutational burdens 10–100 times higher than tumors with functional mismatch repair [16].
Neoepitopes are targets for vaccine development [1719] and adoptive T cell therapy [20]. Neoepitopes occur, in part, as a result of unique somatic mutations in tumor cells, and are predicted to occur in tumors harboring more than 1 somatic mutation per Mb [21]. Mutation-specific CD8+ T cells have been identified in patients who respond to checkpoint blockade [15, 22]. The combination of checkpoint blockade with a patient-specific neoepitope vaccines are being developed [23].
Breast cancer typically harbors lower mutational loads than melanoma and NSCLC, averaging one mutation per Mb, but the mutational burden varies both within and across breast cancer subtypes [24]. The most frequently mutated genes are TP53 and PIK3CA [24]. Mutation-specific tumor-infiltrating T cells have been identified in metastases derived from breast cancer patients, and significant tumor responses have been observed after adoptive T cell transfer [25]. High levels of tumor infiltrating lymphocytes are correlated with improved clinical prognosis [26]. Two clinical trials of PD-1/PD-L1 inhibitors in advanced triple negative breast cancer (TNBC) have been reported, highlighting the clinical importance of immunotherapy in breast cancer. First, in the phase Ib KEYNOTE-012 trial of pembrolizumab, a 19% of overall response rate was observed, and, in the phase Ia trial of atezolizumab, a 24% overall response rate was observed [27, 28]. Identification of neoepitopes in breast cancer is essential for monitoring therapies and to generated personalized vaccines.
Next generation sequencing combined with high performance computing has resulted in the exploration and prediction of neoepitopes in multiple cancers [23, 2931]. The general approach includes HLA-typing and neoepitope prediction from whole exome sequence data [32]. There are also large databases for neoepitope prediction using a combination of somatic mutations and HLA-types; the oldest is SYFPEITHI [33], while the Immune Epitope Database, IEDB [34], is most widely used [32]. A few of the publically available pipelines used NetMHC [35] for epitope prediction like pVAC-seq [36] and INTEGRATE-neo [37]. Prediction methods have been applied to identify neoepitopes in melanoma, NSCLC, and chronic lymphocytic leukemia [14, 19, 23]. In a study of murine melanoma cells, B16F10, 50 validated mutations out of 962 non-synonymous point mutations were tested in vivo, and one-third elicited an immunogenic response [29]. A meta-analysis focused on neoepitopes generated exclusively by missense mutations reported between 1 to 147 immunogenic mutations per patient across 181 patients with different cancer types [38]. However, the neoepitope landscape of breast cancer has not yet been fully explored.
In this study, we evaluate the landscape of neoepitope burden across breast cancers included in The Cancer Genome Atlas (TCGA) [39]. We develop an efficient and publicly available prediction pipeline, EpitopeHunter (Fig. 1), to identify and prioritize clinically relevant mutations from the landscape of somatic mutations from tumor and normal exome data, patient-specific HLA-types, and neoepitope expression. Our prediction strategy employs the IEDB [34] binding affinity prediction algorithm, which dynamically selects the best possible method based on the predictive performance of several prediction methods [4043], for a given MHC molecule. Other publicly available pipelines [36, 37], on the other hand, use only one of the binding affinity prediction method for MHC molecules. We applied our pipeline to 870 breast cancer samples from TCGA [39]. Both somatic mutational burden and neoepitope load varies widely across tumors in breast cancer sub-types, and are highly correlated. We find that half of the predicted neoepitopes are expressed at the RNA level (FPKM≥2). Despite this, 93.5% of the patients have at least one expressed neoepitope that may serve as a candidate for targeted immunotherapy.

Methods

Breast cancer samples

The controlled access sequence data from The Cancer Genome Atlas (TCGA) was obtained for all submitted breast cancer samples via the genomics data commons (GDC) data portal with dbGap approval to Dr. Wilson Sayres (https://​portal.​gdc.​cancer.​gov; dbGap approval #46688).

Calling cancer specific variants

Tumor and germline whole exome sequencing BAM files were obtained for each patient, aligned to the GRCh37 human reference genome. We used this version of the genome to compare with the TCGA RNAseq calling and annotation available at the time, for which all samples were aligned to hg19/GRCh37. Samtools version 1.3.1 [44] was used to convert the aligned bam files to the pileup format. High confidence SNPs and indels were retrieved using VarScan2 version 2.3.9 [45] with minimum coverage of 10, minimum variant allele frequency of 0.08 and somatic p value of 0.05. Additionally, we removed somatic mutations from the high confidence SNPs that fell within 1 bp of an indel position, which are likely false positives due to alignment errors.

Variant annotation and neoepitope generation

We annotated mutations identified in the somatic variant calling using the Variant Effect Predictor tool [46] with ensemble transcripts annotated for the hg19 reference genome. For each non-synonymous amino acid, we generated all possible peptides including the mutated amino acid at every position in a sequence with total lengths of 8, 9, 10, 11 amino acids (called –mers) using the utility generate fasta from pvacseq [36]. That is, all possible 8-mers where the mutated amino acid is at the first position, then the second, third, and so on in a sliding window fashion. We also extracted the corresponding non-mutated reference sequence for each potential neoepitope. Thus, for every mutated amino acid we generated 38 possible neoepitopes.

HLA typing for each patient

We used POLYSOLVER (POLYmorphic loci reSOLVER) [47] to infer the HLA type of each patient using the germline whole exome sequencing data. This method employs a Bayesian classifier and selects and aligns putative HLA reads to an imputed library of full-length HLA alleles. We analyzed three major MHC class I genes (HLA-A, −B, −C) for HLA typing.

Predicting class I binding epitopes

To find neoepitopes predicted to bind to the patient-specific HLA alleles, we used the consensus prediction method from the Immune Epitope Database (IEDB) [34]. We began by matching the 4-dight HLA type of the patient to the HLA alleles in the IEDB database. If the matching HLA type of the patient did not exist in the current IEDB list, we identified the closest allele by keeping the first two digits same and searching for the best available match for the third and fourth digit. For each combination of HLA allele and peptides for each nonsynonymous amino acid (using those generated as 8, 9, 10, and 11-mers above), Epitopehunter selects the epitopes with lowest IEDB score. Thus, for each allele and mutant amino acid combination, it retains only one epitope. To get all high affinity binding neoepitopes with the patient-specific MHC class I molecules, we filtered epitopes with a binding affinity less than or equal to 500 nM, and for the current study, we call these potential binding neoepitopes.

RNAseq expression filtering

We obtained the gene level FPKM (Fragments Per Kilobase of transcript per Million mapped reads) values from the GDC portal for tumor samples with matched transcriptome data (https://​portal.​gdc.​cancer.​gov). Neoantigens were selected if the gene in which the neo-antigen appears was expressed in that patient’s tumor. Based on our previous analysis of FPKM thresholds [48], a gene was considered to be expressed using a cut-off of FPKM≥2. We also evaluated a more stringent cut-off of FPKM≥5.

Statistical analysis

All statistical comparisons and correlations were performed using an unpaired t-test and variation among and between groups was calculated using ANOVA (GraphPad Prism 6). The clinical data were described by the percentage, Kaplan-Meier method for calculation of survival, Log-Rank method for the univariate factor analysis (GraphPad Prism 6). P-values≤0.05 was considered significant. Significance testing of data from the three subtypes was performed using Wilcox rank sum test with *** P < 0.001 ** P < 0.01.

Molecular subtyping of breast cancer samples

To assign samples to molecular subtypes of breast cancer, we used the clinical calls of biomarkers using immunohistochemistry (IHC) status available for TCGA data (https://​cancergenome.​nih.​gov). We subdivided the clinical samples into three categories based on the immunohistochemical expression of the estrogen receptor (ER), progesterone receptor (PR) and the HER-2 receptor, into three types: 1) ER/PR(+)HER-2(−), those with ER and/or PR(+), HER-2(−); 2) HER-2(+), regardless of ER/PR status; and, 3) TNBC, triple negative breast cancer negative for all three ER, PR, and HER-2. The total numbers of TCGA samples and those used in this study for each subtype are reported in Additional file 1: Table S1.

Results

EpitopeHunter pipleline

The EpitopeHunter pipeline (Fig. 1) is broadly divided into the following steps: (1) variant calling and filtering; (2) variant annotation, peptide generation and generating a list of predicted neoepitopes; (3) HLA typing; and, (4) filtering epitope candidates based on IEDB binding score and epitope expression. The first step calls cancer specific mutations as described in the methods section. The user can use any cancer specific variant calling strategy and submit a variant file in vcf format (variant call format) for the next step. Next, we conduct variant annotation and generate a list of neoepitopes from nonsynonymous variants. This step will generate a total of 38 possible neoepitopes of lengths ranging from 8 to 11 amino acids for every non-synonymous cancer-specific variant. HLA typing can be performed using any of the HLA calling programs available for MHC class I; here we use Polysolver. Next, we predict which neoepitopes have a high likelihood to bind to the patient specific HLA alleles using IEDB and filter the high-affinity binding alleles on the basis of RNAseq expression (Methods). The final output file is a list of filtered/selected neoepitopes with binding scores for both WT and mutant epitopes.

Patient characteristics suggest a range of primary breast cancer samples

A total of 870 breast cancer patient samples from TCGA were included in our study; characteristics of the patients are summarized in Additional file 2: Table S2. The ER/PR(+)HER-2(−) was the most common subtype (72.4%) in this study, following by the HER-2(+) subtype (16.2%) and TNBC (11.4%). A higher proportion of patients with the TNBC subtype were under 50 years old (37%) compared to non-TNBC (25–26.5%). The majority of patients had early stage tumors; 83.4–88% of patients had a primary tumor size of T1-T2 across subtypes. Lymph node status was negative in less than half of patients with ER/PR(+)HER-2(−) subtype (44.6%) and patients with the HER-2(+) subtype (41.8%), but 68% of patients with the TNBC subtype had a negative lymph node status. Most of the patients included in our analysis were diagnosed with stage I/II breast cancer, with only 14–28.4% of patients were stage III-IV.

Confirmation of frequently mutated genes in subtypes of breast cancer

We identified the somatic mutational landscape across the three subtypes of breast cancer defined here (Additional file 3: Figure S1). For ER/PR(+)HER-2(−) cases, PIK3CA was mutated in 33% of the samples, TP53 was mutated in 13% of the samples, and TTN was mutated in 9% of the samples. In HER-2(+) cases, PIK3CA was mutated in 33% of the cases, TP53 was mutated in 24% of the cases, and TTN was mutated in 10% of cases. The most frequently mutated genes in TNBC cases are TP53 (present in 40% of samples), TTN (present in 17% of samples) and FAT3 (present in 10% of samples). Mutations called by the current protocol are also comparable with those called previously and available via cBioportal (Additional file 4: Figure S2). The current protocol appears to be more conservative, limiting potential non-synonymous mutations; we have a uniform variant calling protocol to call mutations with high confidence. The total number of nonsynonymous mutations are comparable to reports using previous methods in most cases (Additional file 4: Figure S2), and we chose to err on the side of being conservative with our mutation calling, to limit the number of false positive neoepitopes.

Mutational burden in subtypes of breast cancer

Mutational burden has a large range both within and between breast cancer subtypes (Fig. 2a). Overall, we observed the highest median nonsynonymous mutational burden among samples from patients with TNBC (median = 63 nonsynonymous mutations, range: 2–765); followed by HER-2(+) (median = 39 nonsynonymous mutations, range: 1–1206); and the lowest median mutational burden for samples from patients with ER/PR(+)HER-2(−) tumors (median = 32 nonsynonymous mutations, range: 1–2860). The mutational burden overlaps significantly across breast cancer subtypes, with extreme outliers on either end of the range (Fig. 2a).

Range of generated neoepitopes in subtypes of breast cancer

For each patient sample, we generated all possible neoepitopes from nonsynonymous mutations called in each tumor sample relative to the patient-specific germline (Methods). We selected the potential binding neoepitopes (neoepitope load) as those with ≤500 nM binding affinity to the set of patient-specific HLA class I alleles (Methods). Across tumors subtypes, ~ 37% of the nonsynonymous mutations in the ER/PR(+)HER-2(−) subset, ~ 41% of nonsynonymous mutations in the HER-2(+) subset and ~ 43% of the nonsynonymous mutations in the TNBC subset had a binding affinity ≤500 nM and were called as potential binding neoepitopes. Following the trend for the median mutational burden, the number of potential binding neoepitopes is highest for TNBC (median = 26, range:0–237), followed by HER-2(+) (median = 15, range:0–717); and is lowest for ER/PR(+)HER-2(−) (median = 10, range:0–864; Fig. 2b). The neoepitope load is highly correlated with the mutational burden in all the breast cancer samples considered together (R2 = 0.86, p < 0.001; Fig. 3a), or when broken up by subtypes (Additional file 5: Figure S3): ER/PR(+)HER-2(−) (R2 = 0.90, p < 0.001), HER-2(+) (R2 = 0.86, p < 0.001), and TNBC (R2 = 0.84, p < 0.001). The ratio of somatic mutational burden to predicted neoepitope load for the entire breast cancer dataset is 2.5:1 (Fig. 3a), similar to published values [49, 50].
We tested for the binding of all possible neoepitopes in sliding windows around the mutation of size 8 amino acids (8-mer), 9-mer, 10-mer, and 11-mer, with the patient-specific HLA (Methods), and found potential binding neoepitopes were largely skewed towards 9-mers. In particular, we find that the highest affinity binding epitope for any mutation is the 9-mer 57% of the time, in contrast to the 10-mers (33%), 11-mers (6%) and 8-mers (4%; Additional file 6: Figure S4).

Filtering neoepitopes on the basis of expression

We sought to identify neoepitopes that are expressed, and thus likely to elicit a response with immunotherapy. For this, we measured the expression of potential binding neoepitopes using available RNAseq data. We included neoepitopes as expressed if the gene in which the mutation occurred was expressed with FPKM≥2 (Rupp et al., 2017; Additional file 7: Figure S5). Additionally, we also tested a more stringent cutoff of FPKM≥5 (Fig. 2c). The number of expressed neoepitopes (cutoff of FPKM≥5; Fig. 2c) is highest for TNBC (median = 8 epitopes, range:0–82), followed by HER-2(+) (median = 4 epitopes, range:0–226), and lowest for ER/PR(+)HER-2(−) (median = 3 epitopes, range:0–230) subtype. As we expected, the neoepitope burden across all breast cancers is highly correlated with the number of expressed neoepitopes (R2 = 0.94, p < 0.001) (Fig. 3b), as well as each of the three subtypes individually (Additional file 8: Figure S6): TNBC (R2 = 0.93, p < 0.001), HER-2(+) (R2 = 0.99, p < 0.001), ER/PR(+)HER-2(−) (R2 = 0.95, p < 0.001).
Using a threshold of expression less than 5 FPKM, two thirds (~ 65%) of the neoepitopes are not expressed (TNBC = 65.2%, HER-2(+) = 64.4%, ER/PR(+)HER-2(−) = 65.5%; Fig. 4a), and are thus considered not expressed. For the three subtypes, approximately half of the expressed neoepitopes have an expression value less than 2 FPKM (TNBC = 50.3%, HER-2(+) = 47.2%, ER/PR(+)HER-2(−) = 49.4%; Additional file 9: Figure S7A). Despite most neoepitopes not being expressed, we find that 87% (709/815) of patients have at least one expressed neoepitope (Fig. 4b), with a threshold of expression less than 5 FPKM. Similarly 93.5% (762/815) of patients have at least one expressed neoepitope with FPKM≥2: TNBC (n = 90/94, 96%); HER-2(+) (n = 131/138, 95%); and ER/PR(+)HER-2(−) (n = 541/583, 93%; Additional file 9: Figure S7B).
We also looked at the number of neoepitopes predicted for each HLA class I allele in the TCGA samples. The total number of neoantigens for each allele, as well as for each breast cancer subtype, are shown in Additional file 10: Table S3. We notice that different alleles retrieve different proportion of neoepitopes in the breast cancer subtypes. Overall, patients with the ER/PR(+)HER-2(−) subtype has a higher proportion of HLA-A binding alleles, while patients with the HER-2(+) and the TNBC subtypes have a higher proportion of HLA-B binding alleles (Additional file 11: Figure S8).

Association of total mutation burden with clinical outcome

Mutation burden has been associated with clinical outcome in colorectal and ER(+) breast cancer patients [51, 52]. Here, we first compared the clinical outcome between high and low mutation burden groups. We defined the top quartile of mutation burden as high and the bottom quartile as low. No significant difference of disease-free survival was observed between high and low mutation burden groups in any subtype of breast cancer (Additional file 12: Figure S9). However, we noticed low mutation burden was associated with improved overall survival in the HER-2(+) subtype (p = 0.008; Additional file 12: Figure S9B). There were thirty-three patients in the low mutation burden group and thirty-seven subjects in the high mutation burden group; five patients died in the high mutation burden group, compared to none in the low mutation burden group.
We next compared the clinical outcome between high and low neoepitope load in the three subtypes of breast cancer. Neoepitope load was not associated with disease-free survival or overall survival (Additional file 13: Figure S10). In this study, we observed tumors that presented a higher neoepitope load (NEL) than the matched TMB (tumor mutation burden), indicative of neoepitopes that bind to multiple HLAs with high affinity and are expressed. There were more samples with NEL > TMB in TNBC (40%) than ER/PR(+)HER-2(−) (27%) and HER-2(+) (27%). We did not observe any association between high or low NEL/TMB ratio and prognosis (either disease-free survival or overall survival) across the breast cancer subtypes (Additional file 14: Figure S11).

Discussion

We developed a pipeline to rapidly predict neoepitopes, EpitopeHunter (Fig. 1), and applied it to the TCGA breast cancer exome and expression data to characterize the mutational landscape of somatic mutations and predict clinically-relevant neoepitopes. Our pipeline for neoepitope prediction uses the advanced method (IEDB) for epitope binding affinity prediction, which selects the best method out of a few methods [40], SMM [41], NetMHCpan [42], Comblib [43] for a given MHC molecule. Other publically available pipelines [36, 37], on the other hand, use only one method to predict binding affinity. Using efficient and parallel computational resources, the pipeline can be used in clinical settings to provide a list of prioritized neoantigens that can be tested in vitro to find potential vaccine candidates. We confirmed that, of the three subtypes of cancer, the TNBC subtype has both the highest median mutational burden and highest median neoepitope load. Further, by incorporating analysis of expression, we found that approximately half of predicted neoepitopes are not expressed at the RNA level, suggesting that RNA expression data is needed to predict neoepitope vaccine candidates, as the majority of neoepitopes predicted from exome sequencing alone will not result in effective tumor targeting. That said, as many as 93.5% of breast cancer patients in this study have at least one expressed neoepitope that could serve as a candidate for a vaccine. Our analysis finds that overall neoepitope load is not associated with either disease-free survival, nor overall survival (Additional file 13: Figure S10). In contrast, for HER-2(+) only, we find that overall survival is higher in patients with a lower mutational burden. We hypothesize that the lack of correlation between mutational burden or neoantigen load and survival may be due to several factors, including different treatments among the patients in each class, or potentially that the magnitude of mutation in a tumor isn’t sufficient to predict survival, but rather, that it may be important to use additional criterion to identify the types of mutations and neoantigens that are associated with survival outcomes. Thus, it may be more important to prioritize and identify any high-affinity and expressed neoepitopes for vaccine development, rather than being able to identify many candidates. We also find that 9-mer neoepitopes are most often predicted to be the highest affinity binders across all breast cancer subtypes, with most, but not all, high affinity binding neoepitopes being expressed at least 2 FPKM (Fragments per Kilobase per transcript per Million mapped reads). Rooney et al. [53], identified high mutational burden in breast cancers, that we recapitulate. We extend on this by performing our own variant calling, annotation and high affinity binding neoepitope prediction of cancer specific variants for the three breast cancer subtypes (ER/PR(+)HER-2(−), HER-2(+), and TNBC), and found that TNBC has higher mutational burden than other subtypes. Our findings suggest that most breast cancers, especially TNBC, have strong candidate neoepitopes that may serve as targets for personalized vaccines, and identify patients who may benefit from checkpoint treatment.
The pioneering study of neoepitope prediction of breast cancer only includes neoepitopes restricted to HLA0201 [23], and reported about 10 neoepitopes per sample. Another study, including 760 breast cancer samples from the TCGA dataset, reported the average mutational burden being 52.3 (1–393) and predicted the neoepitopes (9mers and 10mers) binding to each patient’s imputed HLA alleles (< 500 nM), reporting an average of 9.6 neoepitopes per sample (0–64) [53]. An average of 679 and 449 mutations per sample were reported in the study of melanoma and lung cancer, yielding a higher number of mean neoepitopes per sample, 63 and 46 per sample, respectively [21]. Generally, 50% of non-silent mutations were found with ≥1 predicted neo-epitope across cancer types [53].
Checkpoint blockade therapies targeting the immune microenvironment have demonstrated clinical activity in multiple tumors [6, 54, 55], including TNBC [28, 56]. The response rate ranges from 20 to 40% even in sensitive tumors, such as melanoma and NSCLC [57]. The response rate of checkpoint block might be improved when combined with a targeted neoantigen vaccine.
In this study, we only analyzed single nucleotide variant for HLA class I restricted neoepitope prediction. Giannakis et al. has reported that frameshifts generate larger proportion of neoantigens than SNVs [58], and more neoantigen specific CD + 4 T cells have been identified after immunotherapy [19, 59], which indicates the broad potential targets in breast cancer. In the future, we can extend EpitopeHunter to include insertions and deletions, in addition to point mutations.

Conclusions

Therapeutic tumor vaccination and CAR-T therapy have been tested in both preclinical and early phase clinical trials [19, 29, 6063]. Two recent publications in melanoma evaluated mutation-specific personalized vaccination [18, 19], and multiple other trials are ongoing. Sustained progression free intervals have been observed in a subset of patients with vaccine alone or combined with a PD-1 inhibitor [64]. However, neo-epitope specific vaccination remains a manufacturing challenge due to the limited overlap of neoepitopes between patients, and identification of the dominant neoepitopes within tumors with multiple potential targets. Our analysis of the neoepitope landscape across subtypes of breast cancer – notably that over 90% of patients have at least one expressed neoepitope, provides a strong rationale for the development of tumor vaccine trials for breast cancer.

Acknowledgements

We thank Dr. Barbara Pockaj and Dr. Sri Krishna for critical review of this manuscript.

Funding

This study was supported by the Breast Cancer Research Foundation to KSA, and by startup from the School of Life Sciences and the Biodesign Institute to MAW. The funders had no role in the design of the study nor in the collection, analysis, and interpretation of data, nor in writing the manuscript.

Availability of data and materials

The data that support the findings of this study were generated by The Cancer Genome Atlas and are available from Genomic Data Commons but restrictions apply to the availability of these data, which are controlled access and were used under license for the current study, and so are not publicly available. Data, excluding that available from Genomic Data Commons, such as neoepitope prediction are however available either with this manuscript or from the authors upon reasonable request. Code used to run EpitopeHunter is available at: https://​github.​com/​SexChrLab/​EpitopePipeline.
Not applicable.
Not applicable.

Competing interests

The authors declare that they have no competing interests.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.
Anhänge

Additional files

Additional file 4: Figure S2. Comparison of mutations called by the current protocol versus cBioportal (http://​www.​cbioportal.​org). The total number of mutations called by the current protocol (in blue) is comparable to the mutations called from cBioportal (in orange) across all three classifications of breast cancer subtype (PDF 425 kb)
Literatur
1.
Zurück zum Zitat Menon S, Shin S, Dy G. Advances in Cancer immunotherapy in solid tumors. Cancers (Basel). 2016;8:106.CrossRef Menon S, Shin S, Dy G. Advances in Cancer immunotherapy in solid tumors. Cancers (Basel). 2016;8:106.CrossRef
2.
Zurück zum Zitat Schachter J, et al. Pembrolizumab versus ipilimumab for advanced melanoma: final overall survival results of a multicentre, randomised, open-label phase 3 study (KEYNOTE-006). Lancet. 2017;390:1853–62.CrossRef Schachter J, et al. Pembrolizumab versus ipilimumab for advanced melanoma: final overall survival results of a multicentre, randomised, open-label phase 3 study (KEYNOTE-006). Lancet. 2017;390:1853–62.CrossRef
3.
Zurück zum Zitat Vansteenkiste J, et al. Prospects and progress of atezolizumab in non-small cell lung cancer. Expert Opin Biol Ther. 2017;17:781–9.CrossRef Vansteenkiste J, et al. Prospects and progress of atezolizumab in non-small cell lung cancer. Expert Opin Biol Ther. 2017;17:781–9.CrossRef
4.
Zurück zum Zitat Pardoll DM. The blockade of immune checkpoints in cancer immunotherapy. Nat Rev Cancer. 2012;12:252–64.CrossRef Pardoll DM. The blockade of immune checkpoints in cancer immunotherapy. Nat Rev Cancer. 2012;12:252–64.CrossRef
5.
Zurück zum Zitat Hassel JC. Ipilimumab plus nivolumab for advanced melanoma. Lancet Oncol. 2016;17:1471–2.CrossRef Hassel JC. Ipilimumab plus nivolumab for advanced melanoma. Lancet Oncol. 2016;17:1471–2.CrossRef
6.
Zurück zum Zitat Garon EB, et al. Pembrolizumab for the treatment of non-small-cell lung cancer. N Engl J Med. 2015;372:2018–28.CrossRef Garon EB, et al. Pembrolizumab for the treatment of non-small-cell lung cancer. N Engl J Med. 2015;372:2018–28.CrossRef
7.
Zurück zum Zitat Herbst RS, et al. Predictive correlates of response to the anti-PD-L1 antibody MPDL3280A in cancer patients. Nature. 2014;515:563–7.CrossRef Herbst RS, et al. Predictive correlates of response to the anti-PD-L1 antibody MPDL3280A in cancer patients. Nature. 2014;515:563–7.CrossRef
8.
Zurück zum Zitat Schadendorf D, et al. Pooled analysis of long-term survival data from phase II and phase III trials of Ipilimumab in Unresectable or metastatic melanoma. J Clin Oncol. 2015;33:1889–94.CrossRef Schadendorf D, et al. Pooled analysis of long-term survival data from phase II and phase III trials of Ipilimumab in Unresectable or metastatic melanoma. J Clin Oncol. 2015;33:1889–94.CrossRef
9.
Zurück zum Zitat Chow LQM, et al. Antitumor activity of Pembrolizumab in biomarker-unselected patients with recurrent and/or metastatic head and neck squamous cell carcinoma: results from the phase Ib KEYNOTE-012 expansion cohort. J Clin Oncol. 2016;34:3838–45.CrossRef Chow LQM, et al. Antitumor activity of Pembrolizumab in biomarker-unselected patients with recurrent and/or metastatic head and neck squamous cell carcinoma: results from the phase Ib KEYNOTE-012 expansion cohort. J Clin Oncol. 2016;34:3838–45.CrossRef
10.
Zurück zum Zitat Brahmer J, et al. Nivolumab versus docetaxel in advanced squamous-cell non-small-cell lung Cancer. N Engl J Med. 2015;373:123–35.CrossRef Brahmer J, et al. Nivolumab versus docetaxel in advanced squamous-cell non-small-cell lung Cancer. N Engl J Med. 2015;373:123–35.CrossRef
11.
Zurück zum Zitat Topalian SL, et al. Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N Engl J Med. 2012;366:2443–54.CrossRef Topalian SL, et al. Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N Engl J Med. 2012;366:2443–54.CrossRef
12.
Zurück zum Zitat Taube JM, et al. Association of PD-1, PD-1 ligands, and other features of the tumor immune microenvironment with response to anti-PD-1 therapy. Clin Cancer Res. 2014;20:5064–74.CrossRef Taube JM, et al. Association of PD-1, PD-1 ligands, and other features of the tumor immune microenvironment with response to anti-PD-1 therapy. Clin Cancer Res. 2014;20:5064–74.CrossRef
13.
Zurück zum Zitat Chan TA, Wolchok JD, Snyder A. Genetic basis for clinical response to CTLA-4 blockade in melanoma. N Engl J Med. 2015;373:1984.CrossRef Chan TA, Wolchok JD, Snyder A. Genetic basis for clinical response to CTLA-4 blockade in melanoma. N Engl J Med. 2015;373:1984.CrossRef
14.
Zurück zum Zitat Van Allen EM, et al. Genomic correlates of response to CTLA-4 blockade in metastatic melanoma. Science. 2015;350:207–11.CrossRef Van Allen EM, et al. Genomic correlates of response to CTLA-4 blockade in metastatic melanoma. Science. 2015;350:207–11.CrossRef
15.
Zurück zum Zitat Rizvi NA, et al. Cancer immunology. Mutational landscape determines sensitivity to PD-1 blockade in non-small cell lung cancer. Science. 2015;348:124–8.CrossRef Rizvi NA, et al. Cancer immunology. Mutational landscape determines sensitivity to PD-1 blockade in non-small cell lung cancer. Science. 2015;348:124–8.CrossRef
16.
Zurück zum Zitat Le DT, et al. PD-1 blockade in tumors with mismatch-repair deficiency. N Engl J Med. 2015;372:2509–20.CrossRef Le DT, et al. PD-1 blockade in tumors with mismatch-repair deficiency. N Engl J Med. 2015;372:2509–20.CrossRef
17.
Zurück zum Zitat Gilboa E. The makings of a tumor rejection antigen. Immunity. 1999;11:263–70.CrossRef Gilboa E. The makings of a tumor rejection antigen. Immunity. 1999;11:263–70.CrossRef
18.
Zurück zum Zitat Ott PA, et al. An immunogenic personal neoantigen vaccine for patients with melanoma. Nature. 2017;547:217–21.CrossRef Ott PA, et al. An immunogenic personal neoantigen vaccine for patients with melanoma. Nature. 2017;547:217–21.CrossRef
19.
Zurück zum Zitat Sahin U, et al. Personalized RNA mutanome vaccines mobilize poly-specific therapeutic immunity against cancer. Nature. 2017;547:222–6.CrossRef Sahin U, et al. Personalized RNA mutanome vaccines mobilize poly-specific therapeutic immunity against cancer. Nature. 2017;547:222–6.CrossRef
20.
Zurück zum Zitat Rosenberg SA, Restifo NP. Adoptive cell transfer as personalized immunotherapy for human cancer. Science. 2015;348:62–8.CrossRef Rosenberg SA, Restifo NP. Adoptive cell transfer as personalized immunotherapy for human cancer. Science. 2015;348:62–8.CrossRef
21.
Zurück zum Zitat Schumacher TN, Schreiber RD. Neoantigens in cancer immunotherapy. Science. 2015;348:69–74.CrossRef Schumacher TN, Schreiber RD. Neoantigens in cancer immunotherapy. Science. 2015;348:69–74.CrossRef
22.
Zurück zum Zitat Snyder A, et al. Genetic basis for clinical response to CTLA-4 blockade in melanoma. N Engl J Med. 2014;371:2189–99.CrossRef Snyder A, et al. Genetic basis for clinical response to CTLA-4 blockade in melanoma. N Engl J Med. 2014;371:2189–99.CrossRef
23.
Zurück zum Zitat Segal NH, et al. Epitope landscape in breast and colorectal cancer. Cancer Res. 2008;68:889–92.CrossRef Segal NH, et al. Epitope landscape in breast and colorectal cancer. Cancer Res. 2008;68:889–92.CrossRef
24.
Zurück zum Zitat Kandoth C, et al. Mutational landscape and significance across 12 major cancer types. Nature. 2013;502:333–9.CrossRef Kandoth C, et al. Mutational landscape and significance across 12 major cancer types. Nature. 2013;502:333–9.CrossRef
25.
Zurück zum Zitat Goff S, et al. Abstract P2–04-02: Recognition of autologous neoantigens by tumor infiltrating lymphocytes derived from breast cancer metastases. Cancer Res. 2017;77:P2–04-02-P2–04-02.CrossRef Goff S, et al. Abstract P2–04-02: Recognition of autologous neoantigens by tumor infiltrating lymphocytes derived from breast cancer metastases. Cancer Res. 2017;77:P2–04-02-P2–04-02.CrossRef
26.
Zurück zum Zitat Bense RD, et al. Relevance of tumor-infiltrating immune cell composition and functionality for disease outcome in breast Cancer. J Natl Cancer Inst. 2017;109. Bense RD, et al. Relevance of tumor-infiltrating immune cell composition and functionality for disease outcome in breast Cancer. J Natl Cancer Inst. 2017;109.
27.
Zurück zum Zitat Bedognetti D, Maccalli C, Bader SB, Marincola FM, Seliger B. Checkpoint inhibitors and their application in breast Cancer. Breast Care (Basel). 2016;11:108–15.CrossRef Bedognetti D, Maccalli C, Bader SB, Marincola FM, Seliger B. Checkpoint inhibitors and their application in breast Cancer. Breast Care (Basel). 2016;11:108–15.CrossRef
28.
Zurück zum Zitat Nanda R, et al. Pembrolizumab in patients with advanced triple-negative breast Cancer: phase Ib KEYNOTE-012 study. J Clin Oncol. 2016;34:2460–7.CrossRef Nanda R, et al. Pembrolizumab in patients with advanced triple-negative breast Cancer: phase Ib KEYNOTE-012 study. J Clin Oncol. 2016;34:2460–7.CrossRef
29.
Zurück zum Zitat Castle JC, et al. Exploiting the mutanome for tumor vaccination. Cancer Res. 2012;72:1081–91.CrossRef Castle JC, et al. Exploiting the mutanome for tumor vaccination. Cancer Res. 2012;72:1081–91.CrossRef
30.
Zurück zum Zitat Wick DA, et al. Surveillance of the tumor mutanome by T cells during progression from primary to recurrent ovarian cancer. Clin Cancer Res. 2014;20:1125–34.CrossRef Wick DA, et al. Surveillance of the tumor mutanome by T cells during progression from primary to recurrent ovarian cancer. Clin Cancer Res. 2014;20:1125–34.CrossRef
31.
Zurück zum Zitat Duan F, et al. Genomic and bioinformatic profiling of mutational neoepitopes reveals new rules to predict anticancer immunogenicity. J Exp Med. 2014;211:2231–48.CrossRef Duan F, et al. Genomic and bioinformatic profiling of mutational neoepitopes reveals new rules to predict anticancer immunogenicity. J Exp Med. 2014;211:2231–48.CrossRef
32.
Zurück zum Zitat Snyder A, Chan TA. Immunogenic peptide discovery in cancer genomes. Curr Opin Genet Dev. 2015;30:7–16.CrossRef Snyder A, Chan TA. Immunogenic peptide discovery in cancer genomes. Curr Opin Genet Dev. 2015;30:7–16.CrossRef
33.
Zurück zum Zitat Rammensee HG, Friede T. Stevanoviic, S. MHC ligands and peptide motifs: first listing. Immunogenetics. 1995;41:178–228.CrossRef Rammensee HG, Friede T. Stevanoviic, S. MHC ligands and peptide motifs: first listing. Immunogenetics. 1995;41:178–228.CrossRef
34.
Zurück zum Zitat Vita R, et al. The immune epitope database 2.0. Nucleic Acids Res. 2010;38:D854–62.CrossRef Vita R, et al. The immune epitope database 2.0. Nucleic Acids Res. 2010;38:D854–62.CrossRef
35.
Zurück zum Zitat Lundegaard C, et al. NetMHC-3.0: accurate web accessible predictions of human, mouse and monkey MHC class I affinities for peptides of length 8-11. Nucleic Acids Res. 2008;36:W509–12.CrossRef Lundegaard C, et al. NetMHC-3.0: accurate web accessible predictions of human, mouse and monkey MHC class I affinities for peptides of length 8-11. Nucleic Acids Res. 2008;36:W509–12.CrossRef
36.
Zurück zum Zitat Hundal J, et al. pVAC-Seq: a genome-guided in silico approach to identifying tumor neoantigens. Genome Med. 2016;8:11.CrossRef Hundal J, et al. pVAC-Seq: a genome-guided in silico approach to identifying tumor neoantigens. Genome Med. 2016;8:11.CrossRef
37.
Zurück zum Zitat Zhang J, Mardis ER, Maher CA INTEGRATE-n. a pipeline for personalized gene fusion neoantigen discovery. Bioinformatics. 2017;33:555–7. Zhang J, Mardis ER, Maher CA INTEGRATE-n. a pipeline for personalized gene fusion neoantigen discovery. Bioinformatics. 2017;33:555–7.
38.
Zurück zum Zitat Brown SD, et al. Neo-antigens predicted by tumor genome meta-analysis correlate with increased patient survival. Genome Res. 2014;24:743–50.CrossRef Brown SD, et al. Neo-antigens predicted by tumor genome meta-analysis correlate with increased patient survival. Genome Res. 2014;24:743–50.CrossRef
39.
Zurück zum Zitat Cancer Genome Atlas Research Network, et al. The Cancer genome atlas pan-Cancer analysis project. Nat Genet. 2013;45:1113–20.CrossRef Cancer Genome Atlas Research Network, et al. The Cancer genome atlas pan-Cancer analysis project. Nat Genet. 2013;45:1113–20.CrossRef
40.
Zurück zum Zitat Nielsen M, et al. Reliable prediction of T-cell epitopes using neural networks with novel sequence representations. Protein Sci. 2003;12:1007–17.CrossRef Nielsen M, et al. Reliable prediction of T-cell epitopes using neural networks with novel sequence representations. Protein Sci. 2003;12:1007–17.CrossRef
41.
Zurück zum Zitat Peters B, Sette A. Generating quantitative models describing the sequence specificity of biological processes with the stabilized matrix method. Bmc Bioinformatics. 2005;6:132.CrossRef Peters B, Sette A. Generating quantitative models describing the sequence specificity of biological processes with the stabilized matrix method. Bmc Bioinformatics. 2005;6:132.CrossRef
42.
Zurück zum Zitat Hoof I, et al. NetMHCpan, a method for MHC class I binding prediction beyond humans. Immunogenetics. 2009;61:1–13.CrossRef Hoof I, et al. NetMHCpan, a method for MHC class I binding prediction beyond humans. Immunogenetics. 2009;61:1–13.CrossRef
43.
Zurück zum Zitat Sidney J, et al. Quantitative peptide binding motifs for 19 human and mouse MHC class I molecules derived using positional scanning combinatorial peptide libraries. Immunome Res. 2008;4:2.CrossRef Sidney J, et al. Quantitative peptide binding motifs for 19 human and mouse MHC class I molecules derived using positional scanning combinatorial peptide libraries. Immunome Res. 2008;4:2.CrossRef
44.
Zurück zum Zitat Li H, et al. The sequence alignment/map format and SAMtools. Bioinformatics. 2009;25:2078–9.CrossRef Li H, et al. The sequence alignment/map format and SAMtools. Bioinformatics. 2009;25:2078–9.CrossRef
45.
Zurück zum Zitat Koboldt DC, et al. VarScan 2: somatic mutation and copy number alteration discovery in cancer by exome sequencing. Genome Res. 2012;22:568–76.CrossRef Koboldt DC, et al. VarScan 2: somatic mutation and copy number alteration discovery in cancer by exome sequencing. Genome Res. 2012;22:568–76.CrossRef
46.
Zurück zum Zitat McLaren W, et al. The Ensembl variant effect predictor. Genome Biol. 2016;17:122.CrossRef McLaren W, et al. The Ensembl variant effect predictor. Genome Biol. 2016;17:122.CrossRef
47.
Zurück zum Zitat Shukla SA, et al. Comprehensive analysis of cancer-associated somatic mutations in class I HLA genes. Nat Biotechnol. 2015;33:1152–8.CrossRef Shukla SA, et al. Comprehensive analysis of cancer-associated somatic mutations in class I HLA genes. Nat Biotechnol. 2015;33:1152–8.CrossRef
48.
Zurück zum Zitat Rupp SM, et al. Evolution of dosage compensation in Anolis carolinensis, a reptile with XX/XY chromosomal sex determination. Genome Biol Evol. 2017;9:231–40. Rupp SM, et al. Evolution of dosage compensation in Anolis carolinensis, a reptile with XX/XY chromosomal sex determination. Genome Biol Evol. 2017;9:231–40.
49.
Zurück zum Zitat Lauss M, et al. Mutational and putative neoantigen load predict clinical benefit of adoptive T cell therapy in melanoma. Nat Commun. 2017;8:1738.CrossRef Lauss M, et al. Mutational and putative neoantigen load predict clinical benefit of adoptive T cell therapy in melanoma. Nat Commun. 2017;8:1738.CrossRef
50.
Zurück zum Zitat Miller A, et al. High somatic mutation and neoantigen burden are correlated with decreased progression-free survival in multiple myeloma. Blood Cancer J. 2017;7:e612.CrossRef Miller A, et al. High somatic mutation and neoantigen burden are correlated with decreased progression-free survival in multiple myeloma. Blood Cancer J. 2017;7:e612.CrossRef
51.
Zurück zum Zitat Donehower LA, et al. MLH1-silenced and non-silenced subgroups of hypermutated colorectal carcinomas have distinct mutational landscapes. J Pathol. 2013;229:99–110.CrossRef Donehower LA, et al. MLH1-silenced and non-silenced subgroups of hypermutated colorectal carcinomas have distinct mutational landscapes. J Pathol. 2013;229:99–110.CrossRef
52.
Zurück zum Zitat Haricharan S, Bainbridge MN, Scheet P, Brown PH. Somatic mutation load of estrogen receptor-positive breast tumors predicts overall survival: an analysis of genome sequence data. Breast Cancer Res Treat. 2014;146:211–20.CrossRef Haricharan S, Bainbridge MN, Scheet P, Brown PH. Somatic mutation load of estrogen receptor-positive breast tumors predicts overall survival: an analysis of genome sequence data. Breast Cancer Res Treat. 2014;146:211–20.CrossRef
53.
Zurück zum Zitat Rooney MS, Shukla SA, Wu CJ, Getz G, Hacohen N. Molecular and genetic properties of tumors associated with local immune cytolytic activity. Cell. 2015;160:48–61.CrossRef Rooney MS, Shukla SA, Wu CJ, Getz G, Hacohen N. Molecular and genetic properties of tumors associated with local immune cytolytic activity. Cell. 2015;160:48–61.CrossRef
54.
Zurück zum Zitat Callahan MK, et al. Nivolumab plus Ipilimumab in patients with advanced melanoma: updated survival, response, and safety data in a phase I dose-escalation study. J Clin Oncol. 2018;36:391–8.CrossRef Callahan MK, et al. Nivolumab plus Ipilimumab in patients with advanced melanoma: updated survival, response, and safety data in a phase I dose-escalation study. J Clin Oncol. 2018;36:391–8.CrossRef
55.
Zurück zum Zitat Balar AV, et al. Atezolizumab as first-line treatment in cisplatin-ineligible patients with locally advanced and metastatic urothelial carcinoma: a single-arm, multicentre, phase 2 trial. Lancet. 2017;389:67–76.CrossRef Balar AV, et al. Atezolizumab as first-line treatment in cisplatin-ineligible patients with locally advanced and metastatic urothelial carcinoma: a single-arm, multicentre, phase 2 trial. Lancet. 2017;389:67–76.CrossRef
56.
Zurück zum Zitat Dirix LY, et al. Avelumab, an anti-PD-L1 antibody, in patients with locally advanced or metastatic breast cancer: a phase 1b JAVELIN solid tumor study. Breast Cancer Res Treat. 2018;167:671–86.CrossRef Dirix LY, et al. Avelumab, an anti-PD-L1 antibody, in patients with locally advanced or metastatic breast cancer: a phase 1b JAVELIN solid tumor study. Breast Cancer Res Treat. 2018;167:671–86.CrossRef
57.
Zurück zum Zitat Aldous AR, Dong JZ. Personalized neoantigen vaccines: a new approach to cancer immunotherapy. Bioorg Med Chem. 2018;26:2842–9.CrossRef Aldous AR, Dong JZ. Personalized neoantigen vaccines: a new approach to cancer immunotherapy. Bioorg Med Chem. 2018;26:2842–9.CrossRef
58.
Zurück zum Zitat Giannakis M, et al. Genomic correlates of immune-cell infiltrates in colorectal carcinoma. Cell Rep. 2016;17:1206.CrossRef Giannakis M, et al. Genomic correlates of immune-cell infiltrates in colorectal carcinoma. Cell Rep. 2016;17:1206.CrossRef
59.
Zurück zum Zitat Linnemann C, et al. High-throughput epitope discovery reveals frequent recognition of neo-antigens by CD4+ T cells in human melanoma. Nat Med. 2015;21:81–5.CrossRef Linnemann C, et al. High-throughput epitope discovery reveals frequent recognition of neo-antigens by CD4+ T cells in human melanoma. Nat Med. 2015;21:81–5.CrossRef
60.
Zurück zum Zitat Carreno BM, et al. Cancer immunotherapy. A dendritic cell vaccine increases the breadth and diversity of melanoma neoantigen-specific T cells. Science. 2015;348:803–8.CrossRef Carreno BM, et al. Cancer immunotherapy. A dendritic cell vaccine increases the breadth and diversity of melanoma neoantigen-specific T cells. Science. 2015;348:803–8.CrossRef
61.
Zurück zum Zitat Bozeman EN, He S, Shafizadeh Y, Selvaraj P. Therapeutic efficacy of PD-L1 blockade in a breast cancer model is enhanced by cellular vaccines expressing B7-1 and glycolipid-anchored IL-12. Hum Vaccin Immunother. 2016;12:421–30.CrossRef Bozeman EN, He S, Shafizadeh Y, Selvaraj P. Therapeutic efficacy of PD-L1 blockade in a breast cancer model is enhanced by cellular vaccines expressing B7-1 and glycolipid-anchored IL-12. Hum Vaccin Immunother. 2016;12:421–30.CrossRef
62.
Zurück zum Zitat Byrd TT, et al. TEM8/ANTXR1-specific CAR T cells as a targeted therapy for triple-negative breast Cancer. Cancer Res. 2018;78:489–500.CrossRef Byrd TT, et al. TEM8/ANTXR1-specific CAR T cells as a targeted therapy for triple-negative breast Cancer. Cancer Res. 2018;78:489–500.CrossRef
63.
Zurück zum Zitat Ernst B, Anderson KS. Immunotherapy for the treatment of breast cancer. Curr Oncol Rep. 2015;17:5.CrossRef Ernst B, Anderson KS. Immunotherapy for the treatment of breast cancer. Curr Oncol Rep. 2015;17:5.CrossRef
64.
Zurück zum Zitat Zacharakis N, et al. Immune recognition of somatic mutations leading to complete durable regression in metastatic breast cancer. Nat Med. 2018;24:724–30.CrossRef Zacharakis N, et al. Immune recognition of somatic mutations leading to complete durable regression in metastatic breast cancer. Nat Med. 2018;24:724–30.CrossRef
Metadaten
Titel
The neoepitope landscape of breast cancer: implications for immunotherapy
Publikationsdatum
01.12.2019
Erschienen in
BMC Cancer / Ausgabe 1/2019
Elektronische ISSN: 1471-2407
DOI
https://doi.org/10.1186/s12885-019-5402-1

Weitere Artikel der Ausgabe 1/2019

BMC Cancer 1/2019 Zur Ausgabe

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.