Europe PMC

This website requires cookies, and the limited processing of your personal data in order to function. By using the site you are agreeing to this as outlined in our privacy notice and cookie policy.

Abstract 


Neoantigens are antigens encoded by tumor-specific mutated genes. Studies in the past few years have suggested a key role for neoantigens in cancer immunotherapy. Here we review the discoveries of neoantigens in the past two decades and the current advances in neoantigen identification. We also discuss the potential benefits and obstacles to the development of effective cancer immunotherapies targeting neoantigens.

Free full text 


Logo of nihpaLink to Publisher's site
Semin Immunol. Author manuscript; available in PMC 2017 Feb 1.
Published in final edited form as:
PMCID: PMC4862880
NIHMSID: NIHMS737023
PMID: 26653770

Cancer immunotherapy targeting neoantigens

Abstract

Neoantigens are antigens that are encoded by tumor-specific mutated genes. Studies in the past few years have suggested a key role for neoantigens in cancer immunotherapy. Here we review the discoveries of neoantigens in the past two decades and the current advances in neoantigen identification. We also discuss the potential benefits and obstacles to the development of effective cancer immunotherapies targeting neoantigens.

Keywords: cancer immunotherapy, tumor immunology, neoantigen, mutation

1. Introduction

The foundation of immunology is based on self-nonself discrimination [1]. Most of the pathogens contain molecular signatures that can be recognized by the host and trigger immune responses [2]. Unlike pathogens, these molecular signatures are not generally expressed by tumor cells, making them more difficult to be distinguished from normal cells. However, T cells can recognize tumor antigens expressed by tumor cells. A class of tumor antigens, named tumor-associated antigens, is expressed in some normal tissues at low levels but is over-expressed in malignant cells. Many of the tumor-associated antigens have been identified as the targets of tumor-reactive T cells, isolated from tumor infiltrating lymphocytes (TILs), from draining lymph nodes or from peripheral blood [3]. However, expression of these antigens in normal cells can trigger central and peripheral tolerance mechanisms that lead to the selection of T cells with low-affinity T cell receptors (TCR). Conversely, attempts to target tumor-associated antigens with high-affinity TCRs can lead to severe toxicities due to normal tissue destruction [4, 5].

Another class of tumor antigens is tumor-specific neoantigens, which arise via mutations that alter amino acid coding sequences (non-synonymous somatic mutations). Some of these mutated peptides can be expressed, processed and presented on the cell surface, and subsequently recognized by T cells. Because normal tissues do not possess these somatic mutations, neoantigen-specific T cells are not subject to central and peripheral tolerance, and also lack the ability to induce normal tissue destruction. As a result, neoantigens appear to represent ideal targets for T cell-based cancer immunotherapy.

2. Approaches to identify T cell neoantigens

2.1. Classical approaches

Some of the initial attempts were focused on common shared mutations that have been well-characterized. Short peptides were synthesized based on the sequences of mutated BRAF [6, 7], KRAS [8-11] and p53 [12] . T cells from patients’ or healthy donors’ blood were stimulated several times by peptide-pulsed target cells, and expanded T cells were studied for their ability to kill tumors carrying these mutations. Alternatively, cells with overexpressed mutated cDNA, such as mutated NRAS cDNA, could serve as target cells to detect and isolate neoantigen-reactive T cells [13].

However, the majority of the neoantigen-reactive T cells recognized unique mutations not shared between cancer patients. Most of the unique neoantigens were identified by cDNA library screening in the past two decades. In this approach, cDNA library and MHC molecules were over-expressed in cell lines, and then co-cultured with T cells to identify antigens that could induce the T cell activation, measured by cytokine secretion or 4-1BB up-regulation. Table 1 is the list of published neoantigens identified by this approach.

Table 1

Human neoantigens discovered by classical approaches.

Cancer typeYearMutated gene nameApproachSource of T cellsReference
Melanoma1995CDK4cDNA libraryPBL[14]
Melanoma1995MUM1cDNA libraryPBL[57]
Melanoma1996CTNNB1cDNA libraryTIL[15]
Melanoma1999CDC27cDNA libraryTIL[58]
Melanoma1999TRAPPC1cDNA libraryPBL[59]
Melanoma1999TPIChromatographic purificationTIL[60]
Melanoma2000ASCC3cDNA libraryPBL[61]
Melanoma2001HHATcDNA libraryTIL[62]
Melanoma2002FN1cDNA libraryTIL[63]
Melanoma2002OS-9cDNA libraryPBL[64]
Melanoma2003PTPRKcDNA libraryTIL[65]
Melanoma2004CDKN2A** , HLA-A11cDNA libraryTIL[18]
Melanoma2005GAS7, GAPDHcDNA libraryTIL[66]
Melanoma2005SIRT2, GPNMB, SNRP116,
RBAF600, SNRPD1
cDNA libraryPBL[67]
Melanoma2005Prdx5cDNA libraryPBL[68]
Melanoma2011CLPPcDNA libraryTIL[69]
Melanoma2013PPP1R3BcDNA libraryTIL[46]
Lung cancer1998EF2Chromatographic purificationPBL[20]
Lung cancer2001ACTN4cDNA libraryTIL[70]
Lung cancer2001ME1cDNA libraryPBL[71]
Lung Cancer2006NF-YCcDNA librarydraining lymph node[72]
Renal cancer1996HLA-A2cDNA libraryPBL[17]
Renal cancer1999HSP70-2cDNA libraryTIL[73]
Renal cancer2005KIAA1440cDNA libraryPBL[74]
Head and neck
squamous cell carcinoma
1997CASP8cDNA libraryPBL[16]
**frame-shift mutation

TIL: tumor infiltrating lymphocytes; PBL: peripheral blood lymphocytes.

Neoantigens have been identified predominantly in melanoma, likely due to the relatively high mutation rate in this tumor type. Nonetheless, neoepitopes have also been identified in multiple tumor types including lung and renal cancers. The majority of neoantigens were encoded by point-mutated gene products, although frameshift deletion and insertions have also been found to generate neoepitopes. Some mutated gene products recognized by T cells appear to be driver mutation products and play a role in tumorigenesis. These include CDK4, β-catenin (CTNNB1) and Caspase-8 (CASP8) proteins [14-16]. Interestingly, HLA-A2 and A11 containing point-mutations have also been identified as tumor-specific T cell antigens, suggesting that somatic mutations in HLA molecules can be a source of neoantigens [17-19].

In another approach, a mutated (EF2) and non-mutated (gp100:154) T cell epitopes have been identified by using mass spectrometry to sequence peptides that were eluted from HLA molecules isolated from the surface of tumor cells [20]. This approach has been limited by the sensitivity of these methods. Nevertheless, recent advances in the sensitivity and throughput of these techniques may facilitate the application of this approach to identifying tumor antigens.

2.2. Approaches utilizing next-generation sequencing techniques

Although the classical cDNA library screening approach led to the discovery of multiple neoantigens, this approach is labor-intensive and low-throughput. In addition, some large transcripts, GC-rich transcripts and low-expression transcripts cannot be cloned easily, leading to the failure of identifying some mutated antigens. Recently, several investigators have taken advantage of whole-exome sequencing technologies to identify non-synonymous mutations in tumors. These mutated genes were subjected to in silico analysis to predict potential high-affinity epitopes that bind to MHC molecules [21-23]. Additional filters could be applied to eliminate (1) epitopes predicted to be poorly processed by the immunoproteasome and (2) epitopes with lower binding affinity than the corresponding wild-type sequences [24]. Candidate mutated peptides are synthesized and screened to identify T cell neoantigens. This approach can be very efficient, and can identify neoantigens where previous cDNA library screening failed to discover [25].

This peptide-based screening approach, while effective in identifying many epitopes, may nevertheless be limited by the accuracy of MHC/HLA-binding prediction algorithms, which have not been thoroughly examined for MHC class II alleles and infrequent HLA alleles. In addition, the expression of potential T cell epitopes on the cell surface is influenced by a variety of mechanisms, especially the expression of multiple forms of the proteasome, which limit the number of peptides that are truly processed and presented [26, 27]. An alternative approach is pulsing antigen presenting cells with relatively long synthetic peptides that encompass minimal T cell epitopes [28]. In a recent report, nonsynonymous mutated epitopes were identified in three melanoma lesions by evaluating the response of CD4+ TIL to autologous B cells that were pulsed with 31 amino-acid long peptides encompassing individual mutations. Use of this approach resulted in the identification of mutated CIRH1A, GART, ASAP1, RND3, TNIK, RPS12, ZC3H18 and LEMD2 T cell epitopes. Furthermore, in a recent report, a peptide screening was carried out based on the combination of two peptide libraries: (1) 15-mer overlapping long-peptides (2) peptides based on MHC-binding prediction. This screening led to the identification of mutated HSDL1-reactive T cells isolated from an ovarian tumor [29].

To solve all the potential issues mentioned above, we have developed a tandem minigene screening approach [30]. A tandem minigene construct comprises 6 to 24 minigenes that encode polypeptides containing a mutated amino acid residue flanked on their N- and C-termini by 12 amino acids. Tandem minigene constructs are synthesized and used to transfect autologous APCs or cell lines co-expressing autologous HLA molecules. Using this approach, mutated KIF2C and POLA2 epitopes were identified in two melanoma patients [30]. In addition, a mutated ERBB2IP epitope was identified in a patient with cholangiocarcinoma [31]. Recent studies using this approach have led to the identification of mutated antigens express on gastrointestinal, breast and ovarian cancers [32] (and unpublished data). Notably, the neoantigen reactivity could be identified from TILs isolated from patients with cholangiocarcinoma or gastrointestinal cancer, which has a relatively low number of mutations.

A recent approach combined whole-exome/transcriptome sequencing analysis, MHC binding prediction, as well as mass spectrometric technique to detect peptides eluted from HLA molecules [33]. Interestingly, only a small fraction of predicted high-binding peptides were confirmed by mass spectrometry. The relative small number of mutated peptides identified by mass spectrometry might be due to the sensitivity of the peptide purification and mass spectrometry, but it could also suggest that natural antigen process and presentation in cells could be very inefficient. Among 7 neo-epitopes identified by this approach, mutated Adpgk, Reps1 and Dpagt1 epitopes were confirmed to be immunogenic [33].

3. The correlation between neoantigens and tumor regressions

It has been established that T cells recognizing non-mutated tumor antigens could induce tumor regression [34]. This has been demonstrated by clinical trials involving adoptive transfer of T cells targeting HLA-A*0201-restricted NY-ESO-1. Objective responses were observed in 61% of patients with synovial cell sarcoma, 55% of patients with melanoma and 80% of patients with myeloma [35-37]. However, it remains unclear whether T cells recognizing neoantigens can also induce tumor regression in human.

In the past few years, there have been significant advances in checkpoint blockade cancer immunotherapy. Tumor regressions were observed in a portion of cancer patients who underwent CTLA-4, PD-1 or PD-L1 blockade clinical trials [38]. Currently, the widely acceptable hypothesis is that tumors with more mutations likely generate more neoepitopes, which can be recognized by tumor infiltrating T cells. Checkpoint blocking antibodies reactivate these T cells in vivo and induce tumor regressions. As a result, cancers with high mutation rates, such as melanoma and lung cancer, are more susceptible to checkpoint blockade therapies. (One notable exception for this hypothesis is renal cell carcinoma, which is susceptible to checkpoint blockade therapies but with low mutation load). A recent study in a murine model has provided some evidence for this hypothesis. In an anti-PD-1 and/or anti-CTLA-4 immunotherapy model, two dominant neoantigens asparagine-linked glycosylation 8 (Alg8) (A506T) and laminin alpha subunit 4 (Lama4) (G1254V) were identified from a sarcoma cell line d42m1-T3. Mutated Lama4 and Alg8 long peptide vaccines could induce tumor rejection comparable to checkpoint blockade immunotherapy. In addition, the checkpoint blockade immunotherapy increased the number and enhanced the activity of neoantigen-specific CD8+ T cells [24]. These results suggested that neoantigens and neoantigen-specific T cells were strongly associated with tumor regressions after checkpoint blockade therapy.

In human, recent studies have established the correlation between the number of mutations/neoantigens and clinical outcomes. In a PD-1 blockade clinical trial comparing colorectal cancer patients with or without mismatch-repair deficiency, prolonged progression-free survival was associated with high somatic mutations, which were found in tumors with mismatch repair-deficiency [39]. Other studies utilized in silico analyses to predict potential high-affinity neoepitopes for calculating the numbers of neoantigens. In two anti-CTLA-4 melanoma immunotherapy studies, the number of neoantigens was significantly associated with clinical benefits after CTLA-4 blockade therapies [40, 41]. However, it’s unclear whether the presence of “tetrapeptide” signatures within predicted T cell epitopes could be employed to predict the clinical outcomes for the anti-CTLA-4 immunotherapy [40, 41]. Similar to CTLA-4 blockade studies, higher neoantigen burden was correlated with clinical benefit and progression-free survival in PD-1 blockade immunotherapy for patients with non-small cell lung cancer [42]. These studies suggested that the number of neoantigens was positively associated with clinical benefit after immune checkpoint blockade therapies. Lastly, adoptive transfer of mixed TILs could induce tumor regressions in melanoma patients [43]. In a recent study from our group, objective tumor regressions in three melanoma patients were associated with adoptive transferring of TILs all recognizing neoantigens [25]. In a subsequent study, two TIL products were associated with complete tumor regressions observed in two melanoma patients, and each TIL could recognize one unique neoantigen [30]. Taken together, these correlative studies suggested that neoantigen-reactive T cells were likely the dominant player inducing tumor regressions in patients.

4. Current evidence of cancer immunotherapy targeting neoantigens

The first approach to target neoantigens is via cancer vaccine. In an initial mouse study, candidate mutated epitopes were identified by whole-exome sequencing of the B16F10 murine melanoma. Fifty selected mutation-coding long peptides were injected into mice to elicit immune responses, and 11 out of 50 peptides induced immune responses preferentially recognizing the mutated epitopes. Among these 11 peptides, mutated Kif18b (K739N) was found to be the dominant mutated antigens, and mice immunized with mutated Kif18b peptide could slow tumor growth and improve survival [22]. In another study, MC-38 tumor-bearing mice vaccinated with mutated peptides (Adpgk, Reps1 and Dpagt1) showed sustainable inhibition of tumor growth [33]. Recently, a synthetic RNA “pentatope” vaccine was developed. Each pentatope contains five 27-mer minigenes with the mutated amino acids in the center, and each fused to each other by 10-mer glycine-serine linker (pentatopes). The immunization of this RNA pentatope conferred disease control and survival benefit in a murine CT26 tumor model. Notably, mutated MHC class II epitopes were more immunogenic than class I epitopes in this study [44]. A recent study has brought a neoantigen cancer vaccine to a clinical trial in which whole-exome sequencing was carried out to identify somatic mutations in tumors from 3 patients with melanoma. Candidate HLA-A*02:01 epitopes containing residues arising from mutations were initially filtered using an HLA binding prediction algorithm, and then evaluated using competitive binding assays. Three patients were vaccinated with autologous dendritic cells that had been pulsed with the top 7 highest binding peptides identified from each tumor. The breadth and diversity of neoantigen-specific T cells were increased in all 3 patients after the vaccination. In addition, the results indicated that T cells generated by vaccination with 7 out of the 21 epitopes could recognize target cells transfected with the corresponding tandem minigene constructs, indicating that these neoantigens could be endogenously processed and presented [45]. While these results demonstrated that neoantigen cancer vaccines could elicit neoantigen-specific T cells in cancer patients, no clinical benefits to these patients were shown in this study. It remains unclear whether current cancer vaccine therapies are potent enough to provide therapeutic benefits in cancer patients with bulky disease.

The second approach to target neoantigens is by transferring neoantigen-specific T cells directly into host. Although so far no evidence in mouse models has shown that such T cells can induce tumor regressions in vivo, some evidence has linked neoantigen-reactive T cells to tumor regressions in human. A clue came from a report studying a unique melanoma TIL product, with no reactivity against non-mutated antigens. Instead, ~50% of this TIL product showed predominant reactivity against HLA-A*01-restricted mutated PPP1R3B. Following adoptive transfer of this TIL product, the patient experienced a complete response ongoing beyond 10 years. In addition, mutated PPP1R3B-reactive T cells could be still detected in the patient’s peripheral blood 5 years after the immunotherapy [46]. The most direct clinical evidence came from an adoptive cell transfer immunotherapy using nearly pure neoantigen-reactive T cells. A patient with metastatic cholangiocarcinoma was treated with one billion mutated ERBB2IP-reactive CD4+ T cells, and she experienced a partial response ongoing for more than 2 years since treatment [31]. This suggested that neoantigen-reactive T cells could induce long-term tumor regressions in cancer patients, and it also brought up the possibility of applying immunotherapy to a variety of cancer types other than melanoma.

5. Conclusions and future perspectives

Based on current knowledge, neoantigens are ideal targets for cancer immunotherapy. Because neoantigens are specifically expressed in the tumor, it is less likely to induce tolerance and nearly impossible to induce normal tissue toxicity. However, it remains challenging to reduce the cost of such highly personalized therapy.

Many outstanding questions remain unanswered (Table 2). How can we apply to cancer immunotherapy to a variety of cancer types by targeting neoantigens? Does the average number of neoantigens discovered per tumor correlate with the mutation rate in each cancer type? If yes, how can we apply immunotherapy to cancers with low mutation rates? Does the tumor microenvironments in different cancer types influence the efficacy of immunotherapy? Can the fitness or the number of T cells overcome the potentially harsh tumor microenvironment in different cancer types?

Table 2

Outstanding questions for cancer immunotherapy targeting neoantigens.

1.How does this approach overcome the heterogeneity of tumor?
2.Does the number of neoantigens correlate with the mutation rate in each tumor
specimen?
3.Can this approach apply to a variety types of cancers, especially cancers with low
mutation rate?
4.Can neoantigens induce tolerance? Can Treg specifically recognize neoantigens and
induce tolerance?
5.Are the number and fitness of neoantigen-specific T cells important for the efficacy of
cancer immunotherapy?

One major concern is the heterogeneity of tumor. Neoantigens may be expressed in some tumor cells, but not all tumor cells in an individual patient, leading to tumor escape from immunotherapy. Conversely, we and others have observed complete tumor regressions in dozens of patients after immunotherapy. Some potential approaches may address this concern. One is to target multiple neoantigens at the same time, so all tumor cells expressing at least one neoantigen can be destroyed. Another approach is to target a single neoantigen, which is ideally expressed in all tumor cells within a patient.

Targeting driver mutations can be an effective strategy [30]. Aggressive tumor cells are actively expressing driver mutations, which are essential for carcinogenesis or metastasis. Although some tumor cells may not express driver mutations and may escape from immunotherapy, those tumor cells likely lose the metastatic potential and cannot grow aggressively. Nonetheless, defining driver mutations versus passenger mutations remains a challenging issue. The most definitive way to identify driver mutations is based on the functional studies found in the literature. From the neoantigens reviewed in this article, CDK4, β-catenin and Caspase-8 are likely driver mutations. The majority of neoantigens are likely random mutations (passenger mutations) recognized by T cells, but it is also possible that some of the mutations are not well-characterized. The Cancer Genome Atlas attempted to address this issue by sequencing and analyzing hundreds of tumor specimens. The recurrent mutations were identified as the potential driver mutations [47-49].

Recent checkpoint blockade immunotherapies have shown some efficacies in lung cancer, bladder cancer and renal cancer [50-53]. Based on the assumption that neoantigen-reactive T cells are responsible for tumor regressions in checkpoint blockade immunotherapies, directly transferring neoantigen-reactive T cells into cancer patients may achieve a much higher response rate. Additionally, neoantigen cancer vaccines may enhance the strength and persistence of T cells, and ultimately improve the efficacy of immunotherapy. With the recent advances in technology, we can identify neoantigens and isolate neoantigen-specific TCRs from individual patients in a timely and cost effective manner [54-56] (and unpublished data). It is possible that patients with a wide variety of cancer types, including cancers with very few mutations, can receive the proposed cancer immunotherapy targeting neoantigens (Figure 1).

An external file that holds a picture, illustration, etc.
Object name is nihms-737023-f0001.jpg

Cancer immunotherapy targeting neoantigens by the adoptive transfer of genetically-modified T cells. In this proposed study, a tumor will be surgically resected from a cancer patient. Part of the tumor specimen will be subjected to whole-exome sequencing to identify nonsynonymous mutations. The rest of the tumor specimen will be used to grow TILs. Potential neoantigens will be identified by minigene or peptide screening approach described in the text. Neoantigen-specific TCRs will be isolated and introduced into patient’s peripheral blood T cells. These genetically-modified T cells will be adoptively transferred back to the patient to fight cancer. A neoantigen cancer vaccines will be injected to enhance the strength and persistence of T cells.

Highlights

We review the past and present approaches to identify neoantigens.

We emphasize the importance of neoantigens in cancer immunotherapy.

We discuss the benefits and obstacles to develop cancer immunotherapies targeting neoantigens.

Acknowledgements

The authors thank Todd Prickett and Lucas McDuffie for suggestions and discussions. This work was supported by the Intramural Research Program of National Cancer Institute.

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

[1] Burnet FM. The clonal selection theory of acquired immunity. Cambridge University Press; London: 1959. [Google Scholar]
[2] Janeway CA, Jr., Medzhitov R. Innate immune recognition. Annu Rev Immunol. 2002;20:197–216. [Abstract] [Google Scholar]
[3] Cheever MA, Allison JP, Ferris AS, Finn OJ, Hastings BM, Hecht TT, et al. The prioritization of cancer antigens: a national cancer institute pilot project for the acceleration of translational research. Clin Cancer Res. 2009;15:5323–5337. [Europe PMC free article] [Abstract] [Google Scholar]
[4] Johnson LA, Morgan RA, Dudley ME, Cassard L, Yang JC, Hughes MS, et al. Gene therapy with human and mouse T-cell receptors mediates cancer regression and targets normal tissues expressing cognate antigen. Blood. 2009;114:535–546. [Europe PMC free article] [Abstract] [Google Scholar]
[5] Parkhurst MR, Yang JC, Langan RC, Dudley ME, Nathan DA, Feldman SA, et al. T cells targeting carcinoembryonic antigen can mediate regression of metastatic colorectal cancer but induce severe transient colitis. Mol Ther. 2011;19:620–626. [Europe PMC free article] [Abstract] [Google Scholar]
[6] Sharkey MS, Lizee G, Gonzales MI, Patel S, Topalian SL. CD4(+) T-cell recognition of mutated B-RAF in melanoma patients harboring the V599E mutation. Cancer research. 2004;64:1595–1599. [Abstract] [Google Scholar]
[7] Somasundaram R, Swoboda R, Caputo L, Otvos L, Weber B, Volpe P, et al. Human leukocyte antigen-A2-restricted CTL responses to mutated BRAF peptides in melanoma patients. Cancer research. 2006;66:3287–3293. [Abstract] [Google Scholar]
[8] Fossum B, Gedde-Dahl T, 3rd, Hansen T, Eriksen JA, Thorsby E, Gaudernack G. Overlapping epitopes encompassing a point mutation (12 Gly-->Arg) in p21 ras can be recognized by HLA-DR, -DP and -DQ restricted T cells. European journal of immunology. 1993;23:2687–2691. [Abstract] [Google Scholar]
[9] Bergmann-Leitner ES, Kantor JA, Shupert WL, Schlom J, Abrams SI. Identification of a human CD8+ T lymphocyte neo-epitope created by a ras codon 12 mutation which is restricted by the HLA-A2 allele. Cellular immunology. 1998;187:103–116. [Abstract] [Google Scholar]
[10] Kubuschok B, Schmits R, Hartmann F, Cochlovius C, Breit R, Konig J, et al. Use of spontaneous Epstein-Barr virus-lymphoblastoid cell lines genetically modified to express tumor antigen as cancer vaccines: mutated p21 ras oncogene in pancreatic carcinoma as a model. Human gene therapy. 2002;13:815–827. [Abstract] [Google Scholar]
[11] Shono Y, Tanimura H, Iwahashi M, Tsunoda T, Tani M, Tanaka H, et al. Specific T-cell immunity against Ki-ras peptides in patients with pancreatic and colorectal cancers. British journal of cancer. 2003;88:530–536. [Europe PMC free article] [Abstract] [Google Scholar]
[12] Ichiki Y, Takenoyama M, Mizukami M, So T, Sugaya M, Yasuda M, et al. Simultaneous cellular and humoral immune response against mutated p53 in a patient with lung cancer. Journal of immunology. 2004;172:4844–4850. [Abstract] [Google Scholar]
[13] Linard B, Bezieau S, Benlalam H, Labarriere N, Guilloux Y, Diez E, et al. A ras-mutated peptide targeted by CTL infiltrating a human melanoma lesion. Journal of immunology. 2002;168:4802–4808. [Abstract] [Google Scholar]
[14] Wolfel T, Hauer M, Schneider J, Serrano M, Wolfel C, Klehmann-Hieb E, et al. A p16INK4a-insensitive CDK4 mutant targeted by cytolytic T lymphocytes in a human melanoma. Science. 1995;269:1281–1284. [Abstract] [Google Scholar]
[15] Robbins PF, El-Gamil M, Li YF, Kawakami Y, Loftus D, Appella E, et al. A mutated beta-catenin gene encodes a melanoma-specific antigen recognized by tumor infiltrating lymphocytes. J Exp Med. 1996;183:1185–1192. [Europe PMC free article] [Abstract] [Google Scholar]
[16] Mandruzzato S, Brasseur F, Andry G, Boon T, van der Bruggen P. A CASP-8 mutation recognized by cytolytic T lymphocytes on a human head and neck carcinoma. The Journal of experimental medicine. 1997;186:785–793. [Europe PMC free article] [Abstract] [Google Scholar]
[17] Brandle D, Brasseur F, Weynants P, Boon T, Van den Eynde B. A mutated HLA-A2 molecule recognized by autologous cytotoxic T lymphocytes on a human renal cell carcinoma. The Journal of experimental medicine. 1996;183:2501–2508. [Europe PMC free article] [Abstract] [Google Scholar]
[18] Huang J, El-Gamil M, Dudley ME, Li YF, Rosenberg SA, Robbins PF. T cells associated with tumor regression recognize frameshifted products of the CDKN2A tumor suppressor gene locus and a mutated HLA class I gene product. J Immunol. 2004;172:6057–6064. [Europe PMC free article] [Abstract] [Google Scholar]
[19] Shukla SA, Rooney MS, Rajasagi M, Tiao G, Dixon PM, Lawrence MS, et al. Comprehensive analysis of cancer-associated somatic mutations in class I HLA genes. Nat Biotechnol. 2015 [Europe PMC free article] [Abstract] [Google Scholar]
[20] Hogan KT, Eisinger DP, Cupp SB, 3rd, Lekstrom KJ, Deacon DD, Shabanowitz J, et al. The peptide recognized by HLA-A68.2-restricted, squamous cell carcinoma of the lung-specific cytotoxic T lymphocytes is derived from a mutated elongation factor 2 gene. Cancer research. 1998;58:5144–5150. [Abstract] [Google Scholar]
[21] Matsushita H, Vesely MD, Koboldt DC, Rickert CG, Uppaluri R, Magrini VJ, et al. Cancer exome analysis reveals a T-cell-dependent mechanism of cancer immunoediting. Nature. 2012;482:400–404. [Europe PMC free article] [Abstract] [Google Scholar]
[22] Castle JC, Kreiter S, Diekmann J, Lower M, van de Roemer N, de Graaf J, et al. Exploiting the mutanome for tumor vaccination. Cancer research. 2012;72:1081–1091. [Abstract] [Google Scholar]
[23] Cai A, Keskin DB, DeLuca DS, Alonso A, Zhang W, Zhang GL, et al. Mutated BCR-ABL generates immunogenic T-cell epitopes in CML patients. Clinical cancer research : an official journal of the American Association for Cancer Research. 2012;18:5761–5772. [Europe PMC free article] [Abstract] [Google Scholar]
[24] Gubin MM, Zhang X, Schuster H, Caron E, Ward JP, Noguchi T, et al. Checkpoint blockade cancer immunotherapy targets tumour-specific mutant antigens. Nature. 2014;515:577–581. [Europe PMC free article] [Abstract] [Google Scholar]
[25] Robbins PF, Lu YC, El-Gamil M, Li YF, Gross C, Gartner J, et al. Mining exomic sequencing data to identify mutated antigens recognized by adoptively transferred tumor-reactive T cells. Nature medicine. 2013;19:747–752. [Europe PMC free article] [Abstract] [Google Scholar]
[26] Valmori D, Lienard D, Waanders G, Rimoldi D, Cerottini JC, Romero P. Analysis of MAGE-3-specific cytolytic T lymphocytes in human leukocyte antigen-A2 melanoma patients. Cancer research. 1997;57:735–741. [Abstract] [Google Scholar]
[27] Morel S, Levy F, Burlet-Schiltz O, Brasseur F, Probst-Kepper M, Peitrequin AL, et al. Processing of some antigens by the standard proteasome but not by the immunoproteasome results in poor presentation by dendritic cells. Immunity. 2000;12:107–117. [Abstract] [Google Scholar]
[28] Zwaveling S, Ferreira Mota SC, Nouta J, Johnson M, Lipford GB, Offringa R, et al. Established human papillomavirus type 16-expressing tumors are effectively eradicated following vaccination with long peptides. J Immunol. 2002;169:350–358. [Abstract] [Google Scholar]
[29] Wick DA, Webb JR, Nielsen JS, Martin SD, Kroeger DR, Milne K, et al. Surveillance of the tumor mutanome by T cells during progression from primary to recurrent ovarian cancer. Clinical cancer research : an official journal of the American Association for Cancer Research. 2014;20:1125–1134. [Abstract] [Google Scholar]
[30] Lu YC, Yao X, Crystal JS, Li YF, El-Gamil M, Gross C, et al. Efficient identification of mutated cancer antigens recognized by T cells associated with durable tumor regressions. Clin Cancer Res. 2014;20:3401–3410. [Europe PMC free article] [Abstract] [Google Scholar]
[31] Tran E, Turcotte S, Gros A, Robbins PF, Lu YC, Dudley ME, et al. Cancer immunotherapy based on mutation-specific CD4+ T cells in a patient with epithelial cancer. Science. 2014;344:641–645. [Europe PMC free article] [Abstract] [Google Scholar]
[32] Tran E, Ahmadzadeh M, Lu YC, Gros A, Turcotte S, Robbins PF, et al. Immunogenicity of somatic mutations in human gastrointestinal cancers. Science. 2015 [Europe PMC free article] [Abstract] [Google Scholar]
[33] Yadav M, Jhunjhunwala S, Phung QT, Lupardus P, Tanguay J, Bumbaca S, et al. Predicting immunogenic tumour mutations by combining mass spectrometry and exome sequencing. Nature. 2014;515:572–576. [Abstract] [Google Scholar]
[34] Rosenberg SA, Restifo NP. Adoptive cell transfer as personalized immunotherapy for human cancer. Science. 2015;348:62–68. [Europe PMC free article] [Abstract] [Google Scholar]
[35] Robbins PF, Kassim SH, Tran TL, Crystal JS, Morgan RA, Feldman SA, et al. A pilot trial using lymphocytes genetically engineered with an NY-ESO-1-reactive T-cell receptor: long-term follow-up and correlates with response. Clin Cancer Res. 2015;21:1019–1027. [Europe PMC free article] [Abstract] [Google Scholar]
[36] Robbins PF, Morgan RA, Feldman SA, Yang JC, Sherry RM, Dudley ME, et al. Tumor regression in patients with metastatic synovial cell sarcoma and melanoma using genetically engineered lymphocytes reactive with NY-ESO-1. J Clin Oncol. 2011;29:917–924. [Europe PMC free article] [Abstract] [Google Scholar]
[37] Rapoport AP, Stadtmauer EA, Binder-Scholl GK, Goloubeva O, Vogl DT, Lacey SF, et al. NY-ESO-1-specific TCR-engineered T cells mediate sustained antigen-specific antitumor effects in myeloma. Nat Med. 2015;21:914–921. [Europe PMC free article] [Abstract] [Google Scholar]
[38] Nguyen LT, Ohashi PS. Clinical blockade of PD1 and LAG3--potential mechanisms of action. Nat Rev Immunol. 2015;15:45–56. [Abstract] [Google Scholar]
[39] Le DT, Uram JN, Wang H, Bartlett BR, Kemberling H, Eyring AD, et al. PD-1 Blockade in Tumors with Mismatch-Repair Deficiency. N Engl J Med. 2015;372:2509–2520. [Europe PMC free article] [Abstract] [Google Scholar]
[40] Snyder A, Makarov V, Merghoub T, Yuan J, Zaretsky JM, Desrichard A, et al. Genetic basis for clinical response to CTLA-4 blockade in melanoma. N Engl J Med. 2014;371:2189–2199. [Europe PMC free article] [Abstract] [Google Scholar]
[41] Van Allen EM, Miao D, Schilling B, Shukla SA, Blank C, Zimmer L, et al. Genomic correlates of response to CTLA-4 blockade in metastatic melanoma. Science. 2015;350:207–211. [Europe PMC free article] [Abstract] [Google Scholar]
[42] Rizvi NA, Hellmann MD, Snyder A, Kvistborg P, Makarov V, Havel JJ, et al. Cancer immunology. Mutational landscape determines sensitivity to PD-1 blockade in non-small cell lung cancer. Science. 2015;348:124–128. [Europe PMC free article] [Abstract] [Google Scholar]
[43] Rosenberg SA, Yang JC, Sherry RM, Kammula US, Hughes MS, Phan GQ, et al. Durable complete responses in heavily pretreated patients with metastatic melanoma using T-cell transfer immunotherapy. Clin Cancer Res. 2011;17:4550–4557. [Europe PMC free article] [Abstract] [Google Scholar]
[44] Kreiter S, Vormehr M, van de Roemer N, Diken M, Lower M, Diekmann J, et al. Mutant MHC class II epitopes drive therapeutic immune responses to cancer. Nature. 2015;520:692–696. [Europe PMC free article] [Abstract] [Google Scholar]
[45] Carreno BM, Magrini V, Becker-Hapak M, Kaabinejadian S, Hundal J, Petti AA, et al. Cancer immunotherapy. A dendritic cell vaccine increases the breadth and diversity of melanoma neoantigen-specific T cells. Science. 2015;348:803–808. [Europe PMC free article] [Abstract] [Google Scholar]
[46] Lu YC, Yao X, Li YF, El-Gamil M, Dudley ME, Yang JC, et al. Mutated PPP1R3B Is Recognized by T Cells Used To Treat a Melanoma Patient Who Experienced a Durable Complete Tumor Regression. J Immunol. 2013;190:6034–6042. [Europe PMC free article] [Abstract] [Google Scholar]
[47] Lawrence MS, Stojanov P, Polak P, Kryukov GV, Cibulskis K, Sivachenko A, et al. Mutational heterogeneity in cancer and the search for new cancer-associated genes. Nature. 2013;499:214–218. [Europe PMC free article] [Abstract] [Google Scholar]
[48] Lawrence MS, Stojanov P, Mermel CH, Robinson JT, Garraway LA, Golub TR, et al. Discovery and saturation analysis of cancer genes across 21 tumour types. Nature. 2014;505:495–501. [Europe PMC free article] [Abstract] [Google Scholar]
[49] Cancer Genome Atlas N Genomic Classification of Cutaneous Melanoma. Cell. 2015;161:1681–1696. [Europe PMC free article] [Abstract] [Google Scholar]
[50] Topalian SL, Hodi FS, Brahmer JR, Gettinger SN, Smith DC, McDermott DF, et al. Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N Engl J Med. 2012;366:2443–2454. [Europe PMC free article] [Abstract] [Google Scholar]
[51] Brahmer JR, Tykodi SS, Chow LQ, Hwu WJ, Topalian SL, Hwu P, et al. Safety and activity of anti-PD-L1 antibody in patients with advanced cancer. N Engl J Med. 2012;366:2455–2465. [Europe PMC free article] [Abstract] [Google Scholar]
[52] Herbst RS, Soria JC, Kowanetz M, Fine GD, Hamid O, Gordon MS, et al. Predictive correlates of response to the anti-PD-L1 antibody MPDL3280A in cancer patients. Nature. 2014;515:563–567. [Europe PMC free article] [Abstract] [Google Scholar]
[53] Powles T, Eder JP, Fine GD, Braiteh FS, Loriot Y, Cruz C, et al. MPDL3280A (anti-PD-L1) treatment leads to clinical activity in metastatic bladder cancer. Nature. 2014;515:558–562. [Abstract] [Google Scholar]
[54] Kobayashi E, Mizukoshi E, Kishi H, Ozawa T, Hamana H, Nagai T, et al. A new cloning and expression system yields and validates TCRs from blood lymphocytes of patients with cancer within 10 days. Nature medicine. 2013;19:1542–1546. [Abstract] [Google Scholar]
[55] Linnemann C, Heemskerk B, Kvistborg P, Kluin RJ, Bolotin DA, Chen X, et al. High-throughput identification of antigen-specific TCRs by TCR gene capture. Nature medicine. 2013;19:1534–1541. [Abstract] [Google Scholar]
[56] Cohen CJ, Gartner JJ, Horovitz-Fried M, Shamalov K, Trebska-McGowan K, Bliskovsky VV, et al. Isolation of neoantigen-specific T cells from tumor and peripheral lymphocytes. J Clin Invest. 2015;125:3981–3991. [Europe PMC free article] [Abstract] [Google Scholar]
[57] Coulie PG, Lehmann F, Lethe B, Herman J, Lurquin C, Andrawiss M, et al. A mutated intron sequence codes for an antigenic peptide recognized by cytolytic T lymphocytes on a human melanoma. Proceedings of the National Academy of Sciences of the United States of America. 1995;92:7976–7980. [Europe PMC free article] [Abstract] [Google Scholar]
[58] Wang RF, Wang X, Atwood AC, Topalian SL, Rosenberg SA. Cloning genes encoding MHC class II-restricted antigens: mutated CDC27 as a tumor antigen. Science. 1999;284:1351–1354. [Abstract] [Google Scholar]
[59] Chiari R, Foury F, De Plaen E, Baurain JF, Thonnard J, Coulie PG. Two antigens recognized by autologous cytolytic T lymphocytes on a melanoma result from a single point mutation in an essential housekeeping gene. Cancer research. 1999;59:5785–5792. [Abstract] [Google Scholar]
[60] Pieper R, Christian RE, Gonzales MI, Nishimura MI, Gupta G, Settlage RE, et al. Biochemical identification of a mutated human melanoma antigen recognized by CD4(+) T cells. J Exp Med. 1999;189:757–766. [Europe PMC free article] [Abstract] [Google Scholar]
[61] Baurain JF, Colau D, van Baren N, Landry C, Martelange V, Vikkula M, et al. High frequency of autologous anti-melanoma CTL directed against an antigen generated by a point mutation in a new helicase gene. Journal of immunology. 2000;164:6057–6066. [Abstract] [Google Scholar]
[62] Kawakami Y, Wang X, Shofuda T, Sumimoto H, Tupesis J, Fitzgerald E, et al. Isolation of a new melanoma antigen, MART-2, containing a mutated epitope recognized by autologous tumor-infiltrating T lymphocytes. Journal of immunology. 2001;166:2871–2877. [Abstract] [Google Scholar]
[63] Wang HY, Zhou J, Zhu K, Riker AI, Marincola FM, Wang RF. Identification of a mutated fibronectin as a tumor antigen recognized by CD4+ T cells: its role in extracellular matrix formation and tumor metastasis. The Journal of experimental medicine. 2002;195:1397–1406. [Europe PMC free article] [Abstract] [Google Scholar]
[64] Vigneron N, Ooms A, Morel S, Degiovanni G, Van Den Eynde BJ. Identification of a new peptide recognized by autologous cytolytic T lymphocytes on a human melanoma. Cancer immunity. 2002;2:9. [Abstract] [Google Scholar]
[65] Novellino L, Renkvist N, Rini F, Mazzocchi A, Rivoltini L, Greco A, et al. Identification of a mutated receptor-like protein tyrosine phosphatase kappa as a novel, class II HLA-restricted melanoma antigen. Journal of immunology. 2003;170:6363–6370. [Abstract] [Google Scholar]
[66] 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. [Europe PMC free article] [Abstract] [Google Scholar]
[67] Lennerz V, Fatho M, Gentilini C, Frye RA, Lifke A, Ferel D, et al. The response of autologous T cells to a human melanoma is dominated by mutated neoantigens. Proceedings of the National Academy of Sciences of the United States of America. 2005;102:16013–16018. [Europe PMC free article] [Abstract] [Google Scholar]
[68] Sensi M, Nicolini G, Zanon M, Colombo C, Molla A, Bersani I, et al. Immunogenicity without immunoselection: a mutant but functional antioxidant enzyme retained in a human metastatic melanoma and targeted by CD8(+) T cells with a memory phenotype. Cancer research. 2005;65:632–640. [Abstract] [Google Scholar]
[69] Corbiere V, Chapiro J, Stroobant V, Ma W, Lurquin C, Lethe B, et al. Antigen spreading contributes to MAGE vaccination-induced regression of melanoma metastases. Cancer research. 2011;71:1253–1262. [Abstract] [Google Scholar]
[70] Echchakir H, Mami-Chouaib F, Vergnon I, Baurain JF, Karanikas V, Chouaib S, et al. A point mutation in the alpha-actinin-4 gene generates an antigenic peptide recognized by autologous cytolytic T lymphocytes on a human lung carcinoma. Cancer research. 2001;61:4078–4083. [Abstract] [Google Scholar]
[71] Karanikas V, Colau D, Baurain JF, Chiari R, Thonnard J, Gutierrez-Roelens I, et al. High frequency of cytolytic T lymphocytes directed against a tumor-specific mutated antigen detectable with HLA tetramers in the blood of a lung carcinoma patient with long survival. Cancer research. 2001;61:3718–3724. [Abstract] [Google Scholar]
[72] Takenoyama M, Baurain JF, Yasuda M, So T, Sugaya M, Hanagiri T, et al. A point mutation in the NFYC gene generates an antigenic peptide recognized by autologous cytolytic T lymphocytes on a human squamous cell lung carcinoma. International journal of cancer Journal international du cancer. 2006;118:1992–1997. [Abstract] [Google Scholar]
[73] Gaudin C, Kremer F, Angevin E, Scott V, Triebel F. A hsp70-2 mutation recognized by CTL on a human renal cell carcinoma. Journal of immunology. 1999;162:1730–1738. [Abstract] [Google Scholar]
[74] Zhou X, Jun DY, Thomas AM, Huang X, Huang LQ, Mautner J, et al. Diverse CD8+ T-cell responses to renal cell carcinoma antigens in patients treated with an autologous granulocyte-macrophage colony-stimulating factor gene-transduced renal tumor cell vaccine. Cancer research. 2005;65:1079–1088. [Abstract] [Google Scholar]

Citations & impact 


Impact metrics

Jump to Citations

Citations of article over time

Alternative metrics

Altmetric item for https://www.altmetric.com/details/4857272
Altmetric
Discover the attention surrounding your research
https://www.altmetric.com/details/4857272

Article citations


Go to all (137) article citations

Funding 


Funders who supported this work.

Intramural NIH HHS (1)