Skip to main content
Access keys NCBI Homepage MyNCBI Homepage Main Content Main Navigation
Curr Opin Genet Dev. Author manuscript; available in PMC 2019 Jul 25.
Published in final edited form as:
PMCID: PMC6657809
NIHMSID: NIHMS1039657
PMID: 25588790

Immunogenic peptide discovery in cancer genomes

Abstract

As immunotherapies to treat malignancy continue to diversify along with the tumor types amenable to treatment, it will become very important to predict which treatment is most likely to benefit a given patient. Tumor neoantigens, novel peptides resulting from somatic tumor mutations and recognized by the immune system as foreign, are likely to contribute significantly to the efficacy of immunotherapy. Multiple in silico methods have been developed to predict whether peptides, including tumor neoantigens, will be presented by the major histocompatibility complex (MHC) Class I or Class II, and interact with the T cell receptor (TCR). The methods for neoantigen prediction will be reviewed here, along with the most important examples of their use in the field of oncology.

Introduction: why prediction, why now?

When William B. Coley, the ‘father’ of immunotherapy, injected streptococcal organisms into patients with metastatic solid tumors in the 1890s, it was not known what aspects of ‘Coley’s Toxins,’ the patient and/or tumor, were responsible for tumor regression in a subset of patients with metastatic cancer. Nearly a century later, IL-2 was approved for the treatment of metastatic melanoma [1]. Since then, checkpoint blockade therapies and T cell therapies have proliferated, with promising results for both [2,3,4●●]. With this increase in immunotherapies, clinicians will need tools to predict which type of immunotherapy is most likely to benefit a specific patient.

A growing body of literature suggests that response to multiple types of immunotherapy results from the anti-tumor immune response against a critical neoantigen(s), non-self peptides resulting from exonic missense mutations (reviewed in [5●]). Cancer vaccines have historically used tumor-associated antigens which are over-expressed in tumors and have restricted tissue expression. However, these therapies require overcoming central and peripheral tolerance. In contrast, a tumor neoantigen would in theory not be limited by tolerance, with data both in preclinical models and humans supporting this idea [6●,7●]. It is important to note that not all effective immunotherapies target tumor neoantigens, but rather may target viral [8] or other antigens, as with chimeric antigen receptors [9,10] and therapeutic antibodies [11].

The major hurdle to selecting, improving and designing immunotherapies based on neoantigens lies in their accurate prediction, a challenging process in light of the complexities of the immune system. The major histocompatibility complex (MHC) molecules include two classes. MHC Class I (in humans, human leukocyte antigen, or HLA, Class I) molecules bind intracellular antigens of 8–11 amino acids in length and present them to cytotoxic CD8+ T cells. MHC Class II molecules bind extracellular antigens of 11–20 amino acids and present them to T helper CD4+ cells [12]. The HLA alleles are incredibly diverse, with greater than 6000 HLA Class I and Class II alleles described to date [13]. The number of potential peptides processed from a given pathogen is also vast, with a small proportion actually binding MHC Class I or II [14]. These facts make peptide prediction both important and challenging.

An immunogenic peptide fulfills at least two criteria: presentation by an MHC molecule and recognition by a T-cell receptor (Figure 1). In order to be presented by the MHC, a protein may be cleaved, and is typically presented by the antigen presentation machinery. Multiple computational algorithms exist for each step in this process. A fairly exhaustive list of prediction tools is provided (http://cancerimmunity.org/resources/webtools/ and tables in [1517]), and the bioinformatic aspects of many of these programs has been reviewed [17,18●,19●].

An external file that holds a picture, illustration, etc.
Object name is nihms-1039657-f0001.jpg

Summary of antigen presentation. A tumor cell (top) loads a peptide onto the Class I major histocompatibility complex (MHC) for presentation to and interaction with a CD8+ cytotoxic T cell. An antigen presenting cell (APC) loads a peptide onto the Class II MHC for presentation to and interaction with a CD4+ helper T cell (right). Additional co-receptor interactions between the APC and each type of T cell are not displayed here. These interactions play an important role in determining the downstream fate of each T cell, but as yet cannot be predicted bioinformatically. Image based on figures from motifolio.com.

In the absence of data showing which peptides are presented by a given MHC, whole exome sequencing can be used to indirectly predict this. The sequenced DNA can be ‘virtually translated’ into predicted proteins, then the analytic tools described here evaluate which peptides may be presented by a given patient’s HLA repertoire (Figure 2). Here, we will first describe and evaluate the most commonly used MHC Class I, Class II and T cell predictors, highlighting those with data to support their use both in infectious diseases and oncology. We will then describe the most important recent uses of these tools in preclinical and clinical oncologic settings.

An external file that holds a picture, illustration, etc.
Object name is nihms-1039657-f0002.jpg

Example of neoantigen prediction. Red arrow indicates the window that includes a high-affinity nonamer.

MHC Class I prediction

There are many MHC Class I prediction tools (Table 1). Most have been trained on data from the Immune Epitope Database (IEDB) [20]. There are two general categories of prediction tools: allele-specific and pan-specific. Initial programs relied on allele-specific motifs; for example, positions 2 and 9 constitute anchor residues on HLA-A*02:01, commonly occupied by leucine, valine or isoleucine [21]. Other positions are similarly stereotyped [22,23] and unknown input peptides searched for allele-specific motifs. A common problem with these methods is that there are insufficient data from rare alleles to reliably predict peptides which will bind to them. Three-dimensional structural models have been designed, but thus far underperform the models trained on actual data [19●].

Table 1

MHC Class I and Class II prediction tools

NameWebsiteValidated in vitroProsConsMHC Class predictor
BIMAS [31]http://www-bimas.cit.nih.gov/molbio/hla_bind/Yes [32,35]Used historicallyPredictions for selected HLA A, B and C allelesI
RANKPEP [36]http://imed.med.ucm.es/Tools/rankpep.htmlYes [37,38,62]Widely validatedPredictions for selected HLA A, B and C, DP, DQ and DR allelesI, II
SYFPEITHI [26]http://www.syfpeithi.de/Yes [2729, 5961]Widely validatedPredictions for selected HLA A, B and C alleles; Class II limited to HLA DRB1I, II
NetMHC [pan] [24,39] and NetMHCIIpan3.0 [55]http://www.cbs.dtu.dk/services/NetMHCpan/Yes [43●●, 56,57]Pan-specific; Class I has best in vitro and in vivo data, Class II predictor is considered most accurate [18●]Uncommon alleles more difficult to predict due to limited in vitro dataI, II
IEDB recom-mendedhttp://tools.immuneepitope.org/mhci/NoCombines multiple predictive algorithms including NetMHCpanHas not yet been validated, although some component programs have beenI, II
TEPITOPE [pan] [48]http://www.biokdd.fudan.edu.cn/Service/TEPITOPEpan/TEPITOPEpan.htmlYes [49,50, 52,53]Pan-specific; widely validated and user-friendlyMay be inferior to NetMHCIIpan2.0 [18●]II
PickPocket [97]Not publically availableNoPan-specificNot publically availableII
MULTIPRED2 [98]http://cvc.dfci.harvard.edu/multipred2/HTML/prediction2.phpN/APan-specific combination of MULTIPRED, PEPVAC, NetMHCpan and NetMHCIIpanHas not yet been validated, although some component programs have beenII
MultiRTA [99]http://bordnerlab.org/MultiRTANoBased exclusively on experimental binding dataHas not yet been validatedII

To address this problem, pan-specific programs were created to extrapolate from existing data to less common alleles with a paucity of in vitro data. While more useful because of their inclusion of more diverse HLA types, the pan-specific programs do become less accurate as the difference between novel and known HLA allele increases [24]. Prediction tools are generally developed using two steps: model training, then prediction on additional dataset(s). Thus, all of the models have in silico validation by the nature of their development, whereas there are fewer studies validating each in vitro or in vivo.

In the most highly cited paper using bioinformatic software to predict tumor neoantigens [25●●], the authors analyzed 1152 neoepitopes in breast and colorectal cancer assuming HLA-A*0201 and found 10 and 7 novel epitopes per cancer, respectively. The authors used allele-specific predictors BIMAS, SYFPEITHI, RANKPEP and NetMHC.

These MHC Class I predictors remain the most commonly used. SYFPEITHI, one of the first algorithms [26], has been used frequently in the cancer field for epitope prediction [2729] at times in conjunction with other predictors [30]. BIMAS [31] has been used both in infectious disease studies [32,33], and to evaluate cancer-associated antigens [34,35], as has RANKPEP [3638], although less frequently than SYFPEITHI or NetMHC.

The NetMHC programs are the best validated for peptide prediction in infectious diseases and oncology. NetMHC is allele-specific, and generates an IC50 score for the more common HLA types [39]. NetMHCpan is pan-specific and as such has broader applicability [24]. Hereafter, these programs are considered together as they are often cited in general terms by authors.

NetMHC has been used to predict epitopes in viruses associated with malignancy [40,41], neoantigens generated by drug-resistance mutations in Bcr-abl in CML [42] and to identify neoantigens thought to be responsible for spontaneous CD8+ T cell responses in patients with various malignancies [43●●]. The latter example also highlights the limits of predictive programs: although in most cases the cognate wild type peptide was predicted to bind MHC as strongly as the corresponding mutated peptide, in vitro only the mutated peptide stimulated cytolytic activity. This effect was hypothesized to result from the peptide in question affecting TCR interaction rather than MHC binding (24 of 26 cases), retention of one anchor amino acid (2 of 26 cases), or successful tolerance to the native peptide.

Finally, the IEDB Recommended tool provides an amalgam of some of the above predictive strategies (http://tools.immuneepitope.org/mhci/reference/). This program aims to choose the ‘best’ predictor for a given input peptide. While this would seem the ideal method, it has not yet been compared to the other programs listed above.

MHC Class II prediction

MHC Class II is highly polymorphic, and is comprised of, both the core and flanking residues, both of which influence binding to the MHC [44●●,45]. These features make MHC Class II binding prediction very difficult (reviewed in [15]), with MHC Class II predictors consistently underperforming MHC Class I [46]. Ideally, between 100 and 200 experimental peptide binding measurements are needed per MHC molecule for an adequate prediction to be calculated [47], but IEDB, the largest database of MHC binding peptides, has far fewer data points than this on which to train predictive programs.

Multiple tools for predicting MHC Class II binding have been created (Table 1), the most commonly used being TEPITOPEpan [48]. This program attempts to address the problem of insufficient in vitro data by assessing peptide sequence and peptide-MHC binding residues rather than actual binding data. TEPITOPE has been used in infectious disease [4951] although users emphasize its limitations. TEPITOPE has also been used to detect Class II epitopes in malignancy, including for the melanoma antigen gp100 [52] and prostate-specific membrane antigen [53], among many other similar examples. OWA-PSSM is a recently developed tool from the authors of TEPITOPE still undergoing refinement and is not publically available (Wong HS, personal communication) [54].

The other Class II predictors are less commonly used. NetMHCIIpan uses artificial neural network learning to train the model on actual MHC Class II binding data [55]. In one analysis written by the authors of TEPITOPE that compared the pan-specific MHC II predictors, NetMH-CII was found to be superior to both MultiRTA and TEPITOPE [18●]. NetMHCII has been used in rheumatology [56], infectious diseases [57] and malignancy [58]. SYFPEITHI and RANKPEP have also been used in malignancy [5963]. MULTIPRED2, software that combines the NetMHC tools as well as a peptide cleavage tool, is similar to ‘IEDB recommended’ in that it incorporates those programs with the highest validation rate, but it has not yet validated in vitro.

T cell interaction prediction

The MHC-T cell interaction is the most complex, and therefore the hardest to model bioinformatically (Table 2). The most frequently used program is NetCTL, which does not try to directly predict T cell binding. Rather, it uses a combination of MHC binding (NetMHC), C-terminal cleavage affinity (NetCHOP [64]) and transporter associated with antigen processing (TAP) transport [65] to generate its score. This program has been used commonly in infectious disease [6669], and infrequently in malignancy [70,71], where again the authors comment on the limitations of this strategy, including limited selection of HLA alleles, and the presence of epitopes found experimentally that were not predicted bioinformatically. The IEDB T cell epitope prediction tool is a recently developed tool [72] that predicts immunogenicity based on peptide position and amino acid characteristics, but it has not yet been validated in vitro.

Table 2

T cell prediction tools

NameWebsiteValidated in vitroProsCons
EpicCapo [78]http://pirun.ku.ac.th/~fsciiok/EpicCapoREF.zipNo9 amino acid input, with plan to expand to 8–11Predictions for selected HLA alleles
POPISK [74]http://iclab.life.nctu.edu.tw/POPISKNoTakes into account HLA typeHas not yet been validated
EpiMatrix [79●]http://www.epivax.com/epimatrix/Yes [100,101]Validated in vitroProprietary tool
NetCTLpan [102]http://www.cbs.dtu.dk/services/NetCTL/Yes [66,67]Validated in vitro; is really a combination of Class I, peptide cleavage and TAP transport modelsFocuses on MHC presentation rather than TCR itself
iMatrix [75] (formerly PAComplex)http://pacomplex.life.nctu.edu.tw/index_iM.phpNoUser-friendlyHas not yet been validated
IEDB tool [72]http://tools.immuneepitope.org/main/html/tcell_tools.htmlYes [103]User-friendly; good in silico validationPaucity of validation
CTLPred [76]http://www.imtech.res.in/raghava/ctlpred/NoUser-friendlyHas not yet been validated

There are several other T cell epitope predictors, but none has been validated. POPI was the first T-cell reactivity predictor [73]; POPISK [74] takes into account MHC allele restriction has been used in infectious disease [57] but not malignancy. iMatrix models the TCR-peptide and peptide-MHC interfaces [75]. CTLPred [76] has been used retrospectively to predict epitopes from Ebola virus but these were not validated in vitro, and this program underperformed relative to SYFPEITHI and IEDB for predicting HIV-specific epitopes [77]. EpicCapo + REF has been used to search for promiscuous epitopes (epitopes presented by HLA supertypes) of influenza [78], but has not been used in cancer.

EpiMatrix [79●] has been used predominantly by the group involved in its creation to look at infectious organisms [8083]. In its first use in malignancy, an abstract describes the use of EpiMatrix to identify CD4+ and CD8+ T cell epitopes of the protein survivin, which was used to induce tumor regression in a mouse model of malignant mesothelioma [84].

Summary: which tool, individually or concurrently?

At present, MHC prediction tools are typically used in malignancy to retrospectively predict which of a given tumor’s mutation(s) led to neoantigen formation. Two important questions remain: which tool should be used, and should multiple tools be used concurrently?

There have been several comparative assessments of the prediction tools. A 2007 study of 16 MHC Class I prediction tools evaluated each program separately and in conjunction. This work included the commonly used programs BIMAS, SYFPEITHI, RANKPEP and NetMHC, and reported a modest improvement in sensitivity when the programs were combined [85●]. An international competition for machine learning in 2009 found that NetMHC outperformed both PickPocket and the allele-specific programs BIMAS and SYFPEITHI (described in [16]). A 2012 comparison of the MHC Class I and Class II pan-specific methods found the NetMHC tools performed best for both classes [18●].

In light of these studies, and given existing data from both translational and in vitro studies, the NetMHC series programs have been best validated, with the strongest evidence for MHC Class I prediction, weaker for MHC Class II and the least for T cell recognition. The question of whether NetMHC should be combined with other predictive tools remains to be elucidated.

Preclinical data for neoantigen prediction in malignancy

There are several examples of preclinical models showing the importance of neoantigens in malignancy. As early as 1995, Mandelboim et al. described anti-tumor cytotoxic T lymphocyte responses against neoantigens from a murine lung carcinoma [86●●]. He showed that vaccination with these peptides both protected mice from new metastases and reduced metastatic load in mice with micrometastases.

Three landmark studies published in 2012 demonstrated the importance of tumor neoantigens to tumor control versus escape in mouse models, two of which used NetMHC for neoantigen prediction. In a mouse model of chemically induced sarcoma, Matsushita et al. identified the neoantigen responsible for spontaneous tumor rejection [87●●]. Cell lines harboring this mutation were rejected in most wild type mice, whereas 20% of these tumors underwent immunoediting of the neoantigen, and were able to grow. In a subsequent study, the same group defined the neoantigens responsible for the antitumor effects of anti-CTLA-4 and anti-PD-1 immunotherapy [88●●].

The Sahin lab similarly used next generation sequencing and NetMHC prediction to design tumor-specific vaccines using neoantigens derived from the melanoma cell line B16F10 [89●●]. They selected 50 mutations out of the 962 nonsynonymous exonic mutations for validation; of these, 16 elicited immune responses in immunized mice when delivered with polyI:C, and two vaccines delayed tumor growth.

The Jacks lab treated mice with soft tissue sarcoma with and without lentivirus that expressed known T-cell antigens [90●]. Immunocompetent mice had a delayed development of tumors that was even more dramatic in the presence of T-cell antigens, and that was abrogated upon treatment with anti-CD4+ and anti-CD8+ antibodies. These neoantigens were exogenously expressed, rather than representing the mutant form of an existing wild type peptide.

Moving into the clinic: tumor-associated antigens and neoantigens

Seminal papers in both solid and liquid tumors have demonstrated an important role for tumor neoantigens in immunotherapy. Lennerz et al. used a cDNA library from a patient-derived melanoma cell line to screen for tumor-reactive autologous T cells, and identified four neoantigens that induced T cell response as measured by IFN-gamma ELISPOT [7●]. The same year, Rosenberg and Robbins also discovered two neoantigens recognized by adoptively transferred tumor infiltrating lymphocytes in a single patient with metastatic melanoma treated with adoptive T cell immunotherapy [91●]. Intriguingly, their data suggests that the relevant T cell clones existed before their ex vivo expansion: the autologous T cell therapy was amplifying an existing antineoantigen tumor response.

More recently the same investigators used bioinformatic prediction to narrow down candidate neoantigens in three melanoma patients treated with TIL therapy [92●]. After predicting neoantigens from an HLA-A*0201 patient-derived cell line, the group tested the ability for autologous TILs to recognize 55 neoantigen peptides when presented by the cell line T2. Four peptides predicted by NetMHCPan2.4 led to IFN-gamma release by TIL. Little to no response was seen for the corresponding wild type peptides. For a two additional, non-A*0201 patients (in whom the T2 system could not be used), after incubation with HLA-matched target cells 3 of 53 and 4 of 46 tested peptides led to TIL activation. Importantly, responses to some of the neoantigens were measurable in the peripheral blood, albeit greater than two logs less than in the TILs.

The Rosenberg group has since expanded its therapies to include other tumor types. A patient with metastatic cholangiocarcinoma treated with adoptive transfer of tumor infiltrating lymphocytes containing approximately 25% of neoantigen-specific CD4+ T helper cells transiently responded to initial therapy [4●●]. When her cancer subsequently progressed, the patient was retreated with a near-pure population of neoantigen-specific T cells and responded. This case illustrates several important points: in addition to providing proof of principle that these therapies can work in epithelial malignancies, it also shows the necessity for expanding our neoantigen prediction strategies beyond MHC Class I/CD8+ T cell interaction, as the important population in this case was comprised CD4+ T cells.

In the first published study of neoantigens in the response to checkpoint blockade therapy, the Schumacher group sequenced the tumor of a patient with metastatic melanoma who had a partial response to treatment with CTLA-4 blockade by ipilimumab [93●●]. They then used NetMHC3.2, NetCHOP and RNA expression to predict 448 peptide neoantigens. After testing all these peptides using multimer technology, they found one strong and one weak neoantigen peptide. A T cell response to the strong neoantigen was seen both in TILs and in PBMC, and was not seen against the corresponding wild type peptide. Our group applied similar concepts to a study of 64 metastatic melanoma patients treated with the CTLA4 blockade agents ipilimumab or tremelimumab. Mutational load appeared to correlate with response but alone did not perfectly predict response, as there were some tumors with a high mutational load that did not respond to treatment, and conversely relatively lower burden tumors that did respond. Relying primarily on the NetMHC Class I prediction tool for neoantigen prediction, we found that specific neoantigens predicted to be presented by patient-specific HLA correlated with benefit from therapy [94●●]. These neoantigens appeared to have substrings in common, suggesting T cell degeneracy as has been described [95●]. This phenomenon, which is reminiscent of molecular mimicry, is being addressed by ongoing studies by our group. Several predicted neoantigens were validated in vitro with intracellular cytokine staining using autologous peripheral blood from the treated patients stimulated with synthetic peptides.

The Wu group has used neoantigen prediction extensively. In one example, the investigators used NetMHCpan v2.4 to predict peptide neoantigens occurring in chronic lymphocytic leukemia (CLL) that are presented by patient-specific MHC Class I [96●●]. They tested 9 and 10 amino acid peptides predicted to be presented by HLA-A or HLA-B in vitro in a competitive binding assay and validated 76.5% of peptides predicted to bind with an IC50 of less than 150, and 36% of weak binders (≤500 nM), for a total of a 55% correspondence between NetMHC prediction and the competitive binding assay. When they translated this into patient material, 2 out of 30 peptides tested for one patient, and 1 out of 37 for a second patient led to T cell responses. The authors emphasize that this underestimation of neoantigens on the part of NetMHC pre-dominantly results from the relative insensitivity of in vitro methods to validate T cell reactivity. Thus, the actual number of neoantigens is likely much higher. In an exciting next step for the field of neoantigen prediction and application, a trial using the prediction method to create a patient-specific vaccine (termed NeoVax) is currently being run out of the Harvard system cancer centers (Brigham and Women’s and Dana Farber Cancer Institute ()).

Summary and conclusions

With the increasing use of immunotherapies to treat malignancy, it will become critically important to accurately triage patients to treatments that are most likely to be effective. Novel peptide neoantigens resulting from somatic exonic mutations have been shown conclusively to elicit T cell responses in patients. Prediction tools have proliferated for MHC Class I, Class II and T cell interactions, as well as peptide cleavage. Of the existing tools, based on comparative studies and validation data to date, we recommend using the NetMHC programs for MHC Class I prediction. The apparent low positive predictive value of this tool may actually reflect the relatively insensitive methods used to validate the peptides in vitro, rather than a deficiency of the tool itself. The predictive tools for MHC Class II and T cell interaction are not yet sufficiently validated for routine use. Furthermore, a wild type peptide that is predicted to be presented does not necessarily imply that a peptide-specific response will be seen when tested using patient material. Ultimately, predictive tools in their current form may help narrow candidate peptides for in vitro testing, but these algorithms will need ongoing refinement before they are ready for routine clinical use.

References and recommended reading

Papers of particular interest, published within the period of review, have been highlighted as:

● of special interest

●● of outstanding interest

1. Dutcher JP, Creekmore S, Weiss GR, Margolin K, Markowitz AB, Roper M, Parkinson D, Ciobanu N, Fisher RI, Boldt DH et al.: A phase II study of interleukin-2 and lymphokine-activated killer cells in patients with metastatic malignant melanoma. J Clin Oncol 1989, 7:477–485. [PubMed] [Google Scholar]
2. Hodi FS, O’Day SJ, McDermott DF, Weber RW, Sosman JA, Haanen JB, Gonzalez R, Robert C, Schadendorf D, Hassel JC et al.: Improved survival with ipilimumab in patients with metastatic melanoma. N Engl J Med 2010, 363:711–723. [PMC free article] [PubMed] [Google Scholar]
3. Wolchok JD, Kluger H, Callahan MK, Postow MA, Rizvi NA, Lesokhin AM, Segal NH, Ariyan CE, Gordon RA, Reed K et al.: Nivolumab plus ipilimumab in advanced melanoma. N Engl J Med 2013, 369:122–133. [PMC free article] [PubMed] [Google Scholar]
4. ●●. Tran E, Turcotte S, Gros A, Robbins PF, Lu YC, Dudley ME, Wunderlich JR, Somerville RP, Hogan K, Hinrichs CS et al.: Cancer immunotherapy based on mutation-specific CD4+ T cells in a patient with epithelial cancer. Science 2014, 344:641–645. [PMC free article] [PubMed] [Google Scholar]
This study illustrated that expanded tumor infiltrating lymphocytes that recognized a neoantigen predicted by NetMHC led to the regression of a cholangiocarcinoma. Importantly, these were CD4+ TH1 cells.
5. ●. Hacohen N, Fritsch EF, Carter TA, Lander ES, Wu CJ: Getting personal with neoantigen-based therapeutic cancer vaccines. Cancer Immunol Res 2013, 1:11–15. [PMC free article] [PubMed] [Google Scholar]
This review discusses advantages and challenges of vaccination with neoantigens rather than the vaccine strategies used previously, which predominantly relied on overexpressed self antigens.
6. ●. Mandelboim O, Berke G, Fridkin M, Feldman M, Eisenstein M, Eisenbach L: CTL induction by a tumour-associated antigen octapeptide derived from a murine lung carcinoma. Nature 1994, 369:67–71. [PubMed] [Google Scholar]
One of the pioneering studies in the neoantigen field, this study showed that anti-tumor cytotoxic lymphocytes recognized the mutant form for the connexin peptide.
7. ●. Lennerz V, Fatho M, Gentilini C, Frye RA, Lifke A, Ferel D, Wolfel C, Huber C, Wolfel T: The response of autologous T cells to a human melanoma is dominated by mutated neoantigens. Proc Natl Acad Sci U S A 2005, 102:16013–16018. [PMC free article] [PubMed] [Google Scholar]
This study, often cited in the neoantigen literature, identified both tumor-associated and mutated peptides using cDNA expression screening.
8. Kenter GG, Welters MJ, Valentijn AR, Lowik MJ, Berends-van der Meer DM, Vloon AP, Essahsah F, Fathers LM, Offringa R, Drijfhout JW et al.: Vaccination against HPV-16 oncoproteins for vulvar intraepithelial neoplasia. N Engl J Med 2009, 361:1838–1847. [PubMed] [Google Scholar]
9. Davila ML, Riviere I, Wang X, Bartido S, Park J, Curran K, Chung SS, Stefanski J, Borquez-Ojeda O, Olszewska M et al.: Efficacy and toxicity management of 19–28z CAR T cell therapy in B cell acute lymphoblastic leukemia. Sci Transl Med 2014, 6:224ra225. [PMC free article] [PubMed] [Google Scholar]
10. Grupp SA, Kalos M, Barrett D, Aplenc R, Porter DL, Rheingold SR, Teachey DT, Chew A, Hauck B, Wright JF et al.: Chimeric antigen receptor-modified T cells for acute lymphoid leukemia. N Engl J Med 2013, 368:1509–1518. [PMC free article] [PubMed] [Google Scholar]
11. Dao T, Yan S, Veomett N, Pankov D, Zhou L, Korontsvit T, Scott A, Whitten J, Maslak P, Casey E et al.: Targeting the intracellular WT1 oncogene product with a therapeutic human antibody. Sci Transl Med 2013, 5:176ra133. [PMC free article] [PubMed] [Google Scholar]
12. Janeway CA, Travers P, Walport M, Shlomchik M: Immunobiology: The Immune System in Health and Disease edn 6 New York: Garland Science Publishing; 2005,. [Google Scholar]
13. Robinson J, Mistry K, McWilliam H, Lopez R, Parham P, Marsh SG: The IMGT/HLA database. Nucleic Acids Res 2011, 39:D1171–D1176. [PMC free article] [PubMed] [Google Scholar]
14. Assarsson E, Sidney J, Oseroff C, Pasquetto V, Bui HH, Frahm N, Brander C, Peters B, Grey H, Sette A: A quantitative analysis of the variables affecting the repertoire of T cell specificities recognized after vaccinia virus infection. J Immunol 2007, 178:7890–7901. [PubMed] [Google Scholar]
15. Nielsen M, Lund O, Buus S, Lundegaard C: MHC Class II epitope predictive algorithms. Immunology 2010, 130:319–328. [PMC free article] [PubMed] [Google Scholar]
16. Lundegaard C, Lund O, Buus S, Nielsen M: Major Histocompatibility Complex Class I binding predictions as a tool in epitope discovery. Immunology 2010, 130:309–318. [PMC free article] [PubMed] [Google Scholar]
17. Lin HH, Ray S, Tongchusak S, Reinherz EL, Brusic V: Evaluation of MHC Class I peptide binding prediction servers: applications for vaccine research. BMC Immunol 2008, 9:8. [PMC free article] [PubMed] [Google Scholar]
18. ●. Zhang L, Udaka K, Mamitsuka H, Zhu S: Toward more accurate pan-specific MHC-peptide binding prediction: a review of current methods and tools. Brief Bioinform 2012, 13: 350–364. [PubMed] [Google Scholar]
This review describes then evaluates pan-specific methods of MHC binding prediction.
19. ●. Lundegaard C, Lund O, Kesmir C, Brunak S, Nielsen M: Modeling the adaptive immune system: predictions and simulations. Bioinformatics 2007, 23:3265–3275. [PMC free article] [PubMed] [Google Scholar]
This review discusses advances in MHC Class I and Class II prediction as well as the ways in which the latter is not yet as accurate as the former.
20. Vita R, Zarebski L, Greenbaum JA, Emami H, Hoof I, Salimi N, Damle R, Sette A, Peters B: The immune epitope database 2.0. Nucleic Acids Res 2010, 38:D854–D862. [PMC free article] [PubMed] [Google Scholar]
21. Falk K, Rotzschke O, Stevanovic S, Jung G, Rammensee HG: Allele-specific motifs revealed by sequencing of self-peptides eluted from MHC molecules. Nature 1991, 351: 290–296. [PubMed] [Google Scholar]
22. Ruppert J, Sidney J, Celis E, Kubo RT, Grey HM, Sette A: Prominent role of secondary anchor residues in peptide binding to HLA-A2.1 molecules. Cell 1993, 74:929–937. [PubMed] [Google Scholar]
23. Madden DR, Garboczi DN, Wiley DC: The antigenic identity of peptide-MHC complexes: a comparison of the conformations of five viral peptides presented by HLA-A2. Cell 1993, 75:693–708. [PubMed] [Google Scholar]
24. Nielsen M, Lundegaard C, Blicher T, Lamberth K, Harndahl M, Justesen S, Roder G, Peters B, Sette A, Lund O et al.: NetMHCpan, a method for quantitative predictions of peptide binding to any HLA-A and -B locus protein of known sequence. PLoS ONE 2007, 2:e796. [PMC free article] [PubMed] [Google Scholar]
25. ●●. Segal NH, Parsons DW, Peggs KS, Velculescu V, Kinzler KW, Vogelstein B, Allison JP: Epitope landscape in breast and colorectal cancer. Cancer Res 2008, 68:889–892. [PubMed] [Google Scholar]
Perhaps the most cited paper in the neoantigen literature, this paper addressed the relationship between mutations and neoantigens in a series of breast and colorectal cancer patients using a composite of the methods listed in Table 1.
26. Rammensee HG, Friede T, Stevanoviic S: MHC ligands and peptide motifs: first listing. Immunogenetics 1995, 41:178–228. [PubMed] [Google Scholar]
27. Huebener N, Fest S, Strandsby A, Michalsky E, Preissner R, Zeng Y, Gaedicke G, Lode HN: A rationally designed tyrosine hydroxylase DNA vaccine induces specific antineuroblastoma immunity. Mol Cancer Ther 2008, 7:2241–2251. [PubMed] [Google Scholar]
28. Kamata Y, Kuhara A, Iwamoto T, Hayashi K, Koido S, Kimura T, Egawa S, Homma S: Identification of HLA Class I-binding peptides derived from unique cancer-associated proteins by mass spectrometric analysis. Anticancer Res 2013, 33: 1853–1859. [PubMed] [Google Scholar]
29. Cobbold M, De La Pena H, Norris A, Polefrone JM, Qian J, English AM, Cummings KL, Penny S, Turner JE, Cottine J et al.: MHC Class I-associated phosphopeptides are the targets of memory-like immunity in leukemia. Sci Transl Med 2013, 5:203ra125. [PMC free article] [PubMed] [Google Scholar]
30. Greiner J, Ono Y, Hofmann S, Schmitt A, Mehring E, Gotz M, Guillaume P, Dohner K, Mytilineos J, Dohner H et al.: Mutated regions of nucleophosmin 1 elicit both CD4(+) and CD8(+) T-cell responses in patients with acute myeloid leukemia. Blood 2012, 120:1282–1289. [PubMed] [Google Scholar]
31. Parker KC, Bednarek MA, Coligan JE: Scheme for ranking potential HLA-A2 binding peptides based on independent binding of individual peptide side-chains. J Immunol 1994, 152:163–175. [PubMed] [Google Scholar]
32. Sundar K, Boesen A, Coico R: Computational prediction and identification of HLA-A2.1-specific Ebola virus CTL epitopes. Virology 2007, 360:257–263. [PubMed] [Google Scholar]
33. Moutaftsi M, Peters B, Pasquetto V, Tscharke DC, Sidney J, Bui HH, Grey H, Sette A: A consensus epitope prediction approach identifies the breadth of murine T(CD8+)-cell responses to vaccinia virus. Nat Biotechnol 2006, 24:817–819. [PubMed] [Google Scholar]
34. Michaeli Y, Denkberg G, Sinik K, Lantzy L, Chih-Sheng C, Beauverd C, Ziv T, Romero P, Reiter Y: Expression hierarchy of T cell epitopes from melanoma differentiation antigens: unexpected high level presentation of tyrosinase-HLA-A2 complexes revealed by peptide-specific, MHC-restricted. TCR-like antibodies. J Immunol 2009, 182:6328–6341. [PubMed] [Google Scholar]
35. Cortez-Gonzalez X, Sidney J, Adotevi O, Sette A, Millard F, Lemonnier F, Langlade-Demoyen P, Zanetti M: Immunogenic HLA-B7-restricted peptides of hTRT. Int Immunol 2006, 18:1707–1718. [PubMed] [Google Scholar]
36. Reche PA, Reinherz EL: Prediction of peptide-MHC binding using profiles. Methods Mol Biol 2007, 409:185–200. [PubMed] [Google Scholar]
37. Singh R, Paterson Y: Vaccination strategy determines the emergence and dominance of CD8+ T-cell epitopes in a FVB/N rat HER-2/neu mouse model of breast cancer. Cancer Res 2006, 66:7748–7757. [PubMed] [Google Scholar]
38. Fortier MH, Caron E, Hardy MP, Voisin G, Lemieux S, Perreault C, Thibault P: The MHC Class I peptide repertoire is molded by the transcriptome. J Exp Med 2008, 205:595–610. [PMC free article] [PubMed] [Google Scholar]
39. Lundegaard C, Lamberth K, Harndahl M, Buus S, Lund O, Nielsen M: 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–W512. [PMC free article] [PubMed] [Google Scholar]
40. Lyngaa R, Pedersen NW, Schrama D, Thrue CA, Ibrani D, Met O, Thor Straten P, Nghiem P, Becker JC, Hadrup SR: T-cell responses to oncogenic merkel cell polyomavirus proteins distinguish patients with merkel cell carcinoma from healthy donors. Clin Cancer Res 2014, 20:1768–1778. [PMC free article] [PubMed] [Google Scholar]
41. Riemer AB, Keskin DB, Zhang G, Handley M, Anderson KS, Brusic V, Reinhold B, Reinherz EL: A conserved E7-derived cytotoxic T lymphocyte epitope expressed on human papillomavirus 16-transformed HLA-A2+ epithelial cancers. J Biol Chem 2010, 285:29608–29622. [PMC free article] [PubMed] [Google Scholar]
42. Cai A, Keskin DB, DeLuca DS, Alonso A, Zhang W, Zhang GL, Hammond NN, Nardi V, Stone RM, Neuberg D et al.: Mutated BCR-ABL generates immunogenic T-cell epitopes in CML patients. Clin Cancer Res 2012, 18:5761–5772. [PMC free article] [PubMed] [Google Scholar]
43. ●●. Fritsch EF, Rajasagi M, Ott PA, Brusic V, Hacohen N, Wu CJ: HLA-binding properties of tumor neoepitopes in humans. Cancer Immunol Res 2014, 2:522–529. [PMC free article] [PubMed] [Google Scholar]
This restrospective study applies MHC Class I prediction to documented cases of spontaneous tumor regression, showing the utility of this approach to epitope selection.
44. ●●. Godkin AJ, Smith KJ, Willis A, Tejada-Simon MV, Zhang J, Elliott T, Hill AV: Naturally processed HLA Class II peptides reveal highly conserved immunogenic flanking region sequence preferences that reflect antigen processing rather than peptide-MHC interactions. J Immunol 2001, 166:6720–6727. [PubMed] [Google Scholar]
This study describes the importance of the peptide flanking residues, in addition to the core binding residues, to MHC Class II binding, illustrative of the complexities of MHC Class II prediction.
45. Rotzschke O, Falk K, Mack J, Lau JM, Jung G, Strominger JL: Conformational variants of Class II MHC/peptide complexes induced by N-and C-terminal extensions of minimal peptide epitopes. Proc Natl Acad Sci U S A 1999, 96:7445–7450. [PMC free article] [PubMed] [Google Scholar]
46. Lin HH, Zhang GL, Tongchusak S, Reinherz EL, Brusic V: Evaluation of MHC-II peptide binding prediction servers: applications for vaccine research. BMC Bioinform 2008, 9(Suppl. 12):S22. [PMC free article] [PubMed] [Google Scholar]
47. Jorgensen KW, Buus S, Nielsen M: Structural properties of MHC Class II ligands, implications for the prediction of MHC Class II epitopes. PLoS ONE 2010, 5:e15877. [PMC free article] [PubMed] [Google Scholar]
48. Zhang L, Chen Y, Wong HS, Zhou S, Mamitsuka H, Zhu S: TEPITOPEpan: extending TEPITOPE for peptide binding prediction covering over 700 HLA-DR molecules. PLoS ONE 2012, 7:e30483. [PMC free article] [PubMed] [Google Scholar]
49. Iwai LK, Yoshida M, Sidney J, Shikanai-Yasuda MA, Goldberg AC, Juliano MA, Hammer J, Juliano L, Sette A, Kalil J et al.: In silico prediction of peptides binding to multiple HLA-DR molecules accurately identifies immunodominant epitopes from gp43 of Paracoccidioides brasiliensis frequently recognized in primary peripheral blood mononuclear cell responses from sensitized individuals. Mol Med 2003, 9:209–219. [PMC free article] [PubMed] [Google Scholar]
50. Nyarady Z, Czompoly T, Bosze S, Nagy G, Petrohai A, Pal J, Hudecz F, Berki T, Nemeth P: Validation of in silico prediction by in vitro immunoserological results of fine epitope mapping on citrate synthase specific autoantibodies. Mol Immunol 2006,:830–838. [PubMed]
51. Drouin EE, Glickstein L, Kwok WW, Nepom GT, Steere AC: Searching for borrelial T cell epitopes associated with antibiotic-refractory Lyme arthritis. Mol Immunol 2008, 45:2323–2332. [PMC free article] [PubMed] [Google Scholar]
52. Cochlovius B, Stassar M, Christ O, Raddrizzani L, Hammer J, Mytilineos I, Zoller M: In vitro and in vivo induction of a Th cell response toward peptides of the melanoma-associated glycoprotein 100 protein selected by the TEPITOPE program. J Immunol 2000, 165:4731–4741. [PubMed] [Google Scholar]
53. Schroers R, Shen L, Rollins L, Xiao Z, Sonderstrup G, Slawin K, Huang XF, Chen SY: Identification of MHC Class II-restricted T-cell epitopes in prostate-specific membrane antigen. Clin Cancer Res 2003, 9:3260–3271. [PubMed] [Google Scholar]
54. Shen WJ, Zhang S, Wong HS: An effective and efficient peptide binding prediction approach for a broad set of HLA-DR molecules based on ordered weighted averaging of binding pocket profiles. Proteome Sci 2013, 11:S15. [PMC free article] [PubMed] [Google Scholar]
55. Nielsen M, Lundegaard C, Blicher T, Peters B, Sette A, Justesen S, Buus S, Lund O: Quantitative predictions of peptide binding to any HLA-DR molecule of known sequence: NetMHCIIpan. PLoS Comput Biol 2008, 4:e1000107. [PMC free article] [PubMed] [Google Scholar]
56. Duvvuri B, Duvvuri VR, Wang C, Chen L, Wagar LE, Jamnik V, Wu J, Yeung RS, Grigull J, Watts TH et al.: The human immune system recognizes neopeptides derived from mitochondrial DNA deletions. J Immunol 2014, 192:4581–4591. [PubMed] [Google Scholar]
57. Jung KO, Khan AM, Tan BY, Hu Y, Simon GG, Nascimento EJ, Lemonnier F, Brusic V, Miotto O, Tan TW et al.: West Nile virus T-cell ligand sequences shared with other flaviviruses: a multitude of variant sequences as potential altered peptide ligands. J Virol 2012, 86:7616–7624. [PMC free article] [PubMed] [Google Scholar]
58. Godet Y, Fabre E, Dosset M, Lamuraglia M, Levionnois E, Ravel P, Benhamouda N, Cazes A, Le Pimpec-Barthes F, Gaugler B et al.: Analysis of spontaneous tumor-specific CD4 T-cell immunity in lung cancer using promiscuous HLA-DR telomerase-derived epitopes: potential synergistic effect with chemotherapy response. Clin Cancer Res 2012, 18:2943–2953. [PubMed] [Google Scholar]
59. Calvo-Calle JM, Strug I, Nastke MD, Baker SP, Stern LJ: Human CD4+ T cell epitopes from vaccinia virus induced by vaccination or infection. PLoS Pathog 2007, 3:1511–1529. [PMC free article] [PubMed] [Google Scholar]
60. Neumann F, Wagner C, Preuss KD, Kubuschok B, Schormann C, Stevanovic S, Pfreundschuh M: Identification of an epitope derived from the cancer testis antigen HOM-TES-14/SCP1 and presented by dendritic cells to circulating CD4+ T cells. Blood 2005, 106:3105–3113. [PubMed] [Google Scholar]
61. May RJ, Dao T, Pinilla-Ibarz J, Korontsvit T, Zakhaleva V, Zhang RH, Maslak P, Scheinberg DA: Peptide epitopes from the Wilms’ tumor 1 oncoprotein stimulate CD4+ and CD8+ T cells that recognize and kill human malignant mesothelioma tumor cells. Clin Cancer Res 2007, 13:4547–4555. [PubMed] [Google Scholar]
62. Arnold IC, Hitzler I, Engler D, Oertli M, Agger EM, Muller A: The C-terminally encoded, MHC Class II-restricted T cell antigenicity of the Helicobacter pylori virulence factor CagA promotes gastric preneoplasia. J Immunol 2011, 186:6165–6172. [PubMed] [Google Scholar]
63. Bioley G, Dousset C, Yeh A, Dupont B, Bhardwaj N, Mears G, Old LJ, Ayyoub M, Valmori D: Vaccination with recombinant NY-ESO-1 protein elicits immunodominant HLA-DR52b-restricted CD4+ T cell responses with a conserved T cell receptor repertoire. Clin Cancer Res 2009, 15:4467–4474. [PubMed] [Google Scholar]
64. Nielsen M, Lundegaard C, Lund O, Kesmir C: The role of the proteasome in generating cytotoxic T-cell epitopes: insights obtained from improved predictions of proteasomal cleavage. Immunogenetics 2005, 57:33–41. [PubMed] [Google Scholar]
65. Peters B, Bulik S, Tampe R, Van Endert PM, Holzhutter HG: Identifying MHC Class I epitopes by predicting the TAP transport efficiency of epitope precursors. J Immunol 2003, 171:1741–1749. [PubMed] [Google Scholar]
66. Wang M, Lamberth K, Harndahl M, Roder G, Stryhn A, Larsen MV, Nielsen M, Lundegaard C, Tang ST, Dziegiel MH et al.: CTL epitopes for influenza A including the H5N1 bird flu; genome-, pathogen-, and HLA-wide screening. Vaccine 2007, 25: 2823–2831. [PubMed] [Google Scholar]
67. Perez CL, Larsen MV, Gustafsson R, Norstrom MM, Atlas A, Nixon DF, Nielsen M, Lund O, Karlsson AC: Broadly immunogenic HLA Class I supertype-restricted elite CTL epitopes recognized in a diverse population infected with different HIV-1 subtypes. J Immunol 2008, 180:5092–5100. [PubMed] [Google Scholar]
68. Tang ST, van Meijgaarden KE, Caccamo N, Guggino G, Klein MR, van Weeren P, Kazi F, Stryhn A, Zaigler A, Sahin U et al.: Genome-based in silico identification of new Mycobacterium tuberculosis antigens activating polyfunctional CD8+ T cells in human tuberculosis. J Immunol 2011, 186:1068–1080. [PubMed] [Google Scholar]
69. Blom K, Braun M, Ivarsson MA, Gonzalez VD, Falconer K, Moll M, Ljunggren HG, Michaelsson J, Sandberg JK: Temporal dynamics of the primary human T cell response to yellow fever virus 17D as it matures from an effector-to a memory-type response. J Immunol 2013, 190:2150–2158. [PubMed] [Google Scholar]
70. Lamers CH, Willemsen R, van Elzakker P, van Steenbergen-Langeveld S, Broertjes M, Oosterwijk-Wakka J, Oosterwijk E, Sleijfer S, Debets R, Gratama JW: Immune responses to transgene and retroviral vector in patients treated with ex vivo-engineered T cells. Blood 2011, 117:72–82. [PubMed] [Google Scholar]
71. Graf C, Heidel F, Tenzer S, Radsak MP, Solem FK, Britten CM, Huber C, Fischer T, Wolfel T: A neoepitope generated by an FLT3 internal tandem duplication (FLT3-ITD) is recognized by leukemia-reactive autologous CD8+ T cells. Blood 2007, 109:2985–2988. [PubMed] [Google Scholar]
72. Calis JJ, Maybeno M, Greenbaum JA, Weiskopf D, De Silva AD, Sette A, Kesmir C, Peters B: Properties of MHC Class I presented peptides that enhance immunogenicity. PLoS Comput Biol 2013, 9:e1003266. [PMC free article] [PubMed] [Google Scholar]
73. Tung CW, Ho SY: POPI: predicting immunogenicity of MHC Class I binding peptides by mining informative physicochemical properties. Bioinformatics 2007, 23:942–949. [PubMed] [Google Scholar]
74. Tung CW, Ziehm M, Kamper A, Kohlbacher O, Ho SY: POPISK: T-cell reactivity prediction using support vector machines and string kernels. BMC Bioinform 2011, 12:446. [PMC free article] [PubMed] [Google Scholar]
75. Liu IH, Lo YS, Yang JM: PAComplex: a web server to infer peptide antigen families and binding models from TCR-pMHC complexes. Nucleic Acids Res 2011, 39:W254–W260. [PMC free article] [PubMed] [Google Scholar]
76. Bhasin M, Raghava GP: Prediction of CTL epitopes using QM, SVM and ANN techniques. Vaccine 2004, 22:3195–3204. [PubMed] [Google Scholar]
77. Roider J, Meissner T, Kraut F, Vollbrecht T, Stirner R, Bogner JR, Draenert R: Comparison of experimental fine-mapping to in-silico prediction results of HIV-1 epitopes reveals ongoing need for mapping experiments. Immunology 2014, 143: 193–201. [PMC free article] [PubMed] [Google Scholar]
78. Saethang T, Hirose O, Kimkong I, Tran VA, Dang XT, Nguyen LA, Le TK, Kubo M, Yamada Y, Satou K: EpicCapo: epitope prediction using combined information of amino acid pairwise contact potentials and HLA-peptide contact site information. BMC Bioinform 2012, 13:313. [PMC free article] [PubMed] [Google Scholar]
79. ●. Schafer JR, Jesdale BM, George JA, Kouttab NM, De Groot AS: Prediction of well-conserved HIV-1 ligands using a matrix-based algorithm, EpiMatrix. Vaccine 1998, 16:1880–1884. [PubMed] [Google Scholar]
This paper describes the development and mechanism of the predictor EpiMatrix in order to discover new HIV-1 epitopes. It also illustrates that certain peptides are conserved across strains and likely to be presented by more than one HLA molecule.
80. De Groot AS, Ardito M, McClaine EM, Moise L, Martin WD: Immunoinformatic comparison of T-cell epitopes contained in novel swine-origin influenza A (H1N1) virus with epitopes in 2008–2009 conventional influenza vaccine. Vaccine 2009, 27:5740–5747. [PubMed] [Google Scholar]
81. Moise L, McMurry JA, Buus S, Frey S, Martin WD, De Groot AS: In silico-accelerated identification of conserved and immunogenic variola/vaccinia T-cell epitopes. Vaccine 2009, 27:6471–6479. [PMC free article] [PubMed] [Google Scholar]
82. McMurry JA, Gregory SH, Moise L, Rivera D, Buus S, De Groot AS: Diversity of Francisella tularensis Schu4 antigens recognized by T lymphocytes after natural infections in humans: identification of candidate epitopes for inclusion in a rationally designed tularemia vaccine. Vaccine 2007, 25:3179–3191. [PubMed] [Google Scholar]
83. Mishra S, Losikoff PT, Self AA, Terry F, Ardito MT, Tassone R, Martin WD, De Groot AS, Gregory SH: Peptide-pulsed dendritic cells induce the hepatitis C viral epitope-specific responses of naive human T cells. Vaccine 2014, 32:3285–3292. [PubMed] [Google Scholar]
84. De Groot AS, Bertino P, Hoffman P, Carbone M, Siccardi A, Terry F, Cousens L, Moise L, Martin W: Highly immunogenic vaccine for prevention and therapy of malignant mesothelioma (P4437). J Immunol 2013, 190 126.124. [Google Scholar]
85. ●. Trost B, Bickis M, Kusalik A: Strength in numbers: achieving greater accuracy in MHC-I binding prediction by combining the results from multiple prediction tools. Immunome Res 2007, 3:5. [PMC free article] [PubMed] [Google Scholar]
This study argues that combining prediction tools increases the predictive value over each individual tool; this paper does not include the most recent versions of each program, due to its publication in 2007.
86. ●●. Mandelboim O, Vadai E, Fridkin M, Katz-Hillel A, Feldman M, Berke G, Eisenbach L: Regression of established murine carcinoma metastases following vaccination with tumour-associated antigen peptides. Nat Med 1995, 1:1179–1183. [PubMed] [Google Scholar]
This was among the first studies that showed the utility of a neoantigen-based tumor vaccine in protecting mice from metastases.
87. ●●. Matsushita H, Vesely MD, Koboldt DC, Rickert CG, Uppaluri R, Magrini VJ, Arthur CD, White JM, Chen YS, Shea LK et al.: Cancer exome analysis reveals a T-cell-dependent mechanism of cancer immunoediting. Nature 2012, 482:400–404. [PMC free article] [PubMed] [Google Scholar]
This study used the Immune Epitope Database tool to predict the neoantigen that led to tumor rejection in a murine model of sarcoma. It laid the foundation for a similar analysis in human melanoma tumors (Snyder A, Makarov V, Merghoub T, Yuan J et al, below).
88. ●●. Gubin MM, Zhang X, Schuster H, Caron E, Ward JP, Noguchi T, Ivanova Y, Hundal J, Arthur CD, Krebber WJ et al.: Checkpoint blockade cancer immunotherapy targets tumour-specific mutant antigens. Nature 2014, 515:577–581. [PMC free article] [PubMed] [Google Scholar]
This study used a multi-tool prediction method to identify two rejection neoantigens following anti-PD-1 or anti-CTLA-4 treatment of murine sarcomas.
89. ●●. Castle JC, Kreiter S, Diekmann J, Lower M, van de Roemer N, de Graaf J, Selmi A, Diken M, Boegel S, Paret C et al.: Exploiting the mutanome for tumor vaccination. Cancer Res 2012, 72:1081–1091. [PubMed] [Google Scholar]
This work showed that the non-immunogenic murine cell line B16F10 could be made immunogenic by vaccinating with mutant epitopes predicted from whole exome sequencing plus adjuvant.
90. ●. DuPage M, Mazumdar C, Schmidt LM, Cheung AF, Jacks T: Expression of tumour-specific antigens underlies cancer immunoediting. Nature 2012, 482:405–409. [PMC free article] [PubMed] [Google Scholar]
This study identified rejection antigens, but using a genetically engineered authochthonous mouse model of sarcomagenesis rather than a carcinogen-induced model such as in the Schreiber work.
91. ●. Zhou J, Dudley ME, Rosenberg SA, Robbins PF: Persistence of multiple tumor-specific T-cell clones is associated with complete tumor regression in a melanoma patient receiving adoptive cell transfer therapy. J Immunother 2005, 28:53–62. [PMC free article] [PubMed] [Google Scholar]
This study used a cDNA library screen to identify the neoantigens against which anti-tumor TILs were directed in a melanoma patient.
92. ●. Robbins PF, Lu YC, El-Gamil M, Li YF, Gross C, Gartner J, Lin JC, Teer JK, Cliften P, Tycksen E et al.: Mining exomic sequencing data to identify mutated antigens recognized by adoptively transferred tumor-reactive T cells. Nat Med 2013, 19:747–752. [PMC free article] [PubMed] [Google Scholar]
This study used exome sequencing followed by neoantigen prediction by NetMHCpan and peptide stimulation of autologous T cells to identify tumor neoantigens in melanoma patients treated with TIL therapy.
93. ●●. van Rooij N, van Buuren MM, Philips D, Velds A, Toebes M, Heemskerk B, van Dijk LJ, Behjati S, Hilkmann H, El Atmioui D et al.: Tumor exome analysis reveals neoantigen-specific T-cell reactivity in an ipilimumab-responsive melanoma. J Clin Oncol 2013, 31 e439–442. [PMC free article] [PubMed] [Google Scholar]
This was the first study of tumor neoantigens in a patient treated with immune blockade therapy, in this case ipilimumab.
94. ●●. Snyder A, Makarov V, Merghoub T, Yuan J, Zaretsky JM, Desrichard A, Walsh LA, Postow MA, Wong P, Ho TS et al.: Genetic basis for clinical response to CTLA-4 blockade in melanoma. N Engl J Med 2014, 371:2189–2199. [PMC free article] [PubMed] [Google Scholar]
This study elucidated the neoantigen patterns underlying clinical response to anti-CTLA4 therapy. This was the first multi-patient study of the mutational features influencing response to immune checkpoint blockade.
95. ●●. Birnbaum ME, Mendoza JL, Sethi DK, Dong S, Glanville J, Dobbins J, Ozkan E, Davis MM, Wucherpfennig KW, Garcia KC: Deconstructing the peptide-MHC specificity of T cell recognition. Cell 2014, 157:1073–1087. [PMC free article] [PubMed] [Google Scholar]
This study illustrated the ability of the T cell clones under study to recognize and proliferate in response to a diversity of peptides, including those with limited homology of the centrally-located amino acids.
96. ●●. Rajasagi M, Shukla SA, Fritsch EF, Keskin DB, DeLuca D, Carmona E, Zhang W, Sougnez C, Cibulskis K, Sidney J et al.: Systematic identification of personal tumor-specific neoantigens in chronic lymphocytic leukemia. Blood 2014, 124:453–462. [PMC free article] [PubMed] [Google Scholar]
This study demonstrates that the tumor neoantigen association with response also applies to a liquid tumor, CLL. It also discusses predicted neoantigens across in TCGA data.
97. Zhang H, Lund O, Nielsen M: The PickPocket method for predicting binding specificities for receptors based on receptor pocket similarities: application to MHC-peptide binding. Bioinformatics 2009, 25:1293–1299. [PMC free article] [PubMed] [Google Scholar]
98. Zhang GL, DeLuca DS, Keskin DB, Chitkushev L, Zlateva T, Lund O, Reinherz EL, Brusic V: MULTIPRED2: a computational system for large-scale identification of peptides predicted to bind to HLA supertypes and alleles. J Immunol Methods 2011, 374:53–61. [PMC free article] [PubMed] [Google Scholar]
99. Bordner AJ, Mittelmann HD: MultiRTA: a simple yet reliable method for predicting peptide binding affinities for multiple Class II MHC allotypes. BMC Bioinform 2010, 11:482. [PMC free article] [PubMed] [Google Scholar]
100. McMurry JA, Kimball S, Lee JH, Rivera D, Martin W, Weiner DB, Kutzler M, Sherman DR, Kornfeld H, De Groot AS: Epitope-driven TB vaccine development: a streamlined approach using immuno-informatics. ELISpot assays, and HLA transgenic mice. Curr Mol Med 2007, 7:351–368. [PubMed] [Google Scholar]
101. De Groot AS, Bosma A, Chinai N, Frost J, Jesdale BM, Gonzalez MA, Martin W, Saint-Aubin C: From genome to vaccine: in silico predictions, ex vivo verification. Vaccine 2001, 19:4385–4395. [PubMed] [Google Scholar]
102. Stranzl T, Larsen MV, Lundegaard C, Nielsen M: NetCTLpan: pan-specific MHC Class I pathway epitope predictions. Immunogenetics 2010, 62:357–368. [PMC free article] [PubMed] [Google Scholar]
103. Cantor JR, Yoo TH, Dixit A, Iverson BL, Forsthuber TG, Georgiou G: Therapeutic enzyme deimmunization by combinatorial T-cell epitope removal using neutral drift. Proc Natl Acad Sci U S A 2011, 108:1272–1277. [PMC free article] [PubMed] [Google Scholar]