You are on page 1of 19

LETTER doi:10.

1038/nature21433

Antigen presentation profiling reveals recognition


of lymphoma immunoglobulin neoantigens
Michael S. Khodadoust1*, Niclas Olsson2*, Lisa E. Wagar3, Ole A. W. Haabeth1, Binbin Chen1,4, Kavya Swaminathan2,
Keith Rawson2, Chih Long Liu1, David Steiner5, Peder Lund3, Samhita Rao2, Lichao Zhang2, Caleb Marceau3, Henning Stehr1,
Aaron M. Newman1,6, Debra K. Czerwinski1, Victoria E. H. Carlton7, Martin Moorhead7, Malek Faham7, Holbrook E. Kohrt1‡,
Jan Carette3, Michael R. Green1†, Mark M. Davis3,8, Ronald Levy1, Joshua E. Elias2 & Ash A. Alizadeh1,6,9

Cancer somatic mutations can generate neoantigens that distinguish specific for human leukocyte antigen (HLA)-DR, a class II MHC
malignant from normal cells1–7. However, the personalized ­molecule, and analysed by LC–MS/MS. This strategy identified over
identification and validation of neoantigens remains a major 24,000 unique MHC-I-associated peptides and over 12,500 unique
challenge. Here we discover neoantigens in human mantle-cell MHC-II-associated peptides (Fig. 1b). Both MHC-I and MHC-II
lymphomas by using an integrated genomic and proteomic strategy peptide repertoires demonstrated length distributions ­consistent
that interrogates tumour antigen peptides presented by major with those expected for each class (Fig. 1c and Extended Data
histocompatibility complex (MHC) class I and class II molecules. We Fig. 1a, b). Furthermore, MHC-I peptides showed the expected reduced
applied this approach to systematically characterize MHC ligands ­amino-acid complexity at anchor residue positions (Extended Data
from 17 patients. Remarkably, all discovered neoantigenic peptides Fig. 1c) and agreed with a widely used binding affinity model (Extended
were exclusively derived from the lymphoma immunoglobulin Data Fig. 1d–f).
heavy- or light-chain variable regions. Although we identified MHC Through whole-proteome analysis of two MCL cell lines, we found
presentation of private polymorphic germline alleles, no mutated MHC-I and MHC-II presentation was significantly biased towards
peptides were recovered from non-immunoglobulin somatically abundant proteins (Extended Data Fig. 2). In contrast, we found
mutated genes. Somatic mutations within the immunoglobulin mutated proteins tended to be significantly less abundant than ­average.
variable region were almost exclusively presented by MHC class II. We found a high degree of overlap among genes presented by MHC
We isolated circulating CD4+ T cells specific for immunoglobulin- across patients (Extended Data Fig. 3a, b). However, the specific
derived neoantigens and found these cells could mediate killing ­peptides we recovered were generally private to each individual, with
of autologous lymphoma cells. These results demonstrate that the exception of patients who shared MHC-I and/or MHC-II alleles
an integrative approach combining MHC isolation, peptide (Extended Data Fig. 3c–f), further confirming MHC as the source of
identification, and exome sequencing is an effective platform to the recovered peptides. Among the recurrently presented genes were
uncover tumour neoantigens. Application of this strategy to human members of the B-cell receptor signalling pathway including IGHM,
lymphoma implicates immunoglobulin neoantigens as targets for CD79B, BTK, LYN, SYK, MS4A1 (CD20), and CD22, which showed
lymphoma immunotherapy. significant enrichment (Fig. 2a and Extended Data Fig. 3g).
We sought to profile MHC antigen repertoires of primary human There were 13–175 non-synonymous somatic mutations per patient
lymphomas, with the intention of discovering cancer neoantigens. exome, including genes known to be recurrently mutated in MCL
Typically, ‘reverse immunology’ neoantigen identification ­strategies such as Ig variable regions, TP53, CCND1, ATM, UBR5, SMARCA4,
have relied first on the isolation of cognate T cells to then ­identify NOTCH1, FAT4, TRAF2, and WHSC1 (refs 17, 18) (Fig. 2b). We
the ­candidate ­antigens. By contrast, direct proteomic ­analysis of also identified novel genes as recurrently mutated in MCL including
­cancer MHC ­ligands8–14 by liquid chromatography and tandem mass PBRM1 and SMARCB1, with the latter gene previously found to be
­spectrometry (LC–MS/MS) enables discovery of tumour ­antigens, mutated in a single MCL patient19. Most non-synonymous mutations
including n ­ eoantigens, directly from cancer cells. We ­profiled occurred in genes not characteristically expressed in MCL20 (Fig. 2c).
­lymphoma MHC-I and MHC-II ligands from 17 patients with untreated Among expressed genes bearing coding mutations, we found 46%
­mantle-cell lymphoma (MCL) and additionally from two MCL cell lines with at least one peptide presented by either class I or class II MHC,
(Fig. 1a). We focused on MCL, a subtype of B-cell non-Hodgkin lym- suggesting active processing of these mutated proteins. However, with
phoma with characteristically high expression of both class I and class the e­ xception of peptides derived from Ig genes, the MHC-presented
II MHC ­molecules, because of the availability of large numbers of these peptides were derived from the non-mutated regions of these genes,
tumour cells that had been collected as part of an ongoing clinical trial of as exemplified by the two most frequently mutated non-Ig genes, TP53
immunotransplantation (NCT00490529). To define candidate somatic and CCND1 (Fig. 2d).
neoantigens, we used our previously described approach for whole-­ We recovered neoantigen peptides from 13 genes, all of which were
exome sequencing of DNA from highly pure tumour cells and matched derived from Ig variable regions. To test whether the lack of non-Ig
germline, and ­additionally directly sequenced the expressed lymphoma ­neoantigens was due to technical limitations in recovering private
immunoglobulin (Ig) heavy- and light-chain variable regions15,16. peptide variants, we assessed the recovery of peptides encoded by
Peptides bound to MHC-I and MHC-II were purified in parallel via heterozygous germline single nucleotide polymorphisms (SNPs) for
immunoprecipitation with a pan-MHC-I antibody and an antibody each patient. This analysis revealed significantly greater presentation of
1
Department of Medicine, Division of Oncology, Stanford University, Stanford, California 94305, USA. 2Department of Chemical & Systems Biology, Stanford University, Stanford, California
94305, USA. 3Department of Microbiology & Immunology, Stanford University, Stanford, California 94305, USA. 4Department of Genetics, Stanford University, Stanford, California 94305,
USA. 5Department of Pathology, Stanford University, Stanford, California 94305, USA. 6Center for Cancer Systems Biology, Stanford University, Stanford, California 94305, USA. 7Adaptive
Biotechnologies, South San Francisco, California 94080, USA. 8Howard Hughes Medical Institute, Stanford University, Stanford, California 94305, USA. 9Institute for Stem Cell Biology &
Regenerative Medicine, Stanford University, Stanford, California 94305, USA. †Present address: Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska
68198, USA.
‡Deceased.
*These authors contributed equally to this work.

3 0 M A RC H 2 0 1 7 | VO L 5 4 3 | NAT U R E | 7 2 3
© 2017 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
RESEARCH LETTER

a assayed eight patients’ CD8 T-cell responses against ­computationally


n = 17 Lymphoma biopsy predicted HLA-A2 restricted neoantigens with peptide–MHC
­tetramers (Extended Data Fig. 5). No immune responses were detected
against any of the 108 putative neoantigens tested.
Peptide–MHC
immunoprecipitation
Whole-exome sequencing Immunoglobulin sequencing The Ig heavy chain was presented by both MHC-I and MHC-II
Genomic DNA cDNA
in all 17 patients (Fig. 2b). MHC-I and MHC-II displayed strikingly
Exome capture PCR + sequencing
distinct antigen presentation patterns from this protein. Peptides
V DJ C
IgH
Igκ
­presented by MHC-I mapped to the Ig constant regions (199 peptides,
IgL
V J C Supplementary Note 1 and Supplementary Table 1), but there were
Elute few peptides recovered from the Ig variable regions (eight peptides)
peptides
(Fig. 3a). In contrast to MHC-I, antigen presentation of Ig by MHC-II
Patient-specific database was clustered within the variable region (Fig. 3a). Specifically,
MHC-I peptides MHC-II peptides ~​93% of MHC-II-bound peptides (105/113) spanned the junction
Whole-exome variants of c­ omplementarity-determining region 3 (CDR3) and framework
LC–MS/MS
region 3 (Fig. 3a). Nearly half of the peptides presented by MHC-II
Immunoglobulin variants from this hotspot were neoantigens created by somatic h ­ ypermutation
or V(D)J gene rearrangement (52/105 peptides, 49.5%) (Fig. 3b and
785.49664

987.35614

584.16870

Supplementary Table 2). In contrast, only a single non-germline ­peptide


859.29712
684.34528
1126.46814

1068.95374
1272.48901 Swiss-Prot/TrEMBL
databases
was recovered from MHC-I. Peptides derived from light-chain v­ ariable
456.17139 1395.68408

286.13760

regions showed similar profiles: 14 neoantigen peptides formed via


Peptide identification somatic hypermutation were presented by MHC-II, but none by MHC-I
b (Fig. 3c). Using the Raji lymphoma cell line, we tested whether our
HLA-DR
HLA-A2

Class I MHC
approach did not detect MHC-I-restricted Ig neoantigens as a result of
Source

HLA-A2
Class II MHC
Ki67
IgVH

Non-HLA-A2
technical obstacles. We ectopically expressed an MHC-I allele with high
MCL001
MCL041 DRB-01 predicted affinity for Raji Ig neoantigens. Antigen presentation profiling
MCL034
MCL052
DRB-04
of the resultant Raji cells revealed three Ig neoantigens, thus confirming
DRB-07
MCL030
DRB-11
our ability to recover MHC-I Ig neoantigens (Extended Data Fig. 6).
MCL012
MCL008 To validate that neoantigens are true HLA-DR ligands, we s­ ynthesized
MCL037
MCLX001
Blood neoantigen peptides from three patients with an HLA-DR*​0401
Lymph node
MCL022
Spleen
allele and confirmed their affinity for HLA-DR by exchange onto
MCL005
MCL049 recombinant HLA-DR*​0401 molecules for five of the six tested
MCL043
MCL038
<10%
10–30%
peptides (Extended Data Fig. 7). We next determined whether
MCL007
MCLX002 ≥30% these patients harboured T cells that recognized their ­autologous Ig
MCL014 ­neoantigens. Neoantigen peptide-HLA-DR*​0401 tetramers revealed
JEKO Unmutated
L128 Hypermutated ­corresponding neoantigen-specific peripheral blood CD4 T cells in
one of three patients tested (Fig. 4a). These CD4+ T cells seemed
0

00
0
0

00

00
00

to be memory T cells because they lacked CD45RA expression, but


,0
2,

8,
4,

14

Unique peptides per patient they also lacked PD-1 expression, which has been associated with
c 30,000 5,000 ­neoantigen-specific T cells in patients with melanoma21 (Extended
Class I MHC
Data Fig. 8a). Tetramer-binding T cells were sorted as single cells for
Class II MHC
sequencing of T-cell receptor (TCR) α​- and β​-chains, and single-cell
Unique peptides, class II
Unique peptides, class I

4,000
gene expression phenotyping22. Of 79 cells from which TCR sequences
20,000 were obtained, we found eight derived from a single dominant T-cell
3,000
clone, and three from a second clonotype (Fig. 4b). Neither clone
was seen among 192 analysed cells that did not bind the neoantigen
2,000 tetramer. All cells expressed GATA3 and RORC, consistent with a
10,000
Th2/Th17-skewed phenotype, and a subset also expressed granzyme B
1,000 (Fig. 4b). We next profiled this patient’s neoantigen-specific T cells
following autologous tumour vaccination (Fig. 4c, top). TCR repertoire
0 0 sequencing of peripheral blood T cells collected before and after the
immunizations revealed that both neoantigen-specific T-cell clones
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
7
8
9

Length were induced by vaccination (Fig. 4c, bottom).


Figure 1 | Integrative genomic and proteomic approach for tumour antigen We next explored the therapeutic potential of patient-derived
discovery. a, Whole-exome and targeted Ig sequencing of lymphoma tumour ­neoantigen-specific T cells. In a second patient, we recovered ­neoantigen-
specimens and germline DNA was performed for 17 patients. Sequencing data specific CD4 T cells, identified by CD137 expression, after stimu­lation
were integrated with a human proteome database to create patient-specific with autologous lymphoma Ig neoepitopes (Fig. 4d). We expanded
catalogues incorporating somatically mutated proteins, lymphoma-specific these isolated T cells ex vivo and found they produced both IL-4 and
Ig, and germline variants. MHC-ligands were directly immunoprecipitated granzyme in response to lymphoma neoepitopes, suggesting potential
using both anti-HLA-A, -B, -C and anti-HLA-DR antibodies. Peptides were
then acid-eluted, profiled by LC–MS/MS, and identified with reference to
cytolytic function as previously described3,23 (Fig. 4e and Extended
patient-specific catalogues. The number of unique peptides per case (b) and Data Fig. 8b, c). Remarkably, we found these CD4 neoantigen-­specific
the length distribution of identified MHC ligands (c) are depicted. Igh, Igκ, Igl, T cells could mediate cell killing of autologous lymphoma cells
immunoglobulin heavy chain, kappa light chain, and lambda light chain genes. (Fig. 4f). Autologous Epstein–Barr virus (EBV)-transformed normal
B cells that lacked the lymphoma neoantigen were unaffected, further
germline versus somatic allelic variants across the genome (P <​  0.001, demonstrating the antigen specificity of these T cells (Fig. 4f). Similarly,
Extended Data Fig. 4). To determine whether our approach was insen- pathogen-specific T cells that were purified and expanded in parallel
sitive in detecting clinically significant neoantigens, we additionally were unable to kill lymphoma cells (Extended Data Fig. 9).

7 2 4 | NAT U R E | VO L 5 4 3 | 3 0 M A RC H 2 0 1 7
© 2017 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
LETTER RESEARCH

Antigen presentation Somatic mutations


a b

MC X002
1
MCLX00

4
MC 001

MC 052
MC 022
L 5
MC 007
MC 038
L 0
MC 037
MC 008
MC 012
L 1
MC 014
MC 049
MC 043
L03
L00

L03

L04
MHC-I only MHC-II only

L
L

L
L

L
L
L
MC

MC

MC

MC
MHC-I and MHC-II

MC X001
02
MC 001
5
MC 007
MC 008
MC 012
MC 014
MC 022
MC 030
4
MC 037
MC 038
MC 041
MC 043
MC 049
2

LX0
L00

L03

L05
175 31 24 23 22 15 17 25 24 19 38 32 22 21 17 15 13 Coding mutations

8
o

L
L
L
L
L
L

L
L
L
L
L

L
L12
Patients (%)

MC

MC

MC

MC
J ek
IGHM 100
LYN 100
PLCG2 100 605 77 71 56 63 58 43 28 64 16 13765 40 67 37 83 20 Predicted neoantigens
IGHM 96 99 95 97 99100 97 96 93 96 97 98 95 99 98 96 96
CARD11 100 TP53 VJ unmutated
PTPN6 100 CCND1 VJ hypermutated
PIK3AP1 95 UBR5
INPP5D 95 TTN
CD79B 95 PAPPA
SMARCA4 Missense
SYK 95 ATM
NFATC1 89 LRRIQ3 Frameshift
PIK3CD 89 DNAH2 Nonsense
CD22 84 SHROOM3
AKT2 74 TRAF2 Non-frameshift
BTK 74 NOTCH1
WHSC1 deletion
PPP3CA 74 PBRM1
NFKB1 63 SP140
PIK3CG 63 NRF1
RAC1 58 THBS1
CD79A 58 SMARCB1
RIMS2
RAC2 58 CSMD3
CD19 53 CRISPLD1
GRB2 53 ATG2A
PIK3CA 53 MALRD1
SOS1 53 FAT4
COL7A1

c d
Non-synonymous mutations Transactivation Tetramerization
n = 571 genes
Mutated gene Mutated protein TP53
Expressed SH3 DNA- NLS Regulatory
n = 254 genes binding
Recovered MHC-I
peptides
Protein presented
n = 117 genes Cyclin box
LXCXE PEST
Neoantigens CCND1
presented
LLXXXL
n = 13 genes Recovered MHC-I
IgVH - 9 peptides
Coding mutation
IgVκ - 4
MHC-I binding peptide

Figure 2 | Characterization of lymphoma-specific MHC-I and MHC-II genes across the cohort. Nested ovals depict increasing evidence levels
epitopes and somatic mutations. a, B-cell receptor pathway components for candidate neoantigens, starting with non-synonymous mutations
presented by >​50% of patients in the context of MHC-I (pink), MHC-II (outermost oval) to direct evidence of neoantigen presentation (innermost
(blue), or both (red). b, Non-synonymous somatic mutations, predicted oval). IgVh and IgVκ, Ig heavy variable and kappa variable regions.
neoantigens, and the degree of somatic hypermutation in IgVh or IgJh d, TP53- or CCND1-derived MHC-I peptides and observed somatic
heavy chain segments are depicted, along with genes recurrently mutated mutations are depicted in relation to the corresponding protein domains.
in the cohort by exome sequencing. c, Antigen presentation of mutated

The unexpected bias against Ig variable-region presentation by Whereas the lack of MHC-I Ig variable-region presentation may
MHC-I has important biological and therapeutic implications. r­ epresent a method of immune evasion, the contrasting f­ requent
Immunoediting represents one possible explanation for the observed ­presentation of variable-region peptides by MHC-II suggests that
bias24,25. Although we found the lymphoma Ig sequences included Ig ­neoantigen recognition by CD4 does not inhibit lymphoma
numerous variable-region peptides with predicted high affinity for self- ­development. Mouse models have suggested that such presentation
MHC-I, none were measured with our direct profiling (Extended Data may serve to promote lymphomagenesis via cognate CD4-expressing
Fig. 10). It remains unclear how primary lymphoma cells could avoid helper T cells26. While Ig neoantigen-specific T cells displayed a
presenting high-affinity Ig variable-region peptides on MHC-I, while Th2-like phenotype similar to these mouse models, they could
peptides derived from the constant region are prominently presented ­nevertheless be engaged and expanded, and they demonstrated
(Supplementary Note 2 and Supplementary Table 3). anti-tumour functions.

a MHC-I MHC-II b MHC-I MHC-II c MHC-I MHC-II


VH

Heavy-chain variable Kappa-chain variable


region (VH) VH region (Vκ) Vκ

1
CH

MCL005 peptides peptides


MHC-I n=8 MHC-I n = 13

F4 C3 F3 C2 F2 C1 F1 F4 C3 F3 C2 F2 C1 F1
MHC-II n = 113 MHC-II n = 34
CH2

IgVκ MCL034
MCL005 MCL008
neoantigen
Peptide coverage peptides MCL030*
1 8 80 MCL008
C H3

MCL012
IgVH IgVH/Vκ domains
MCL014
MCL030* neoantigen
C H4

MCL034 peptides F1 = FR1 C1 = CDR1


MCL041* F2 = FR2 C2 = CDR2
IgH peptides
MCL052 F3 = FR3 C3 = CDR3
VH n=8 n = 113 F4 = FR4
CH n =199 n = 204 MCLX001

Figure 3 | MHC-I and MHC-II presentation of lymphoma Ig. peptides created by either somatic hypermutation or V(D)J rearrangement
a, Heatmap reflects the frequency and distribution of MHC-I (left) and are aligned to expanded heatmaps, and grouped by patient (boxes).
MHC-II (right) presentation of IgM-derived peptides across the cohort. Red, somatically mutated positions within recovered peptides creating
b, c, Expanded views of antigen presentation from variable regions of neoantigens. *​Patients/peptides selected for subsequent functional
Ig heavy and kappa light chains (Vh and Vκ​), respectively. Neoantigen immunological studies.

3 0 M A RC H 2 0 1 7 | VO L 5 4 3 | NAT U R E | 7 2 5
© 2017 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
RESEARCH LETTER

a b c
Patient MCL041 TCR-β CDR3 Vaccine Immunizations
Single-cell 8 production

IgVH-specific neoantigen HLA-DR*04:01 tetramer


profiling 3
0.0
0.01%
01%
1 2

ChemoRx Remission
n = 79
cells
102
&$66<4*35*<7) &$66<4*35*<7)
MCL041 &$66<6*57<(4<) &$66<6*57<(4<)

Frequency per million reads


TN F B 1
Z 3
G XP3

TB R C
IL MB
G TA

ROF1

TG T
IL A
101

FO L6

F
12

PR
A

2
BC
CDR3 beta Clone
&$66<4*35*<7) 1 + + +
0.00% &$66<4*35*<7) 1 + + + +
&$66<4*35*<7) 1 + + +
&$66<4*35*<7) 1 + + + 100
&$66<4*35*<7) 1 + + +
&$66<4*35*<7) 1 + + +
&$66<4*35*<7) 1 + + +
&$66<4*35*<7) 1 + +
ND
&$66<6*57<(4<) 2 + + +
Healthy donor &$66<6*57<(4<) 2 + + + 1 2

CMV-specific &$66<6*57<(4<) 2 + + ine tions ine


cc a cc
HLA-DR*04:01 tetramer r e -va uniz st-va
P
Im
m Po

d Patient MCL030 e f
Antigenic stimulus: Expanded IgV neoantigen- Effectors:
mutant IgV neoepitopes specific CD4+ T cells IgV neoantigen-specific T cells

0.03% 45% Targets: 41%


Sort and Co- autologous
Granzyme B

expand culture lymphoma cells

Counts
CD137 (4-1BB)

0.31%
CD4 Effectors:
Germline IgV epitopes IgV neoantigen-specific T cells

0.00% Targets:
autologous
EBV-transformed
B cells
Counts

9%
0.18%

CD69 Death (7-AAD)

Figure 4 | Detection and cytololytic activity of neoantigen-specific from TCR-β​repertoire sequencing. ChemoRx, chemotherapy; ND, not
CD4 T cells. a, Peripheral blood CD4 T cells from patient MCL041 (top) detected. d, CD4 T cells from a second patient were stimulated with
or a healthy donor (bottom) were stained with neoantigen-specific or either Ig neoantigen peptides (top) or corresponding unmutated germline
cytomegalovirus (CMV)-specific HLA-DR tetramers. b, Neoantigen- counterparts (bottom). e, Sorted neoantigen-specific cells were expanded
specific T cells were sorted and subjected to single-cell TCR-β​ sequencing and immunophenotyped for granzyme B expression. f, Cytotoxic activity
(top) and RNA expression profiling (bottom), with recurrent neoantigen- of neoantigen-specific T cells against either autologous lymphoma cells
specific TCR-β​clones highlighted. c, Pre- and post-immunization (top) or EBV-transformed B cells (bottom), determined by 7-AAD uptake.
peripheral blood frequencies of corresponding neoantigen-specific clones

These findings also inform upon the previous experience with from specimens with large mutation burdens, including cancer cell
i­ diotype therapy. Idiotype vaccines seemed to provoke predominantly lines13 and melanoma11,12. By incorporating MHC class II ligands
CD4 and/or humoral responses, consistent with our finding of selective into our analysis, we uncover a different category of lymphoma-­
MHC-II presentation of Ig antigens and CD4 Ig neoantigen-specific specific n ­ eoantigens. We find that, apart from Ig, mutated antigens
T cells27,28. Clinical trials of vaccine therapy targeting lymphoma Ig are not prominently presented as MHC epitopes in MCL, similar to
variable regions showed early promise, but three independent phase other h ­ aematological malignancies with low to moderate mutation
III trials of idiotype vaccine found either modest or no overall c­ linical ­burden8,9,14. It remains unclear whether the lack of recovered MHC
benefit29–31. Nevertheless, the subgroup of patients who generated neoantigens from primary human tumours reflects their true absence
immune responses were noted to derive substantial benefit30. Our or a limitation of our method in detecting rare epitopes. Nevertheless,
results suggest that MHC presentation of variable-region epitopes is our findings suggest that the epitopes uncovered through direct,
not equivalent across all individual lymphomas, raising the possibility proteomic antigen profiling represent actionable antigen targets for
that the patients who benefited from idiotype vaccination were those T-cell therapy. Here, we leverage our ability to recover precise CD4
with active presentation of their Ig neoepitopes. neoantigens to discover and augment the cytotoxic potential of the
The number of somatic mutations, and therefore potential neoantigen-specific CD4+ T cells. Our results suggest that e­ xpansion
­neoantigens, has been posited as a predictor of immune checkpoint of endogenous Ig n ­ eoantigen-specific CD4+ T cells may represent
response in malignancies with high mutational burden, such as lung a viable treatment strategy for lymphoma. We demonstrate that by
cancer and melanoma4,32,33. Neoantigen discovery based on mass integrating genomic and proteomic analysis, antigen presentation
­spectrometry has successfully identified MHC class I neoantigens profiling ­effectively identifies clinically relevant tumour antigens, and

7 2 6 | NAT U R E | VO L 5 4 3 | 3 0 M A RC H 2 0 1 7
© 2017 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
LETTER RESEARCH

reveals class II MHC restricted Ig neoantigens as an effective target for 24. Trojan, A. et al. Immunoglobulin framework-derived peptides function as
cytotoxic T-cell epitopes commonly expressed in B-cell malignancies. Nature
­lymphoma immunotherapy. Med. 6, 667–672 (2000).
25. Strothmeyer, A. M. et al. Comparative analysis of predicted HLA binding of
Online Content Methods, along with any additional Extended Data display items and immunoglobulin idiotype sequences indicates T cell-mediated
Source Data, are available in the online version of the paper; references unique to immunosurveillance in follicular lymphoma. Blood 116, 1734–1736
these sections appear only in the online paper. (2010).
26. Zangani, M. M. et al. Lymphomas can develop from B cells chronically helped
Received 10 August 2016; accepted 30 January 2017. by idiotype-specific T cells. J. Exp. Med. 204, 1181–1191 (2007).
Published online 22 March 2017. 27. Kwak, L. W. et al. Induction of immune responses in patients with B-cell
lymphoma against the surface-immunoglobulin idiotype expressed by their
tumors. N. Engl. J. Med. 327, 1209–1215 (1992).
1. Robbins, P. F. et al. Mining exomic sequencing data to identify mutated
28. Neelapu, S. S. et al. Vaccine-induced tumor-specific immunity despite
antigens recognized by adoptively transferred tumor-reactive T cells. Nature
severe B-cell depletion in mantle cell lymphoma. Nature Med. 11, 986–991
Med. 19, 747–752 (2013).
(2005).
2. Linnemann, C. et al. High-throughput epitope discovery reveals frequent
29. Freedman, A. et al. Placebo-controlled phase III trial of patient-specific
recognition of neo-antigens by CD4+ T cells in human melanoma. Nature Med.
immunotherapy with mitumprotimut-T and granulocyte-macrophage
21, 81–85 (2015).
colony-stimulating factor after rituximab in patients with follicular lymphoma.
3. Tran, E. et al. Cancer immunotherapy based on mutation-specific CD4+ T cells
J. Clin. Oncol. 27, 3036–3043 (2009).
in a patient with epithelial cancer. Science 344, 641–645 (2014).
30. Levy, R. et al. Active idiotypic vaccination versus control immunotherapy for
4. van Rooij, N. et al. Tumor exome analysis reveals neoantigen-specific T-cell
follicular lymphoma. J. Clin. Oncol. 32, 1797–1803 (2014).
reactivity in an ipilimumab-responsive melanoma. J. Clin. Oncol. 31,
31. Schuster, S. J. et al. Vaccination with patient-specific tumor-derived antigen in
e439–e442 (2013).
first remission improves disease-free survival in follicular lymphoma. J. Clin.
5. Rajasagi, M. et al. Systematic identification of personal tumor-specific
Oncol. 29, 2787–2794 (2011).
neoantigens in chronic lymphocytic leukemia. Blood 124, 453–462 (2014).
32. Van Allen, E. M. et al. Genomic correlates of response to CTLA-4 blockade in
6. Khodadoust, M. S. & Alizadeh, A. A. Tumor antigen discovery through
metastatic melanoma. Science 350, 207–211 (2015).
translation of the cancer genome. Immunol. Res. 58, 292–299 (2014).
33. Rizvi, N. A. et al. Mutational landscape determines sensitivity to PD-1 blockade
7. Kreiter, S. et al. Mutant MHC class II epitopes drive therapeutic immune
in non-small cell lung cancer. Science 348, 124–128 (2015).
responses to cancer. Nature 520, 692–696 (2015).
8. Kowalewski, D. J. et al. HLA ligandome analysis identifies the underlying
specificities of spontaneous antileukemia immune responses in chronic Supplementary Information is available in the online version of the paper.
lymphocytic leukemia (CLL). Proc. Natl Acad. Sci. USA 112, E166–E175 (2015).
9. Berlin, C. et al. Mapping the HLA ligandome landscape of acute myeloid Acknowledgements We are grateful to the patients and their families who
leukemia: a targeted approach toward peptide-based immunotherapy. participated in this study, as well as to M. Diehn, S. Levy, and members of their
Leukemia 29, 647–659 (2015). laboratories for feedback. This work was supported by the following grants:
10. Dutoit, V. et al. Exploiting the glioblastoma peptidome to discover novel National Institutes of Health (NIH) U01 CA194389 (M.M.D., R.L., J.E.E., A.A.A.),
tumour-associated antigens for immunotherapy. Brain 135, 1042–1054 NIH PPG CA49605 (R.L.); American Society of Hematology Scholar Award
(2012). (A.A.A.), V-Foundation (A.A.A.), Damon Runyon Cancer Research Foundation
11. Kalaora, S. et al. Use of HLA peptidomics and whole exome sequencing to (A.A.A.); Damon Runyon-Rachleff Innovation Award (J.E.E.), W.M. Keck
identify human immunogenic neo-antigens. Oncotarget 7, 5110–5117 (2016). Foundation Medical Research Grant (J.E.E.); Conquer Cancer Foundation
12. Bassani-Sternberg, M. et al. Direct identification of clinically relevant Young Investigator Award (M.S.K.), Fellow Award from the Leukemia
neoepitopes presented on native human melanoma tissue by mass & Lymphoma Society (M.S.K.); Knut and Alice Wallenberg Foundation
spectrometry. Nature Commun. 7, 13404 (2016). Postdoctoral Fellowship (N.O.); Stanford Translational Research and Applied
13. Yadav, M. et al. Predicting immunogenic tumour mutations by combining mass Medicine Pilot Grant (M.R.G., H.E.K., and A.A.A.); NIH S10 RR02933801.
spectrometry and exome sequencing. Nature 515, 572–576 (2014). We thank the NIH Tetramer Facility for providing recombinant HLA-A0201 for
14. Walz, S. et al. The antigenic landscape of multiple myeloma: mass tetramer experiments.
spectrometry (re)defines targets for T-cell-based immunotherapy. Blood 126,
1203–1213 (2015).
Author Contributions M.S.K., N.O., M.M.D., R.L., J.E.E., and A.A.A. developed
15. Green, M. R. et al. Hierarchy in somatic mutations arising during genomic
the concept, designed the experiments, and analysed the data. R.L. provided
evolution and progression of follicular lymphoma. Blood 121, 1604–1611
patient material from the associated clinical trial. M.S.K., M.R.G., and A.A.A.
(2013).
performed molecular biology, genomic analyses, and neoantigen classification
16. Green, M. R. et al. Mutations in early follicular lymphoma progenitors are
with assistance from C.L.L., H.S., H.E.K., D.S., and B.C. N.O., K.R., M.S.K., and
associated with suppressed antigen presentation. Proc. Natl Acad. Sci. USA
J.E.E. performed proteomic studies and analyses with assistance from B.C.,
112, E1116–E1125 (2015).
S.R., L.Z., K.S., P.L., C.M., and J.C. M.S.K., L.E.W., O.A.W.H., and D.K.C. performed
17. Beà, S. Cyclin D1 transcriptional activation in MCL. Blood 123, 1979–1980
immunological experiments and analyses. V.E.H.C., M.M., and M.F. performed
(2014).
TCR repertoire sequencing. M.S.K., N.O., B.C., K.R., A.M.N., C.L.L., D.S., J.E.E., and
18. Zhang, J. et al. The genomic landscape of mantle cell lymphoma is related to
A.A.A. performed integrative data analyses. M.S.K., N.O., R.L., J.E.E., and A.A.A.
the epigenetically determined chromatin state of normal B cells. Blood 123,
wrote the manuscript with contributions from all authors, who commented on
2988–2996 (2014).
it at all stages.
19. Beà, S. et al. Landscape of somatic mutations and clonal evolution in mantle
cell lymphoma. Proc. Natl Acad. Sci. USA 110, 18250–18255 (2013).
20. Kridel, R. et al. Whole transcriptome sequencing reveals recurrent NOTCH1 Author Information Reprints and permissions information is available at
mutations in mantle cell lymphoma. Blood 119, 1963–1971 (2012). www.nature.com/reprints. The authors declare competing financial interests:
21. Gros, A. et al. PD-1 identifies the patient-specific CD8+ tumor-reactive details are available in the online version of the paper. Readers are welcome to
repertoire infiltrating human tumors. J. Clin. Invest. 124, 2246–2259 (2014). comment on the online version of the paper. Publisher’s note: Springer Nature
22. Han, A., Glanville, J., Hansmann, L. & Davis, M. M. Linking T-cell receptor remains neutral with regard to jurisdictional claims in published maps and
sequence to functional phenotype at the single-cell level. Nature Biotechnol. institutional affiliations. Correspondence and requests for materials should be
32, 684–692 (2014). addressed to A.A.A. (arasha@stanford.edu) or J.E.E. (josh.elias@stanford.edu).
23. Quezada, S. A. et al. Tumor-reactive CD4+ T cells develop cytotoxic activity and
eradicate large established melanoma after transfer into lymphopenic hosts. Reviewer Information Nature thanks C. Melief and the other anonymous
J. Exp. Med. 207, 637–650 (2010). reviewer(s) for their contribution to the peer review of this work.

3 0 M A RC H 2 0 1 7 | VO L 5 4 3 | NAT U R E | 7 2 7
© 2017 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
RESEARCH LETTER

METHODS the full-length variable region41. Tumour RNA was used to create complementary
No statistical methods were used to predetermine sample size. The experiments DNA (cDNA) using Superscript Reverse Transcriptase (ThermoFisher Scientific)
were not randomized. The investigators were not blinded to allocation during according to standard protocol using random hexamers. PCR was performed with
experiments and outcome assessment. Kapa HiFi HotStart ReadyMix (Kapa Biosystems) with an annealing temperature
Tumour specimens. All specimens were obtained with informed consent in of 68–70 °C using a mix of forward V-leader family primers and a reverse primer
accordance with the Declaration of Helsinki and this study was approved by in the constant region (Supplementary Table 4). PCR products were purified by
Stanford University’s Administrative Panels on Human Subjects in Medical agarose gel electrophoresis and extraction. PCR products were sequenced by
Research. Samples were collected from patients with untreated MCL by e­ xcisional Sanger sequencing either directly or after subcloning into the pCR-Blunt vector
lymph node biopsy, or splenectomy, or peripheral blood leukapheresis, and (ThermoFisher Scientific). VH usage and germline homology was determined
cryopreserved. using IMGT, the international ImMunoGeneTics information system (http://www.
Vaccine trial. Patient MCL041 was treated as part of an ongoing clinical trial of imgt.org)42.
tumour vaccination (NCT00490529). Vaccine was prepared by ex vivo culture of Generation of custom reference proteome databases. Two protein-­sequence
circulating autologous lymphoma cells in AIM-V media and human AB-negative databases were constructed. Both were built upon the human proteome
sera containing 3 μ​g/ml PF-3512676 (CpG molecule) for 72 h. Cells were then UniprotKB database including Swiss-Prot and TrEMBL databases (version May
irradiated with 200 Gy and cryopreserved. The patients were treated with ­induction 2015) with common contaminant protein sequences included (for example,
chemotherapy and those with a complete remission then received a series of Staphylococcus protein A). The first was a somatic personalized database which
three weekly autologous tumour vaccinations. Leukapheresis was used to collect included ­predicted mutated peptides inferred from somatic SNVs and indels
­peripheral blood mononuclear cells before and after the series of vaccinations. from exome sequencing, as well as the lymphoma-specific IgVh and light-chain
Cell lines. JEKO and L128 MCL lines were originally obtained from the Lymphoma variable sequences as inferred from Ig V(D)J sequencing. The second database
Research Foundation/Mantle Cell Lymphoma Consortium American Type Culture ­consisted of the same human proteome UniProt database but with added peptides
Collection (ATCC) cell line resource, tested for mycoplasma, and authenticated derived from patient-specific coding, heterozygous SNPs, and cell-line-specific
by STR profiling. Cell lines were grown according to ATCC recommendations. SNPs. Personalized entries consisted of the mutated amino acid(s) with flanking
Exome sequencing. Tumour whole-exome sequencing was performed for 21 amino acids both amino (N)-terminal and carboxy (C)-terminal to the mutated
17 patients and the JEKO and L128 MCL cell lines as described previously15,16. residue(s). Reversed ‘decoy’ protein sequences were appended to both of these
Tumour DNA was collected from either peripheral blood or lymph node biopsy. databases for ‘target-decoy’ error estimation43.
Tumour cells were purified by either flow cytometry or by magnetic bead Purification of HLA peptides from cells. MHC class I and class II pepti-
­purification for CD19 expression, or in some cases were not purified if the biopsy domes were extracted from the MCL tumours of 17 patients and from two cell
sample had ≥​90% tumour purity. Germline DNA was obtained either from lines (JEKO, L128). The MHC class I or MHC class II molecules were isolated
­peripheral blood collected at time of complete remission or by fluorescence-­ and the associated peptides extracted as previously described44,45 with some
activated cell sorting (FACS) of CD3+ T cells from the initial tumour specimen. ­modifications. Before lysis, all cells were washed twice with PBS. For all patient
DNA and RNA were extracted from fresh single-cell suspensions using a Qiagen samples, 1 ×​  108 cells were lysed per MHC preparation, while for the cell lines
Allprep kit (Qiagen). Library preparation and exome capture were performed 1 ×​  108 and 1 ×​  109 cells were used to evaluate the extent to which limited input
as previously described16. Next-generation sequencing libraries were prepared material may reduce the sensitivity ligand detection. In brief, cells were lysed for
using KAPA Libarary Preparation kits (KAPA Biosystems). Briefly, 100 ng to 20 min on ice in 20 mM Tris-HCl (pH8), 150 mM NaCl, 1% (w/v) CHAPS, 0.2 mM
1.5 μ​g of DNA per sample was sheared by sonication using a Covaris S-series PMSF, 1×​Halt Protease, and Phosphatase Inhibitor Cocktail (ThermoFisher
instrument (Covaris). Sheared DNA was end-repaired according to the manu- Scientific) supplemented with complete Protease Inhibitor Cocktail (Roche).
facturer’s protocol and ligated with Illumina sequencing adaptors overnight at The lysate was subjected to ­centrifugation (2 ×​ 30 min, 13,200 r.p.m. at 4 °C) and
16 °C. Adaptor-ligated product was enriched using eight to ten cycles of PCR with the resulting supernatant was pre-cleared for 30 min using rProtein A Sepharose
Phusion High-Fidelity PCR Master Mix (New England Biolabs), and size-selected Fast Flow beads (GE Healthcare). For all MHC-1 immunoprecipitations, the
using Agencourt Ampure XP beads (Beckman Coulter). Purified libraries were pre-cleared lysate was ­incubated with the pan HLA-A-, B-, and C- antibody
evaluated for size distribution using a BioAnalyzer High-Sensitivity DNA kit W6/32 ­(purified in-house from hybridoma provided by B. Mellins and with Ig
(Agilent), and quantified using spectrophotometry and a Qubit dsDNA HS assay produced by Genentech46) ­coupled to rProtein A Sepharose Fast Flow beads for
(Invitrogen). Next-generation sequencing libraries were pooled and enriched 5 h at 4 °C. For MHC-II ­captures, the pre-cleared lysate was incubated with the
for coding sequences using the SeqCap EZ Exome version 3.0 capture reagent HLA-DR-specific ­antibody L243 (produced and purified by Genentech from our
(NimbleGen). Hybrid captures were performed according to the manufacturer’s ­hybridoma)47 ­coupled to rProtein A Sepharose Fast Flow beads for 5 h at 4 °C. After
protocol, with all 47 °C steps conducted on a thermal cycler to ensure maximal ­immunoprecipitation, the beads were washed with TBS (pH 7.4) and peptides were
temperature stability. Pooled libraries were amplified post-enrichment using six to eluted from the ­purified MHC molecules using 10% acetic acid. Eluted peptides
eight cycles of PCR. Illumina sequencing services were performed by the Stanford were further p ­ urified with a 10 kDa molecular weight cut-off size filter, followed
Center for Genomics and Personalized Medicine, with 101-base-pair paired-end by a ­concentration step using vacuum centrifugation, de-salted on C18-based
reads on a HiSeq 2000 instrument (Illumina) loading one capture pool per lane. STAGE tips48. Peptides were lyophilized and stored at −​80 °C until LC–MS/MS
Exome sequencing achieved a median depth of coverage over this region 94×​ in analysis.
purified tumour B cells and 71×​in germline samples. Mass spectrometry analysis of HLA-associated peptides. For MS analysis,
Variant calling and annotation. Raw sequences were demultiplexed and adaptor ­isolated peptides were reconstituted in 0.1% formic acid and analysed on an LTQ
sequences trimmed. Reads were aligned to UCSC hg19 reference genome with Orbitrap Elite mass spectrometer (Thermo Fisher Scientific) in three ­replicate
the BWA mem aligner34 and duplicate reads were removed. Single-nucleotide injections of 25% of the total specimen amount. Samples were separated by
­variant (SNV) and indel variants were called by VarScan2 (ref. 35) using reads with ­capillary reverse-phase chromatography on an 18 cm reversed-phase column
­mapping quality >​10, minimum base quality >​20, minimum variant f­ requency (100 μ​m inner diameter, packed in-house with ReproSil-Pur C18-AQ 3.0 m resin
0.1, minimum of three variant reads, strand bias <​90%, minimum normal (Dr. Maisch)) over a total run time of 160 min using a two-step linear g­ radient with
­coverage at least six reads, and minimum tumour coverage at least eight reads. 4–25% buffer B (0.2% (v/v) formic acid, 5% DMSO, and 94.8% (v/v) a­ cetonitrile)
High-confidence somatic SNVs were then realigned around indels by Varscan2 and for 120 min followed by 25–40% buffer B for 30 min using an Eksigent ekspert
filtered using the false-positive filter script fpffilter.pl36. Variants were annotated nanoLC-425 system (SCIEX, Framingham, Massachusetts, USA). Three injections
and coding changes were predicted using Annovar37. Germline SNPs were called were made per sample to utilize multiple fragmentation modes (HCD (higher-­
using VarScan2 with the above parameters. For cell lines, SNPs were assigned as energy collisional dissociation) or CID (collision-induced dissociation)). The third
germline if they were present in the NCBI dbSNP137 database. injection was performed with CID including singly charged species. Acquisition
Neoantigen prediction. Coding variants were annotated to provide the was executed in data-dependent mode with the full MS scans acquired in the
­corresponding mutated protein sequences using Annovar. Patient HLA alleles were Orbitrap mass analyser with a resolution of 60,000 and m/z scan range 340–1,600.
extracted from sequencing of autologous normal cells using Phlat38. Neoantigen The top ten most intense ions were then selected for sequencing and f­ ragmented
predictions were performed with NetMHC 3.4 (ref. 39) or, when the HLA allele was in the Orbitrap mass analyser at a resolution of 15,000 (full width at half
not included in the NetMHC model, NetMHCpan2.8 (ref. 40). Unless otherwise maximum). Data-dependent scans were acquired from precursors with masses
specified, peptides with predicted half-maximum inhibitory concentrations (IC50) ranging from 700 to 1,800 Da for all MHC class 1 samples and from 700 to 2,750 Da
<​500 nM were considered neoantigens. for MHC class II samples. Precursor ions were fragmented with a normalized
Targeted Ig variable-region sequencing. Sequencing of Ig heavy and light chains collision energy of 35% and an activation time of 5 ms for CID and 30 ms for HCD.
was performed using modification of the BIOMED-2 protocol to reliably sequence Repeat count was set to 2 and fragmented m/z values were dynamically excluded

© 2017 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
LETTER RESEARCH

from further selection for a period of 30 s. The minimal signal threshold was set Wisconsin, USA) at a ratio of 1:25 enzyme:substrate for 16 h at 37 °C. The ­reaction
to 500 counts. was quenched with formic acid and the peptides de-salted using a Sep-Pak C18 1 cc
Computational identification of HLA peptides from mass spectra. All t­ andem Vac Cartridge, 50 mg (Waters, Milford, Massachusetts, USA). Digested p ­ eptides
mass spectra were queried against personalized somatic and personalized germline were re-suspended in 130 μ​l 10 mM ammonium formate, pH 10, followed by high-
proteome databases described above using both SEQUEST49 and PEAKS DB pHRP fractionation54,55 using a 65 min +​ 15 min step-gradient (buffer A (10 mM
search engines (PEAKS Studio 7, Bioinformatics Solutions)50. Spectra were also ammonium formate, pH 10) and buffer B (10 mM ammonium formate, 90% ACN,
interpreted by de novo sequencing (PEAKS Studio 7, Bioinformatics Solutions) to 10% H2O, pH 10)) using an Agilent 1200 HPLC (Agilent Technologies). In total, 84
improve high-confidence peptide identification. The msConvert program (­ version fractions were collected, concatenated, and combined55 into a total of 12 ­fractions
3.0.45) was used to generate peak lists from the RAW data files, and spectra were and lyophilized by vacuum centrifugation. All fractions were cleaned using C18-
then assigned to peptides using the SEQUEST (version 28.12) ­algorithm. RAW based STAGE Tips48, lyophilized, and stored at −​80 °C until final LC–MS/MS
data files were imported into PEAKS Studio 7 before PEAKS DB and PEAKS measurement.
de novo searching and subjected to default data refinement (deisotoping, charge Mass spectrometry analysis of cell line proteomes. Each concatenated peptide
­deconvolution, peak centroiding). For all searches, the parent mass error t­ olerance fraction was re-suspended in 20 μ​l 0.1% formic acid. In total 10% of the material
was set to 20 p.p.m. and the fragment mass error tolerance to 0.02 Da. For was injected and separated on an 18 cm reversed-phase column (100 μ​m inner
SEQUEST and PEAKS DB, enzyme specificity was set to none and oxidation of diameter), packed in-house with ReproSil-Pur C18-AQ 3.0 m resin (Dr. Maisch
methionines and deamidation (N, Q), cysteinylation, and phosphorylation (S, T, Y) GmbH) over a total run of 160 min using a two-step linear gradient with 4–25%
were considered as variable modifications. buffer B (0.2% (v/v) formic acid, 5% DMSO, and 94.8% (v/v) acetonitrile) for
High-confidence peptide identifications were selected at a 1% false ­discovery 120 min followed by 25–40% buffer B for 30 min using an Eksigent ekspert
rate with a version of the Percolator algorithm, which we modified for nanoLC-425 system (SCIEX, Framingham, Massachusetts, USA). The LC system
­proteogenomic immunopeptide analysis51. Percolator implements a linear SVM was coupled online with an Orbitrap Elite instrument (Thermo Fisher Scientific,
­(support vector machine) to score peptide identifications based on multiple peptide Bremen, Germany) via a nano-electrospray source. Full scans were acquired in
­attributes. We modified the Percolator method to incorporate peptide attributes the Orbitrap mass analyser with resolution 60,000 at 340–1,600 m/z. Unassigned
that influence the confidence in immunopeptide identifications. These included charge states were rejected and the top 20 most intense ions with charge states 2
whether the de novo sequence matched database assignments; the magnitude of and up were sequentially isolated for MS/MS analysis using CID fragmentation.
scores assigned by all three search algorithms; the source of the identified peptide A minimal signal of 500 was required, the normalized collision energy was set at
(Swiss-Prot, TrEMBL, or our own sequencing); and the presence of one or more 35%, and the fragmented peptide masses were collected in the ion-trap. Dynamic
post-translational modifications in the identified peptide. Unlike conventional exclusion was enabled with a repeat count of 1 and the repeat duration set to 30 s.
proteome analysis, false discovery rates were not evaluated at the level of assembled HLA-B*35 construct and genetic transduction of Raji cells. The cDNA sequence
proteins, as this would have unnecessarily penalized proteins identified by small of full-length HLA-B*​35:01:01:01:01 was synthesized (IDT) with 25 base pairs of
numbers of peptide. overlapping vector sequence on either end and cloned into pLenti-CMV-Puro-
Confirmation of HLA identifications with synthetic peptides and mass DEST (w118-1, a gift from E. Campeau) at the EcoRV sites using Gibson assembly
­spectrometry. Synthetic peptides corresponding to the idiotype peptides (NEB). A two-step PCR was used to insert an N-terminal Flag tag ­downstream
from MCL-41 (NMLFLQMNSLKTEDT, DDSKNMLFLQMNSLKTEDT, of the signal peptide. Lentivirus produced in HEK293FT cells was used to
FLQMNSLKTEDT, IKNKIDGETTDYAAPV) were purchased from JPT Peptide ­transduce Raji cells overnight. Transduced cells were selected by treatment with
Technologies (Berlin, Germany) with a purity of >​70%, dissolved, and re-­ ­puromycin (1 μ​g/ml, InvivoGen) for 7 days. Raji cells overexpressing the viral
suspended into 0.1% formic acid. For each peptide, 250 fmol was analysed on an entry ­receptor DC-SIGN (a gift from E. Harris, University of California, Berkeley)
LTQ-Orbitrap Elite mass ­spectrometer (ThermoFisher, Bremen, Germany) using with and ­without HLA*​B35:01 overexpression were cultured in RPMI medium
the same instrument s­ ettings as described above. Synthetic peptides were measured supplemented with 5% fetal bovine serum and 1×​penicillin/streptomycin and
with two replicate LC–MS analyses, using either the CID or HCD fragmentation l-­glutamine.
methods as defined above. Both included singly charged species. Computational interpretation of cell-line proteomes. Data were analysed with
Quality assessment of HLA ligands. Sequence logos for recovered MHC-I ProteomeDiscover version 2.1.0.81 (Thermo Fisher Scientific) and SEQUEST-HT
­peptides were produced using Icelogo (http://iomics.ugent.de/icelogoserver) to with the following settings: the parent mass error tolerance was set to 20 p.p.m. and
reveal position weighting at anchor residues52. The size distribution of r­ ecovered the fragment mass error tolerance to 0.6 Da. Strict trypsin specificity was required,
MHC-I and MHC-II bound peptides was compared with size distributions of allowing for up to two missed cleavages. Carbamidomethylation of cysteine was
known MHC-I and MHC-II ligands from the Immune Epitope Database and set as fixed modification and oxidation of methionines as a variable modification.
Analysis resource (http://www.iedb.org, January 2016) using filters for host The minimum required peptide length was set to seven amino acids. All spectra
(human) and assays (MHC ligand assays only)53. To assess compatibility of were queried in two independent searches against the patient-specific or the SNP-
­recovered MHC-I ligands with the netMHCpan40 prediction algorithm, we tested derived databases as earlier described for the ligandome analysis. A false discovery
cross-prediction for two patients with completely private HLA-A, -B, and -C alleles. rate of 1% was required at both the peptide level and the protein level. For each
Affinities for peptides recovered from one patient were predicted against self or protein group, summed peptide intensities were used to estimate copies-per-cell
unrelated HLA-A, -B, -C alleles. abundance using the histone ruler approach56.
Correlation of MHC presentation with gene expression. To estimate gene Three subsets of proteins were compared with the complete set of all recovered
­expression, we used previous transcriptome profiling of MCL performed in proteins: (1) proteins for which at least one corresponding peptide was r­ ecovered
ref. 20. The median log2-transformed reads per kilobase of transcript per million from the MHC-I immuno-peptidome dataset; (2) proteins for which at least one
mapped reads (RPKM) from the 15 patients from that study was used for predicted corresponding peptide was recovered from the MHC-II immuno-peptidome
gene expression, and genes with a median log2(RPKM) >​0 were considered to be dataset; (3) proteins that were predicted to be mutated by exome sequencing and
expressed. Every recovered MHC-bound peptide was assigned the log2(RPKM) for which the predicted mutated peptide was identified by proteome analysis.
of its corresponding gene. The distributions of abundance for each of these subsets was compared with the
Total cell-line proteome sample preparation and high pH reversed-phase distribution of all proteins using a two-tailed Kolmogorov–Smirnov test for each
­fractionation for MS-analysis. One hundred million cells from each of two MCL cell line, given the unequal variance between groups.
cell lines (JEKO and L128) were used for protein extractions. In brief, cells were Inter-patient similarity for MHC presentation of peptide and genes. The
lysed in 8 M urea, 150 mM NaCl, 5 mM DTT, 50 mM Tris pH 8 supplemented with lists of unique peptides or genes presented by MHC-I and MHC-II for each
Complete Protease Inhibitor Cocktail tablet (Roche) and 1×​Halt Protease and patient were used to calculate inter-patient similarity. Peptides with a post-­
Phosphatase Inhibitor Cocktail (ThermoFisher Scientific). After lysis, the lysate translation ­modification were considered distinct from unmodified peptides (for
was centrifuged at 13,200 r.p.m. for 15 min, the supernatant was t­ ransferred to ­example, a peptide with a phosphorylated residue was not identical to the same
fresh tubes, and a second round of centrifugation was performed. The ­resulting ­unphosphorylated peptide). The Sørensen similarity index was calculated between
­supernatant was alkylated using 14 mM iodoacetamide for 45 min at room patients as 1 −​ (Bray–Curtis dissimilarity) using the R ‘vegan’ package based on
­temperature in the dark and the reaction was then quenched with d ­ ithiothreitol peptide or gene similarity. A force-directed graph was generated based upon
(DTT). Proteins were methanol–chloroform precipitated and washed twice with MHC peptide presentation similarity used in the R ‘igraph’ package. A weighted
acetone before being re-suspended in 300 μ​l 8 M urea, 50 mM Tris pH 8. Total ­adjacency matrix was created and plotted with a Fruchterman–Reingold layout. For
protein concentration was determined using a Pierce BCA Protein Assay Kit ­visualization, a minimum similarity of 0.15 was set as a threshold to produce an edge.
(Pierce, Rockford, Illinois, USA). The samples were diluted to 1 M urea using HLA allele families for each patient were manually superimposed over the layout
50 mM Tris pH 8 before digestion with trypsin/Lys-C Mix (Promega, Madison, to highlight the relationship between peptide similarity and shared HLA alleles.

© 2017 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
RESEARCH LETTER

The identified MHC-associated peptide repertoires showed relatively low beads and co-stained with both anti-HLA-DR and anti-DNP antibodies. Beads
inter-patient similarity (MHC-I: Sørensen similarity 0.01–0.45, median 0.12; were then analysed by flow cytometry. One peptide that was not computationally
MHC-II: Sørensen similarity 0.05–0.55, median 0.17), but similarity was predicted to bind either of the patient’s HLA-DR alleles (IKNKIDGETTDYAAPV)
increased between patients with shared or similar HLA alleles. Genes represented was found to have high affinity for HLA-DR*​0401, highlighting the sensitivity of
in the a­ ntigen repertoire were more similar between patients (MHC-I: Sørensen proteomic methods for MHC ligand discovery.
­similarity 0.27–0.60, median 0.41; MHC-II: Sørensen similarity 0.04–0.48, HLA-DR tetramer staining and single-cell sequencing. Total peripheral blood
median 0.26). mononuclear cells (PBMCs) or purified CD4+ T cells were treated with 50 nM
Gene set enrichment of MHC-presented peptides. All recovered MHC-I bound dasatinib (CST) for 30 min at 37 °C to inhibit TCR downregulation. Samples were
peptides from all 17 patients were included. The total number of unique MHC- then stained with tetramers (0.5 μ​g/ml each specificity) and Fc block for 1 h at
presented peptides recovered per gene was used to rank gene ­presentation. If room temperature, with agitation. Antibodies and live/dead staining reagents
an identical peptide was recovered from multiple patients, it was treated as an were added during the last 30 min of tetramer staining. Samples were washed, and
additional unique peptide mapping to that gene. The top fifth percentile of tetramer-positive CD4 T cells (and tetramer-negative control cells) were single-cell
genes ranked in this manner were used for gene set enrichment analysed by the sorted with a FACSAria sorter (BD Biosciences). The TCR and select phenotypic
PANTHER statistical overrepresentation test57. mRNA were amplified from each cell by nested PCR, the products were pooled
Comparison of germline SNP peptide presentation versus somatically mutated and purified, and sequenced using Illumina MiSeq as previously described22.
peptide presentation. Germline SNPs for all 17 patients were identified as above. For single-cell sequencing, reads were assembled, de-barcoded, and TCR α​- and
Recovered MHC-I- and MHC-II-associated peptides were matched with 100% β​-chains were called for each cell that met acceptable confidence thresholds using
identity against both the SNP variant allele and the corresponding reference allele a customized pipeline22. For phenotyping analysis, cells with at least ten reads of a
as listed in hg19. Peptide expression of both alleles was considered true if peptides particular mRNA were considered positive.
from both the SNP and corresponding reference allele were recovered in the same TCR-β repertoire sequencing. DNA was extracted from peripheral blood
patient. The same analysis was performed for non-Ig somatic SNVs. No non-Ig ­mononuclear cells, then TCR-β​CDR3 regions were amplified and sequenced using
somatically mutated peptides were presented by MHC-I or MHC-II. However, ImmunoSEQ (Adaptive Biotechnologies, Seattle, Washington, USA) from 400 ng
nine MHC-I peptides and nine MHC-II peptides from the corresponding germline of DNA template59. Bias-controlled V and J gene primers were used to amplify
allele were recovered, which overlapped the predicted mutated amino-acid rearranged V(D)J segments for high-throughput sequencing at ~​20×​ coverage.
­position. A 2 ×​ 3 Fisher’s exact test was used to compare SNP recovery versus SNV After correcting sequencing errors via a clustering algorithm, CDR3 segments
recovery (where the three outcomes were recovery of variant peptide, r­ ecovery of were annotated according to the International ImMunoGeneTics collaboration,
the reference peptide, and recovery of both reference and variant peptide). identifying which V, D, and J genes contributed to each rearrangement60.
Comparison between Ig variable versus constant region presentation. Unique HLA-A2 tetramer staining and preparation. HLA-A02:01 tetramers containing
MHC-presented peptides were aligned to the patients’ IgVh and IgHμ (Ig heavy an ultraviolet-cleavable peptide61 were synthesized by the NIH Tetramer Facility.
variable and heavy constant mu regions) sequences. IgVh sequences from all Potential HLA-A2-binding (predicted IC50 <​ 500 nM by netMHC62) neoantigen
patients were aligned to each other using IMGT nomenclature and gap insertion peptides from eight patients with HLA-A2 were synthesized (Sigma-Aldrich or
when required for discrepant lengths. For each position on the Ig sequence, the JPT Peptide Technologies), exchanged, and multimerized with streptavidin-PE as
numbers of unique peptides that overlapped that position were summed for all 17 previously described63. Vaccine-primed PBMCs were collected by apheresis. Cells
patients and presented as a heatmap. The numbers of experimentally r­ ecovered were pre-treated with 50 nM dasatinib for 60–180 min. To determine ­background
peptides and the number of predicted peptides were determined for each p ­ osition staining, we used T cells from leukocyte reduction system cones from two or
along the consensus Ig molecule. A two-sided Mann–Whitney–Wilcoxon test was three healthy HLA-A2 donors from the Stanford Blood Center. These cells were
used to compare the number of peptides per position for N-terminal ­(variable ­barcode-labelled with distinct levels of CellTrace Violet dye (ThermoFisher) to
region plus N-terminal 50 amino acids of CH1) versus C-terminal (last 350 enable multiplexing. Donor cells were treated with dasatinib in parallel and then
amino acids in constant regions) positions. This was performed separately for pooled with T cells from patients immediately before tetramer staining. Cells were
­experimentally identified peptides and predicted peptides. mixed so that the total number of pooled healthy donor cells equalled that of the
Prediction of Ig presentation. The full-length Ig heavy-chain sequence for each patient’s T cells. Background staining was determined as the highest frequency
patient was used as input into netMHCpan and predictions were performed for seen in a s­ ingle donor. Tetramer staining was otherwise performed as previously
all peptides with a length of 8–11 amino acids for each of the patients’ HLA-A, described63. HIV-gag SLYNTVATL peptide was used as a negative control, and
HLA-B, and HLA-C alleles. Two IC50 thresholds were evaluated: 50 nM and CMV-pp65 NLVPMVATV and Flu-M1 GILGFVFTL peptides were used as pos-
500 nM. For each Ig position, a patient was counted as presenting that position if itive controls.
at least one peptide spanning it was predicted to meet the threshold for self-MHC Neoantigen T-cell purification and expansion. Patient leukocytes were collected
prediction. The number of patients with predicted presentation of that position by leukapheresis approximately 2 weeks after the series of autologous tumour
was then summed, with a maximum value of 17 patients presenting a position. ­vaccinations. CD4-expressing T cells were purified by negative magnetic bead
We evaluated whether the reduced MHC-I presentation of the N-terminal selection using a MACS CD4+ T-cell isolation kit (Miltenyi Biotec). CD4 T cells
170-amino-acid region of the heavy chain (corresponding to the variable region were co-incubated with autologous PBMCs collected at a later time, at a ratio of 1:1.
plus N-terminal portion of the CH1 constant domain) was related to differences in Cells were cultured in a 1:1 mix of AIM-V media and RPMI1640 (Thermo Fisher)
the predicted presentation between this region and the rest of the constant regions. with 10% autologous plasma and 50 μ​M β​-mercaptoethanol. Neoantigen peptides
Peptide–MHC binding predictions were performed as above for each patient’s Ig or matching germline peptides were synthesized (ElimBio) and added to a final
heavy-chain sequence. The total number of predicted peptides for each position concentration of 10 μ​g/ml. For each of the three MHC-II peptide families sharing
was summed across all 17 patients for the N-terminal 170 amino acids and the a core sequence, a single 29- to 30-amino-acid peptide was constructed with the
C-terminal 400 amino acids. shared core sequence in the centre. As a positive control, cells were ­stimulated with
HLA-DR tetramer preparation. A baculovirus construct encoding Jun/Fos a mixture of pathogen-associated peptides, CEFT pool (JPT Peptide Technologies).
dimerization domain-modified HLA-DRB1*​04:01 with tethered CLIP peptide Cells were incubated for 30 h before FACS sorting. Cells were sorted on live,
was used to produce DR4 protein in insect cells. The product was FPLC purified, CD4+, CD137+ cells using a FACS Aria sorter (BD Biosciences). A total of 278
biotinylated, and peptide exchanged as described in ref. 58. Candidate peptides ­neoantigen-specific cells were collected and co-cultured with a pooled mix of
predicted to interact with DR4 were assayed for binding by flow cytometry, and 50,000 feeder PBMCs from three irradiated (40 Gy) allogeneic healthy donors
those that successfully exchanged into the peptide-binding groove were used in in 1:1 AIM-V/RPMI media with 10% pooled human AB sera (Gemini Bio),
subsequent class II tetramer studies. Peptide–MHC monomers were t­ etramerized β​-mercaptoethanol, 30 ng/ml anti-CD3 antibody (OKT3, eBiosciences), 1 μ​g/ml
using phycoerythrin (PE)- or allophycocyanin (APC)-linked streptavidin anti-CD28 antibody (CD28.2 eBiosciences), and 10 U/ml recombinant human
­(eBioscience), and residual non-tetramer components were eliminated with IL-2 (Peprotech). Media were supplemented every 1–3 days with media containing
biotin agarose (Sigma) and streptavidin agarose (ThermoFisher). The peptides fresh IL-2 and β​-mercaptoethanol. Every 7 days, cells were co-incubated with fresh
used in this study were MCL008_IgK1 APRLLIYDAANRATG, MCL008_IgH1 irradiated allogeneic feeder cells, anti-CD3 antibody, and anti-CD28 antibody.
KNQFSLELNSVTAADTA, MCL034_IgH1 GQVTISVDKSISTAYL, MCL041_ Confirmation of neoantigen T-cell specificity. After ~​2.5 weeks of expansion,
IgH1 NMLFLQMNSLKTEDT, MCL041_IgH2 IKNKIDGETTDYAAPV, and a fraction of the T-cell culture was rested for 5 days in 1:1 AIM-V/RPMI media
MCL041_IgH3 DDSKNMLFLQMNSLKTEDT. To test binding affinity to with 10% pooled human AB sera (Gemini Bio), β​-mercaptoethanol, but lacking
HLA-DR, peptides were also synthesized with an N-terminal dinitrophenyl (DNP) feeder cells, anti-CD3, anti-CD28, or rhIL-2. The T-cell cultures were labelled with
modification. After exchanging the peptides into HLA-DR as described above, the CellTrace Violet (Thermo Fisher), incubated with autologous PBMCs loaded with
biotinylated HLA-DR molecules were bound to streptavidin-coated microsphere a mix of pathogen-derived peptides, or a pool of neoantigen peptides, or a pool

© 2017 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
LETTER RESEARCH

of matching germline-derived peptides at a concentration of 10 μ​g/ml. Cells were 44. Hunt, D. F. et al. Characterization of peptides bound to the class I MHC
co-cultured for 24 h before antibody staining and FACS analysis. Cells undergoing molecule HLA-A2.1 by mass spectrometry. Science 255, 1261–1263 (1992).
intracellular staining were treated with monensin (Golgi Stop, BD Biosciences) for 45. Hunt, D. F. et al. Peptides presented to the immune system by the murine class
II major histocompatibility complex molecule I-Ad. Science 256, 1817–1820
6 h before antibody staining. (1992).
T-cell-mediated lymphoma cytotoxicity. Neoantigen or viral-specific T-cell 46. Barnstable, C. J. et al. Production of monoclonal antibodies to group A
­cultures were produced as above. After 3 weeks of expansion, activated T-cell erythrocytes, HLA and other human cell surface antigens-new tools for genetic
­cultures were mixed with autologous lymphoma cells at an effector to target ratio of analysis. Cell 14, 9–20 (1978).
10:1. Autologous lymphoma cells were labelled with CellTrace Violet dye (Thermo 47. Lampson, L. A. & Levy, R. Two populations of Ia-like molecules on a human B
cell line. J. Immunol. 125, 293–299 (1980).
Fisher). After 24 h, cells were stained with 7-AAD to determine cell death. Target 48. Rappsilber, J., Ishihama, Y. & Mann, M. Stop and go extraction tips for
lymphoma cells were gated by CellTrace Violet dye labelling. matrix-assisted laser desorption/ionization, nanoelectrospray, and LC/MS
Glycosylation site prediction. N-linked glycosylation motifs in patient Ig heavy- sample pretreatment in proteomics. Anal. Chem. 75, 663–670 (2003).
and light-chain variable regions were predicted on the basis of NXS/T motif. 49. Eng, J. K., McCormack, A. L. & Yates, J. R. An approach to correlate tandem
Briefly, we computationally searched for peptide sequences containing proline mass spectral data of peptides with amino acid sequences in a protein
database. J. Am. Soc. Mass Spectrom. 5, 976–989 (1994).
followed by a random amino acid and a serine or threonine. The first random
50. Zhang, J. et al. PEAKS DB: de novo sequencing assisted database search for
amino acid could not be proline. sensitive and accurate peptide identification. Mol. Cell Proteom. 11,
Data availability. Peptide data have been deposited in the PRIDE Archive M111.010587 (2012).
at www.ebi.ac.uk/pride/archive under accession numbers PXD004746 and 51. Käll, L., Canterbury, J. D., Weston, J., Noble, W. S. & MacCoss, M. J. Semi-
PXD005704. All other data are available from the corresponding authors upon supervised learning for peptide identification from shotgun proteomics
reasonable request. datasets. Nature Methods 4, 923–925 (2007).
52. Colaert, N., Helsens, K., Martens, L., Vandekerckhove, J. & Gevaert, K. Improved
visualization of protein consensus sequences by iceLogo. Nature Methods 6,
34. Li, H. & Durbin, R. Fast and accurate short read alignment with Burrows– 786–787 (2009).
Wheeler transform. Bioinformatics 25, 1754–1760 (2009). 53. Vita, R. et al. The immune epitope database (IEDB) 3.0. Nucleic Acids Res. 43,
35. Koboldt, D. C. et al. VarScan 2: somatic mutation and copy number alteration D405–D412 (2015).
discovery in cancer by exome sequencing. Genome Res. 22, 568–576 (2012). 54. Gilar, M., Olivova, P., Daly, A. E. & Gebler, J. C. Orthogonality of separation in
36. Koboldt, D. C., Larson, D. E. & Wilson, R. K. Using VarScan 2 for germline two-dimensional liquid chromatography. Anal. Chem. 77, 6426–6434 (2005).
variant calling and somatic mutation detection. Curr. Protoc. Bioinformatics 44, 55. Wang, Y. et al. Reversed-phase chromatography with multiple fraction
15.14.1–15.14.17 (2013). concatenation strategy for proteome profiling of human MCF10A cells.
37. Wang, K., Li, M. & Hakonarson, H. ANNOVAR: functional annotation of genetic Proteomics 11, 2019–2026 (2011).
variants from high-throughput sequencing data. Nucleic Acids Res. 38, e164 56. Wiśniewski, J. R., Hein, M. Y., Cox, J. & Mann, M. A “proteomic ruler” for protein
(2010). copy number and concentration estimation without spike-in standards.
38. Bai, Y., Ni, M., Cooper, B., Wei, Y. & Fury, W. Inference of high resolution HLA Mol. Cell. Proteomics 13, 3497–3506 (2014).
types using genome-wide RNA or DNA sequencing reads. BMC Genomics 15, 57. Mi, H., Poudel, S., Muruganujan, A., Casagrande, J. T. & Thomas, P. D. PANTHER
325 (2014). version 10: expanded protein families and functions, and analysis tools.
39. Lundegaard, C. et al. NetMHC-3.0: accurate web accessible predictions of Nucleic Acids Res. 44 (D1), D336–D342 (2016).
human, mouse and monkey MHC class I affinities for peptides of length 8–11. 58. Day, C. L. et al. Ex vivo analysis of human memory CD4 T cells specific for
Nucleic Acids Res. 36, W509–W512 (2008). hepatitis C virus using MHC class II tetramers. J. Clin. Invest. 112, 831–842
40. Nielsen, M. et al. NetMHCpan, a method for quantitative predictions of peptide (2003).
binding to any HLA-A and -B locus protein of known sequence. PLoS ONE 2, 59. Kirsch, I. R. et al. TCR sequencing facilitates diagnosis and identifies mature
e796 (2007). T cells as the cell of origin in CTCL. Sci. Transl. Med. 7, 308ra158 (2015).
41. van Dongen, J. J. et al. Design and standardization of PCR primers and 60. Lefranc, M. P. et al. IMGT, the international ImMunoGeneTics information
protocols for detection of clonal immunoglobulin and T-cell receptor gene system. Nucleic Acids Res. 37, D1006–D1012 (2009).
recombinations in suspect lymphoproliferations: report of the BIOMED-2 61. Rodenko, B. et al. Generation of peptide-MHC class I complexes through
Concerted Action BMH4-CT98-3936. Leukemia 17, 2257–2317 (2003). UV-mediated ligand exchange. Nature Protocols 1, 1120–1132 (2006).
42. Lefranc, M. P. et al. IMGT, the international ImMunoGeneTics database. Nucleic 62. Nielsen, M. et al. Reliable prediction of T-cell epitopes using neural networks
Acids Res. 27, 209–212 (1999). with novel sequence representations. Protein Sci. 12, 1007–1017 (2003).
43. Elias, J. E. & Gygi, S. P. Target-decoy search strategy for increased confidence in 63. Newell, E. W., Klein, L. O., Yu, W. & Davis, M. M. Simultaneous detection of many
large-scale protein identifications by mass spectrometry. Nature Methods 4, T-cell specificities using combinatorial tetramer staining. Nature Methods 6,
207–214 (2007). 497–499 (2009).

© 2017 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
RESEARCH LETTER

Extended Data Figure 1 | MHC ligand characteristics. a, Length the patient’s own HLA alleles (‘self ’, brown) or the HLA alleles of patient
distribution of recovered MHC-I peptides plotted alongside the completed MCL052 (‘nonself ’, blue) using netMHC. f, MHC-I ligands from patient
Immuno Epitope Database (IEDB) of human HLA ligands. b, Length MCL052 were analysed for predicted affinity to either the patient’s own
distribution of recovered MHC-I peptides plotted with IEDB human HLA alleles (‘self ’, brown) or the HLA alleles of patient MCL001 (‘nonself ’,
HLA ligand dataset. c, Weblogo from all 9-amino-acid MHC-I ligands blue). White breaks indicate a change in scale of the x axis. g, The number
for four representative patients. d, Overlap of MHC-I ligands for two of unique peptides presented by MHC-I across all patients was determined
patients with a completely distinct MHC-I profile (that is, no shared HLA- for each gene. The genes that produced the most unique peptides (top fifth
A, HLA-B, or HLA-C alleles). The overlap of peptides (left) and genes percentile) were analysed by gene set enrichment using the PANTHER
(right) presented by MHC-I for each patient is shown. e, MHC-I ligands pathways database, revealing enrichment of B-cell activation genes.
from patient MCL001 were analysed for their predicted affinity to either

© 2017 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
LETTER RESEARCH

Extended Data Figure 2 | Correlation of protein abundance with MHC-I *​*​*​P <​  10−9. b, The abundance of each protein was estimated using the
and MHC-II presentation. Two MCL cell lines, JEKO (left) and L128 histone ruler approach (black). Each protein with at least one peptide
(right), were digested with trypsin and peptides identified by LC–MS/MS. presented by MHC-I (blue) or MHC-II (red) is marked with an adjacent
a, Violin plots: distribution of protein abundance is plotted as kernel tick to the right and left (respectively) of the estimated protein abundance
density violin plots with mean value indicated by a bar for all proteins line. Proteins for which a mutated peptide was recovered by whole-
(grey), proteins presented by MHC-I (blue), proteins presented by MHC-II proteomic analysis (not from MHC immunoprecipitation) are indicated by
(red), and mutated proteins where a mutated peptide was identified from ticks on the left of the figure (green). Noteworthy genes involved in MCL
whole-proteome analysis (green). The distributions of protein abundance pathogenesis are highlighted. Proteins for which a mutated peptide was
for each set of proteins (MHC-I presented, MHC-II presented, and recovered by proteomic analysis are indicated by ticks on the left of the
mutated proteins) were compared with the distribution for all proteins figure.
by Kolmogorov–Smirnov test. For each line, *​P <​  10−3, *​*​P <​  10−5,

© 2017 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
RESEARCH LETTER

Extended Data Figure 3 | Comparison of antigen presentation between directed graph of Sørensen similarity (edges) between patients (nodes)
patients. a, b, Heatmap of Sørensen similarity coefficient between patients for MHC-I (e) or MHC-II (f). For MHC-I, patients with at least one HLA
for the set of genes presented by MHC-I (a) and MHC-II (b). Patients allele belonging to the HLA-A02 (pink), HLA-B44 (blue), or HLA-A03
were clustered by hierarchical clustering. Gene presentation by MHC (green) supertype family are coloured accordingly. For HLA-DR, patients
was true if one or more peptides encoded by the gene were presented by are coloured by presence of four specific HLA-DR alleles. Edge weight
MHC. c, d, Heatmap of Sørensen similarity index between patients for and colour are determined by strength of Sørensen similarity (minimum
peptide ligands presented from MHC-I (c) or MHC-II (d). Patients were Sørensen similarity 0.15 per edge). Node size is determined by number
clustered by hierarchical clustering. e, f, Two-dimensional visualization of MHC ligands for each patient. Nodes are coloured by membership to
of the similarity in MHC ligands between patients. Relationship between MHC-I supertype family (e) or HLA-DR allele (f).
samples is shown through a Fruchterman–Reingold layout of a force-

© 2017 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
LETTER RESEARCH

Extended Data Figure 4 | Presentation of heterozygous SNPs. Top, Overlap (purple) indicates an SNP locus for which both SNP and hg19
germline heterozygous non-synonymous SNPs were determined for all reference alleles were identified from the same patient. Bottom, analogous
17 patients. MHC-I- and MHC-II-associated peptides spanning each depiction of recovery of peptides containing a somatic variant (null set;
SNP were identified from our LC–MS/MS dataset. The number of SNP open circle) or the corresponding germline peptide of the variant (yellow
loci that produced an MHC-bound peptide bearing the respective SNP circle). Comparison of recovery for germline SNPs and somatic variants
is shown in red. The number of SNP loci that produced an MHC-bound was performed by Fisher’s exact test.
peptide bearing the corresponding hg19 reference allele is shown in blue.

© 2017 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
RESEARCH LETTER

Extended Data Figure 5 | Peptide–MHC tetramer T-cell responses MHC tetramer is shown. b, Frequency of neoantigen-specific CD8
against predicted HLA-A2 neoantigens. HLA-A2 restricted neoantigens T cells from peripheral blood (filled circles). The frequency of neoantigen-
were predicted for eight patients. Peptide–MHC tetramers were specific T cells from healthy donor PBMCs for each neoantigen is also
synthesized for 108 predicted HLA-A2 neoantigens. Patient peripheral shown (open squares). Staining of a CMV peptide–MHC tetramer for each
blood cells were pre-treated with dasatinib and then stained with each patient is shown in red. c, Representative results for three viral peptides
tetramer. a, To enable a more accurate estimation of background staining, derived from HIV, influenza A (flu), and CMV are shown along with
T cells from patients were mixed and co-stained with fluorescently patient-specific neoantigen staining. The neoantigen SLC9C2 tetramer is
barcoded cells from two or three healthy donors. A representative reaction shown as an example of a tetramer with high background staining of both
with multiplexing of three healthy donors using cytomegalovirus peptide– patient and healthy donor CD8 cells.

© 2017 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
LETTER RESEARCH

Extended Data Figure 6 | MHC-I presentation of lymphoma Ig against the Raji Ig heavy- and light-chain sequences. Positions differing
neoantigens in the Raji lymphoma cell line. a, Top, MHC-I binding from the germline variable region altered by either somatic hypermutation
affinity predictions were performed for all potential 9-amino-acid or V(D)J recombination are shown in red. Three peptides corresponding
peptides from the Raji Ig heavy chain. The best binding affinity for an to Raji Ig were identified, shown in boxes. All three were only found
endogenous class I MHC allele is shown in black. The predicted binding in the B35-transfected Raji cells. Asterisks indicate peptides that were
to an ectopically expressed HLA-B35:01 allele is shown in red. Bottom, identified proteomically. b, The predicted binding IC50 (nM) based on
the Raji lymphoma cell line was transfected with the HLA-B35:01 allele. netMHCpan is shown for each recovered peptide for each of the Raji HLA
MHC-I antigen presentation profiling was performed for both the parental alleles. Red shading indicates a predicted high-affinity interaction with the
cell line and the B35-transfected line. Recovered peptides were matched corresponding HLA allele.

© 2017 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
RESEARCH LETTER

Extended Data Figure 7 | Experimental determination affinity of co-stained with the anti-HLA-DR antibody and anti-DNP antibody. Beads
HLA-DRB1*04:01 with associated Ig neoantigens. Six neoantigen were then washed and analysed by flow cytometry for dual staining against
peptides identified from three patients were synthesized with an HLA-DR and DNP-labelled peptide. A known CMV-derived peptide
N-terminal DNP modification and tested for binding to recombinant ligand of HLA-DR4 was used as a positive control. Shown above each
HLA-DR4 molecules. Recombinant, biotinylated HLA-DR4 molecules plot is the predicted affinity of each peptide for both HLA-DR4 and the
were produced with a thrombin-cleavable CLIP peptide. Neoantigen associated patient’s alternative HLA-DR allele as predicted by netMHCII.
peptides were exchanged onto the DR4 molecules. HLA-DR4 molecules Red letters indicate amino acids that differ from the germline variable gene
were then bound to streptavidin-coated microsphere beads and sequence owing to somatic hypermutation events.

© 2017 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
LETTER RESEARCH

Extended Data Figure 8 | Phenotyping of neoantigen-specific T cells. low-dose IL-15. Cells were then fluorescently labelled and stimulated
a, Peripheral blood CD4+ T cells were isolated from patient MCL041 for 24 h with unlabelled autologous PBMCs loaded with a pool of three
and stained with HLA-DR*​04:01 tetramers loaded with patient-specific Ig neoantigen peptides, or a pool of three corresponding peptides with
neoantigen and a CMV peptide. Right, gated PD1 and CD45RA expression somatic alterations reverted back to the variable gene sequence, or a pool
is shown for neoantigen-specific CD4 T cells (top) and for non-specific of pathogen-associated peptides. Activation of the T cells was determined
CD4 T cells (bottom). b, Vaccine-primed CD4+ T cells from patient by induction of CD25 and CD69 in response to peptide stimulation.
MCL030 were stimulated with either a pool of neoantigen peptides or c, Neoantigen-specific CD4 T cells from patient MCL030 were expanded,
a pool of pathogen-associated (CMV, EBV, influenza, tetanus) peptides labelled, and re-stimulated as in b. Expression of CD25, Ki67, IL-4, and
and were sorted for CD137 upregulation. The sorted population was granzyme B is shown for CD4-gated T cells re-stimulated with neoantigen
expanded ex vivo for 2.5 weeks, then rested for 5 days in the presence of peptide-loaded PBMC.

© 2017 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
RESEARCH LETTER

Extended Data Figure 9 | Lack of cytotoxic activity by pathogen- sorted pathogen antigen-specific cells were expanded using anti-CD3,
specific T cells against autologous lymphoma cells. Left, CD4 T cells anti-CD28, IL-2, and allogeneic feeders for 3 weeks. The expanded T cells
were purified from patient MCL030 after autologous tumour vaccination. were co-cultured with fluorescently labelled autologous lymphoma cells
T cells were stimulated with autologous PBMCs loaded with a pool of for 24 h (top right). Background cell death of the lymphoma cells is also
pathogenic peptides including antigens derived from CMV, EBV, influenza shown (bottom right). Cytotoxicity of lymphoma cells was determined by
A, and tetanus. After 30 h, cells were sorted for CD137 expression. Right, 7-AAD uptake of the labelled population.

© 2017 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
LETTER RESEARCH

Extended Data Figure 10 | Predicted MHC-I presentation of lymphoma position across the Ig heavy chain is shown. b, For each position along the
Ig molecules. a, Left, peptides recovered from MHC-I purification were Ig molecule, the number of peptides that were experimentally recovered
mapped to the Ig heavy and light chain. The colour heatmap corresponds per position (left-hand y axis) was determined. Similarly, for each
to the number of peptides recovered at each position by LC–MS/MS. position, the number of patients (of 17 in total) with at least one peptide
Right, for each patient’s unique lymphoma Ig sequence and HLA profile, with predicted peptide–MHC affinity IC50 <​500 nM was determined
the predicted peptide-HLA affinity was calculated for all possible peptides (right-hand y axis). Positions from the variable region to the N-terminal
with a length of 8–11 amino acids created from their Ig using netMHCpan. 50 amino acids of CH1 (red) were compared with the rest of the constant
A heatmap illustrating the number of patients (from a total of 17) with region (green). P value was calculated using a Mann–Whitney test. Error
at least one peptide predicted to bind self-HLA (IC50 ≤​ 500 nM) at each bars show range, horizontal black bar shows median.

© 2017 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.

You might also like