25
Supplementary Materials for Personalized cancer vaccine effectively mobilizes antitumor T cell immunity in ovarian cancer Janos L. Tanyi, Sara Bobisse, Eran Ophir, Sandra Tuyaerts, Annalisa Roberti, Raphael Genolet, Petra Baumgartner, Brian J. Stevenson, Christian Iseli, Denarda Dangaj, Brian Czerniecki, Aikaterini Semilietof, Julien Racle, Alexandra Michel, Ioannis Xenarios, Cheryl Chiang, Dimitri S. Monos, Drew A. Torigian, Harvey L. Nisenbaum, Olivier Michielin, Carl H. June, Bruce L. Levine, Daniel J. Powell Jr., David Gfeller, Rosemarie Mick, Urania Dafni, Vincent Zoete, Alexandre Harari, George Coukos, Lana E. Kandalaft* *Corresponding author. Email: [email protected] Published 11 April 2018, Sci. Transl. Med. 10, eaao5931 (2018) DOI: 10.1126/scitranslmed.aao5931 This PDF file includes: Materials and Methods Fig. S1. Schema of the clinical study. Fig. S2. Analysis of DC vaccine product. Fig. S3. Immune response in patient CTE-0017. Fig. S4. Immune competency of patients. Fig. S5. Clinical response does not depend on prevaccination T cell gene expression. Fig. S6. Validation of CD8 + T cell responses against neoepitopes and wild-type peptides. Fig. S7. Representative example of the gating strategy applied for intracellular cytokine staining analyses. Fig. S8. TCRα V-J segments recombination landscape of sorted HHAT neoepitope-specific CD8 + T lymphocytes. Table S1. Adverse events detected in cohorts 1 to 3 Table S2. Comparative parameters in immune responder and nonresponder patients. Table S3. Gene set enrichment analysis between clinical responders and nonresponders does not find immune-related pathways differentiating both groups of patients before the vaccination. www.sciencetranslationalmedicine.org/cgi/content/full/10/436/eaao5931/DC1

Supplementary Materials for - stm.sciencemag.org · Supplementary Materials for Personalized cancer vaccine effectively mobilizes antitumor T cell immunity in ovarian cancer Janos

Embed Size (px)

Citation preview

Supplementary Materials for

Personalized cancer vaccine effectively mobilizes antitumor T cell

immunity in ovarian cancer

Janos L. Tanyi, Sara Bobisse, Eran Ophir, Sandra Tuyaerts, Annalisa Roberti,

Raphael Genolet, Petra Baumgartner, Brian J. Stevenson, Christian Iseli,

Denarda Dangaj, Brian Czerniecki, Aikaterini Semilietof, Julien Racle,

Alexandra Michel, Ioannis Xenarios, Cheryl Chiang, Dimitri S. Monos,

Drew A. Torigian, Harvey L. Nisenbaum, Olivier Michielin, Carl H. June,

Bruce L. Levine, Daniel J. Powell Jr., David Gfeller, Rosemarie Mick, Urania Dafni,

Vincent Zoete, Alexandre Harari, George Coukos, Lana E. Kandalaft*

*Corresponding author. Email: [email protected]

Published 11 April 2018, Sci. Transl. Med. 10, eaao5931 (2018)

DOI: 10.1126/scitranslmed.aao5931

This PDF file includes:

Materials and Methods

Fig. S1. Schema of the clinical study.

Fig. S2. Analysis of DC vaccine product.

Fig. S3. Immune response in patient CTE-0017.

Fig. S4. Immune competency of patients.

Fig. S5. Clinical response does not depend on prevaccination T cell gene

expression.

Fig. S6. Validation of CD8+ T cell responses against neoepitopes and wild-type

peptides.

Fig. S7. Representative example of the gating strategy applied for intracellular

cytokine staining analyses.

Fig. S8. TCRα V-J segments recombination landscape of sorted HHAT

neoepitope-specific CD8+ T lymphocytes.

Table S1. Adverse events detected in cohorts 1 to 3

Table S2. Comparative parameters in immune responder and nonresponder

patients.

Table S3. Gene set enrichment analysis between clinical responders and

nonresponders does not find immune-related pathways differentiating both groups

of patients before the vaccination.

www.sciencetranslationalmedicine.org/cgi/content/full/10/436/eaao5931/DC1

Table S4. Description of validated HLA class I neoepitopes identified.

Table S5. Protein Data Bank entries used as templates to model the TCRα, TCRβ,

MHC, β-microglobulin, and the peptide epitope.

Table S6. Molecular interactions between the peptide epitope (KQWLVWLFL)

and MHC HLA-A*0206, as predicted by homology modeling.

Table S7. Molecular interactions between the TCR and pMHC, as predicted by

homology modeling.

References (61–66)

Materials and methods

Characterization of OCDC vaccine product

Frozen DC vaccine products from several patients were thawed and directly stained with a

panel of markers to characterize their maturation: CD14-BV510 (Biolegend, 301842),

CD11c-BV711 (BD, 563130), HLA-DR-ECD (BC, IM3636), HLA-A,B,C-PerCP-Cy5.5

(Biolegend, 311420), CD40-BV605 (Biolegend, 334335), CD80-PE-Cy7 (BD, 561135),

CD83-FITC (BC, IM2410), CCR7-APC-Cy7 (Biolegend, 353211) and Zombie UV fixable

Viability Kit (Biolegend, 423108). The samples were acquired at the BD Fortessa instrument,

the data analysed with FlowJo v10.2 and the graphs were done with GraphPad Prism v7.03.

Detection of T-cell responses to whole-tumor antigen

To quantify T-cell responses against whole-tumor antigen, we interrogated peripheral blood

T cells against autologous activated DCs previously pulsed with autologous oxidized tumor

lysate, i.e. cryopreserved OCDC vaccine aliquots. Dissociated autologous tumor cells were

depleted from CD45+ cells using anti-CD45 magnetic cell sorting (Miltenyi) and were also

used as targets in downstream functional assays to activate PBL, when available. Responder

T cells were quantified using IFNγ ELISpot kits (Diaclone), according to the manufacturer's

instructions. In brief, cryopreserved PBMC were thawed and rested overnight in RPMI

supplemented with 8% human serum, (Biowest). Cryopreserved OCDC vaccine aliquots or

dissociated autologous tumor cells were thawed on the following day and plated with

250,000 PBMC per well in 10:1 PBMC/DC ratio or 100,000 PBMC per well in 1:1

PBMC/tumor cell ratio. PBMC used in these assay were freshly thawed cryopreserved cells

and were not expanded in vitro. Twenty-four hours later, the spots were developed and

counted automatically using the Bioreader 6000 (BioSys). Background reactivity was

determined using PBMC plus media alone and was subsequently subtracted in calculating

responding cells. For intracellular cytokine staining, cryopreserved PBMC were thawed and

rested overnight in RPMI supplemented with 8% human serum. Cryopreserved DCs were

thawed on the following day and plated with the PBMC in 1:10 PBMC/DC ratio. Following

co-incubation for 24 hours, ICS was performed as detailed above.

In vitro identification of neo-epitope specific T cells

Candidate neo-epitope peptides with a predicted binding affinity of ≤500 nM, and their wild-

type non-mutant predicted peptides were synthesized (at >90% HPLC purity) at the Protein

and Peptide Chemistry Facility (PPCF), University of Lausanne. Candidate peptides were

screened for T-cell recognition either as pools of ≤50 peptides or individually following

deconvolution of pool peptide reactivities. Cryopreserved PBMCs were thawed and used to

isolate CD3+CD8+ and CD3+CD4+ T cells using Dynabeads FlowComp Human kits

(Invitrogen), according to the manufacturer’s instructions. CD8+ T-cells were co-incubated in

p96-well plates (100,000 per well with ≥ 6 wells per condition) in RPMI supplemented with

8% human serum, (Biowest) with autologous irradiated CD8+- and CD4+-depleted PBMCs

and 1µg/mL of synthetic peptides at 1:1 ratio. Medium was initially supplemented with 20

IU/mL human recombinant interleukin 2 (IL-2, a generous gift by GlaxoSmithKline to Dr.

Daniel Speiser) and replaced after 48 hours with fresh medium containing 100 IU/mL IL-2.

On day 12, expanded T cells were collected and tested for peptide reactivity in standard IFN

Enzyme-Linked ImmunoSpot (ELISpot) and intracellular cytokine staining assays (see

below). Cross-reactivity to wild-type peptides was assessed to define T-cell specificity, and

peptide-MHC multimers were used to confirm specificity and class-I HLA restriction. CD4+

blasts were generated by stimulating isolated CD3+CD4+ T cells in RPMI supplemented with

8% human serum, PHA (1 μg/mL) and IL-2 (150 U/mL), and used as antigen-presenting cells

for T-cell reactivity analysis (61). When larger number of cells were requested, e.g. prior to a

cell sorting with multimers, positive cell cultures were further enriched by multiple

stimulation rounds. T-cell reactivity was validated by ≥ 2 independent experiments.

For IFN ELISpot, to assess the reactivity of T cells against candidate mutant

peptides, in-vitro stimulated CD8+ T cells were plated (0.5-1x105 cells per well) and re-

challenged for 16 hours in triplicates with autologous CD4+ blasts (1:1 ratio) in the presence

of peptide pools or single peptides at a final concentration of 1µg/mL. ELISpot assays were

performed using pre-coated 96-well ELISpot plates (Mabtech), following the manufacturer’s

instructions. Spot-forming units were counted using a Bioreader-6000-E automated counter

(BioSys). Positive conditions were defined as having more spots than negative control wells

(no peptide) plus 3-times the standard deviation.

For intracellular cytokine staining, in-vitro stimulated T cells were plated with

peptide-pulsed autologous CD4+ blasts in a 1:1 ratio and Golgiplug (BD biosciences, USA)

was added 2 hours later to the co-cultures. After 16-18 hours of re-stimulation with

peptide(s), ≥2x106 cells were harvested and stained with anti-CD3, anti-CD8, anti-CD4, anti-

IL-2, anti-TNFα, anti-IFNγ (BD biosciences) and with viability dye (Life technologies). Flow

cytometry was performed using LSR Fortessa (BD biosciences) and analyzed with FlowJo v9

(TreeStar) and SPICE 4.2.3 (developed by Mario Roederer, Vaccine Research Center,

NIAID, NIH), as described elsewhere(62, 63). The number of lymphocyte-gated events

ranged between 105 and 106 in the flow cytometry experiments shown. Background reactivity

was determined using PBMCs plus media alone, and was subsequently subtracted in

calculating responding cells.

In all patients, PBMCs were interrogated after a first round of in vitro stimulation

using peptide pool(s), to assess presence or absence of neo-epitope reactivity. A repeat single

round of in vitro stimulation was performed with fresh PBMCs in order to confirm reactivity

against specific peptides that were part of the original pool(s); to calculate the specific

frequency of neoepitope-specific T cells; and test their cross-reactivity against the wild-type

control peptide(s). For certain tumors, we needed a second round of in vitro stimulation to

confirm flow cytometry experiments (i.e. peptide-MHC multimer binding or intracellular

cytokine staining, both requiring large number of cells) and assessment of cross-reactivity

with native (wild-type) peptides. For assays such as avidity measurements and TCR

sequencing, T cells were sorted after the second round of in vitro stimulation with the

cognate peptide, and expanded as described below. Importantly, quantitative analyses related

to the frequencies of neo-epitope specific CD8 T cells were consistently performed with cells

obtained after one in vitro stimulation.

Assessment of the cytolytic ability of tumor-specific T cells in vitro and in vivo

NOD.Cg-Prkdcscid

Il2rgtm1WjlTg(HLA-A2.1)1Enge/SzJ mice (NSG; Jackson Laboratory) were

bred and maintained under pathogen-free conditions under animal protocols approved by the

Veterinary Service of Canton Vaud. All mice received β-estradiol (8.5 mg/ml) diluted in

drinking water during the course of the experiments. Patient-derived xenografts (PDX) were

generated from 2x106 dissociated tumor cells depleted of human CD45+ cells (Miltenyi) in

25% Matrigel and grafted subcutaneously into NSG mice. PDX tumors were propagated in

mice every 3 months. We generated tumor cell lines for the in vitro killing assays by

dissociating PDX tumors using type IV collagenase (1 mg/mL, Life Technologies), dispase 1

(1 U/ml, STEMCELL Technologies) and Dnase I (10 μg/mL, Sigma) in DMEM with 10%

FBS (Gibco; Life Technologies). Cryopreserved PBMC were thawed and stimulated for 10

days with CD3/CD28 beads (Miltenyi) in 1:1 bead/T-cell ratio in the presence of 150 U/ml

IL-2 (gift of GlaxoSmithKline). For the in vitro 51Cr-release killing assay, autologous tumor

cells were labeled with 51Cr. 1000 51Cr-labeled tumor cells were co-cultured for 4 hours with

CD3/CD28-expanded T cells at the ratios indicated in the figure legend, in the presence or

absence of 20μg/ml of anti-HLA-A,B,C w6/32 blocking antibody (Ebioscience USA). At the

end of the co-culture, supernatant was collected and analyzed for radio-reactivity using

Topcount Instrument (Perkin Elmer, USA). The percent of specific lysis was calculated as:

100× [(experimental-spontaneous release)/(total-spontaneous release)]. To test the in vivo

specificity of the anti-tumor T cells, PDX tumors were resected from the mice, dissociated

enzymatically, and 1x106 dissociated tumor cells were injected subcutaneously into new NSG

mice either alone or together with 7x106 autologous CD3/CD28-expanded T cells. Tumor

dimensions were longitudinally measured with calipers, and tumor volumes calculated as V =

(length × width2)/2, where length is greatest longitudinal diameter and width is greatest

transverse diameter.

Labelling, sorting and expansion of neo-epitope specific T-cells

PE-labeled multimers were manufactured in-house at the Lausanne Ludwig Institute branch

and used for T-cell staining as previously reported(64). Briefly, ≥2x106 T cells were

incubated with multimers for 45 minutes at 4°C, washed, then stained with anti-CD8

antibody (APC, Biolegend), and finally stained for dead-cell exclusion with L/D stain kit

(Life Technologies) according to the manufacturer’s instructions. Multimer-positive cells

were sorted using a FACSAria IIu (Beckton Dickinson; purity >95%) and used for TCR

sequencing immediately after sorting or expanded using a rapid expansion protocol (REP).

To this end, cells were plated in p96-well plates and stimulated with irradiated feeder cells

(PBMC from 2 healthy donors) in RPMI supplemented with 8% human serum, PHA (1

μg/mL) and IL-2 (150 U/mL). Fresh medium was replaced every 2-3 days. At the end of the

REP, the purity of multimer-positive cells was >95%.

Cloning of neo-epitope specific T-cells

Neo-epitope (HHAT) specific CD8 T cells from patient CTE-0013 were sorted by flow

cytometry using PE-labeled multimer (TCmetrix), cloned by limiting dilution and expanded

in RPMI-1640 medium supplemented with 8% human serum, 150 U/mL recombinant human

IL-2, 1 μg/mL phytohemagglutinin (PHA), and irradiated (30 Gy) allogeneic PBMCs as

feeder cells as described (65).

Functional avidity analysis

Peptide stimulations were performed as described above. Functional avidity of neo-epitope-

specific CD8 T-cell responses was assessed by performing limiting peptide dilutions (ranging

from 30 µg/ml to 0.3 pg/ml) in in vitro IFN ELISpot assays as described(62) with 1000

effector cells per well. The peptide concentration required to achieve a half-maximal IFNγ

response (EC50) was determined.

Analysis of serum cytokines

Serum samples were thawed and evaluated for IFNγ levels using Meso scale discovery assay

(MSD) according to the manufacturer instructions. TGFβ levels in serum were analyzed

using an ELISA kit (DRG diagnostics) according to the manufacturer instructions.

TCR sequencing

Total RNA was isolated using the RNeasy Micro Kit (Qiagen) and mRNA was then

amplified using the MessageAmp II aRNA Amplification Kit (Ambion) with the following

modifications: 500 ng of total RNA was used as starting material. The in vitro transcription

was performed at 37°C for 16h. First strand cDNA was synthesized using the Superscript III

(Thermofisher) and a collection of TRAV/TRBV specific primers. TCRs were then amplified

by PCR (20 cycles with the Phusion from NEB) with a single primer pair binding to the

constant region and the adapter linked to the TRAV/TRBV primers added during the reverse

transcription. A second round of PCR (25 cycles with the Phusion from NEB) was performed

to add the Illumina adapters containing the different indexes. The TCR products were

purified with AMPure XP beads (Beckman Coulter), quantified and loaded on the MiniSeq

instrument (Illumina) for deep sequencing of the TCR/TCR chain. The TCR sequences

were further processed using ad hoc Perl scripts to: (i) pool all TCR sequences coding for the

same protein sequence; (ii) filter out all out-frame sequences; (iii) determine the abundance

of each distinct TCR sequence. TCR with a single read were not considered for the analysis.

Modelling the structure of pre- and on-treatment TCR-pMHC complexes.

The protocol used to model the TCR-p-MHC complexes was adapted from our TCRep 3D

approach (1). Starting from V and J segment identifiers and from the CDR3 sequences, the

full sequence of the constant and variable domains of TCR and TCR were reconstituted

based on IMGT/GENE-DB reference sequences (2). For each time-point, dominant TCR

and TCR chains were paired and analyzed, as reported in Table S5. Homology models of

the TCR-p-MHC complexes were obtained using the Modeller program (3, 4), version 9.

Template experimental structures were taken from the Protein Data Bank (5), and selected

based on the sequence similarity to the different components of the complexes, i.e. peptide,

MHC, -microglobulin, TCR and TCR (Table S5). Sequence alignments between the

target and template proteins were obtained using the MUSCLE (6, 7) program. 500 models

were produced for each TCR:peptide-MHC complex, and ranked according to the Modeller

Objective Function. The best ranked model was selected for CDR loop refinement. The latter

was performed by creating 4 x 500 alternative loop conformations using the “loop modeling”

module of Modeller. During this refinement, loops were treated by pairs as follows: TCR

CDR1 and CDR3 were optimised simultaneously by creating 500 loop conformations (while

other CDR loops were held fixed), followed by TCR CDR1 and CDR2, TCR CDR1 and

CDR3 and finally TCR CDR1 and CDR2, in this order. After each of these four loop

refinement steps, all models were ranked according to the Molecular Mechanics –

Generalized Born Surface Area (MM-GBSA) score we used previously to perform TCR

engineering (8-10). The total energy of the system was calculated using the CHARMM27

(11) force field, and the CHARMM v39 molecular mechanics package (12). The electrostatic

solvation free energy was calculated using the GB-MV2 (13) implicit solvent model, with a

dielectric of 1 and 80 for the protein and solvent, respectively, and no cut-off on the non-

bonded terms. The non-polar solvation energy was estimated by weighting the solvent

accessible surface area calculated analytically with CHARMM (with a probe radius of 1.4 Å)

by a 0.0072 kcal/mol/Å2 surface tension. After each step of loop refinement, the model with

the most favourable MM-GBSA energy was selected for the next step. Molecular graphics

and analyses were performed with the UCSF Chimera package (14).

Gene expression profiling

Gene expression data (RNA-seq) from pre-vaccination tumor biopsies was normalized to

log2-counts per million with help of the functions calcNormFactors from the R-package

edgeR version 3.16.5 (15) and voom from the package limma version 3.30.13 (16).

Differential expression for all genes between the vaccine responders (patients with stable

disease or partial response) and the non-responders (patients with progressive disease) was

then performed with help of DESeq2 (17).Genes were ranked based on the fold change

between the two groups and pathways differentially expressed between the two groups were

searched for with help of gene set enrichment analysis using the GSEA software version

3.0(65, 66). All pathways found in the “Canonical pathways” gene sets (version 6.1) were

tested for enrichment and only those with an FDR below 0.1 are reported in Table S3.

Supple

Fig. S1

Patients

given u

mainten

mentary Fi

. Schema o

s were enrol

under stud

nance.

igures

of the clinic

lled in one

dy NCT011

cal study.

of the three

132014. C

e cohorts. V

hemo: che

Vaccination a

emotherapy

and vaccine

; V: vacc

e maintenan

ine; Vm:

nce were

vaccine

Fig. S2

A) Exp

represen

monocy

differen

(MFI) o

. Analysis o

pression of

ntative exam

ytes (blue).

nt markers

of DC matur

of DC vacc

different m

mples of th

B) The grap

characteris

ration mark

cine produc

markers we

he expressio

ph shows th

stic for DC

ker (n=6).

ct.

ere analysed

on of the m

he percentag

C maturatio

d by flow

markers on D

ges of DC v

n (n=6). C

cytometry.

DC vaccine

vaccine pro

C) Mean flu

Histogram

e product (r

duct expres

uorescent i

ms show

red) and

ssing the

intensity

Fig. S3

A) Fre

autolog

pre- an

CD4+ T

and on

analyse

any in v

. Immune r

equency of

gous oxidize

nd on-vaccin

T cell respo

n-treatment

es (A and B

vitro expans

response in

f IFNγ-secr

ed tumor ly

nation (No

nse to auto

in patient

B) were perf

sion.

n patient CT

reting PBM

ysate measu

Ag: no pe

logous DCs

CTE-0017

formed with

TE-0017.

MC in resp

ured by IFN

eptide, SEB

s pulsed wi

. The gatin

h cryoprese

ponse to a

Nγ ELISpot

B: positive c

ith autologo

ng strategy

erved cells

autologous

detected in

control). B

ous tumor ly

y is shown

directly aft

DCs pulse

n patient CT

) Blood CD

ysate analyz

in Fig S7

er thawing

ed with

TE-0017

D8+ and

zed pre-

7. These

without

Fig. S4. Immune competency of patients.

General immune competency of bulk peripheral blood mononuclear cells (PBMC) from

vaccine-responders and non-responders patients assessed both pre-treatment (baseline, BSL)

and at the end of study (EOS). Data show the mean±SEM number of spot-forming units

(SFU) per 105 PBMCs as assessed by IFNγ ELISpot. N=6 for each group.

Baseline End of Study

IFNγ producing cells

per 1

05 PBMC

Fig. S5

Gene e

between

respond

signific

5. Clinical

expression o

n clinical r

ders (patient

cance.

response d

of specific

responders

ts with prog

does not de

T-cell gen

(patients w

gressive dis

epend on p

nes in tumo

with stable

sease). Wilc

prevaccina

or biopsies

disease o

coxon tests w

ation T cell

pre-vaccin

r partial re

were used t

l gene exp

nation is co

esponse) an

to test for st

ression.

ompared

nd non-

tatistical

Fig. S6. Validation of CD8+ T cell responses against neoepitopes and wild-type peptides.

Validation of neoepitope-specific T cell responses in IFNγ ELISpot assay. For each

neoepitope, wild-type (native) peptides are also shown. PHA was used as positive control

and IFNγ ELISpot T-cell responses were saturared (i.e. not quantitative and identified with

the “>” symbol). Peptide sequences are listed in Table S4.

No Ag

SEMA3G

mut

SEMA3G

wt

PHA 0

20

40

60

80

IFN

prod

ucin

g ce

lls/1

05 C

D8+

T c

ells

CTE‐0008

No Ag

TRIM26

mut

TRIM26

wt

PHA 0

500

1000

IFN

prod

uci

ng

cells

/10

5 C

D8+

T c

ells

No Ag

CHD3 m

ut

CHD3 wt

PHA 0

500

1000

1500

IFN

prod

uci

ng

cells

/10

5 C

D8+

T c

ells

No Ag

DCHS1 m

ut

DCHS1 wt

PHA 0

500

1000

IFN

prod

ucin

g ce

lls/1

05 C

D8+

T c

ells

No Ag

EBF4 m

ut

EBF4 wt

PHA 0

20

40

60

80

100

120

140

IFN

prod

ucin

g ce

lls/1

05 C

D8

+ T

cel

ls

CTE‐0010

No Ag

OR2T3

mut

OR2T

3 wt

PHA 0

100

200

300

400

500

IFN

prod

ucin

g c

ells

/105

CD

8+

T c

ells

>

No Ag

ZCCHC11 m

ut

ZCCHC11 w

t

PHA 0

100

200

300

400

500

IFN

prod

uci

ng c

ells

/105

CD

8+ T

cel

ls

>

No Ag

TNRC6A m

ut

TNRC6A w

t

PHA 0

1000

2000

3000

IFN

prod

ucin

g ce

lls/1

05 C

D8+

T c

ells

No Ag

SEPT9 m

ut

SEPT9 wt

PHA 0

400

800

1200

1600

IFN

prod

ucin

g ce

lls/1

05 C

D8

+ T

cel

ls

CTE‐0011

No Ag

HHAT m

ut

HHAT wt

PHA 0

20

40

60

80

100

IFN

prod

ucin

g c

ells

/105

CD

8+

T c

ells

>

CTE‐0013

CTE‐0015 CTE‐0019

No Ag

ODZ3 m

ut

ODZ3 wt

PHA 0

50

100

150

200

250

IFN

pro

duci

ng c

ells

/10

5 C

D8+

T c

ells

No Ag

P2RX5

mut

P2RX5

wt

PHA 0

200

400

600

800

1000

IFN

prod

ucin

g ce

lls/1

05

CD

8+ T

cel

ls

No Ag

PLK3

mut

PLK3

wt

PHA 0

5

10

15

20

IFN

prod

ucin

g ce

lls/1

05 C

D8+

T c

ells >

No Ag

DNAAF2 m

ut

DNAAF2 wt

PHA 0

5

10

15

20

IFN

prod

ucin

g ce

lls/1

05 C

D8

+ T

cel

ls >

Fig. S7

staining

The figu

positive

granulo

exclusio

respecti

are show

. Represen

g analyses.

ure shows a

e control. Th

osity, exclus

on of resp

ively CD8-

wn for both

ntative exam

an example

he different

sion of dou

pectively C

and CD4-p

h CD8 (left)

mple of the

of cytokine

t gating step

ublets, exclu

CD4- and C

positive cell

and CD4 (r

e gating stra

e production

ps correspon

usion of de

CD8-positiv

ls. Finally,

right) viable

ategy appli

n following

nd to a lymp

ad cells, se

ve cells fo

IFN versu

e T cells.

ied for intr

g stimulation

phocyte gat

election of C

ollowed by

us IL-2 and

racellular c

n with SEB

te based on

CD3-positiv

y the selec

IFN versu

cytokine

used as

size and

ve cells,

ction of

us TNF

Fig. S8

specific

TCRalp

CD8+ T

on-treat

are repr

each rec

8. TCRα V

c CD8+ T ly

pha V-J se

T lymphocyt

tment were

resented acc

combination

V-J segmen

ymphocytes

egments rec

tes from pa

FACS sort

cording to th

n is shown

nts recomb

s.

combination

atient CTE-0

ed using pe

heir chromo

on the z-axi

bination la

n landscape

0013 (neo-e

eptide-MHC

osomal loca

is and highl

ndscape of

e of sorted

epitope spec

C multimer c

ation on the

lighted by c

f sorted H

d HHAT ne

cific T-cell

complexes)

x and y-ax

colors.

HHAT neoe

eo-epitope

responses p

). V and J se

is; the frequ

epitope-

specific

pre- and

egments

uency of

Table SS1. Adversee events detected in coohorts 1 to 3

Table S2. Comparative parameters in immune responder and nonresponder patients.

Number of baseline peripheral blood monocyte or lymphocyte counts between responder and

non-responder patients, number of DC doses produced and output DC phenotype.

Table S3. Gene set enrichment analysis between clinical responders and nonresponders

does not find immune-related pathways differentiating both groups of patients before

the vaccination. GSEA was performed as described in the Methods section. Upregulated

(dark grey) corresponds to pathways with genes more expressed in the responders than non-

responders and the “downregulated” (light grey) corresponds to genes more expressed in the

non-responders than responders.

NAME SIZE ES NES NOM p‐val FDR q‐val FWER p‐val RANK AT MAX

REACTOME_INTERFERON_ALPHA_BETA_SIGNALING 48 0.6418454 2.3285804 < 0.0002 < 0.005 < 0.005 3598 tags=60%, list=19%, signal=75%REACTOME_PHASE1_FUNCTIONALIZATION_OF_COMPOUNDS 47 ‐0.5317925 ‐1.9112967 4.06E‐04 0.08660447 0.273 2671 tags=40%, list=14%, signal=47%

PID_S1P_S1P1_PATHWAY 21 ‐0.6445061 ‐1.9275385 0.002434077 0.08743567 0.229 4414 tags=67%, list=24%, signal=87%KEGG_O_GLYCAN_BIOSYNTHESIS 25 ‐0.62068397 ‐1.947337 0.001658375 0.09008205 0.1804 2933 tags=48%, list=16%, signal=57%

KEGG_PEROXISOME 71 ‐0.4870827 ‐1.8916212 < 0.0002 0.09059492 0.3322 3828 tags=38%, list=21%, signal=48%

LEADING EDGE

Table S

immuno

same m

(higher

characte

HLA c

substitu

algorith

determi

stimulat

treatme

and tho

magnitu

S4. Descrip

ogenic neo-

mutated seq

frequency

erization. P

class-I rest

utions); pre

hm); as we

ined by eith

tion) are sh

ent are show

ose only det

udes.

ption of va

-epitopes id

quence were

y of respo

Patient iden

triction; pe

edicted affi

ell as frequ

her IFNγ EL

hown. All th

wn in red, th

tected on- t

alidated H

dentified. Pe

e excluded

onding CD

ntification n

eptide sequ

inity of ne

uency of n

LISpot or m

he neo-epito

hose detect

treatment ar

HLA class I

eptides ove

from the l

D8+ T cel

number; mu

uence (bold

eo-epitope

neo-epitope

multimer sta

ope specific

ted both pre

re shown in

I neoepitop

rlapping fo

list while u

lls) were

utated gene;

d and und

(binding s

specific C

aining after

CD8+ T-ce

e- and on-tr

n green. Co

pes identifi

or more than

unique dom

chosen fo

; specific m

derlined are

score predi

CD8+ T ce

one or two

ell responses

reatment are

olor intensit

ied. List of

n 5 residue

minant neo-e

or validati

mutation ide

e the amin

icted by N

ells (percen

o rounds of

s only detec

e shown in

ty refers to

f all the

s on the

epitopes

on and

entified;

no acid

NetMHC

ntage as

specific

cted pre-

yellow,

relative

Table S5. Protein Data Bank entries used as templates to model the TCRα, TCRβ,

MHC, β-microglobulin, and the peptide epitope.

Table S6. Molecular interactions between the peptide epitope (KQWLVWLFL) and

MHC HLA-A*0206, as predicted by homology modeling. bb: backbone, sc: side chain; hb:

hydrogen bond, io: ionic interaction; np: non-polar interaction, :-interaction, c cation-

interaction. Unless indicated, interactions are taking place between side chains.

Table S7. Molecular interactions between the TCR and pMHC, as predicted by

homology modeling. bb: backbone, sc: side chain; hb: hydrogen bond, io: ionic interaction;

np: non-polar interaction; -: - interaction

Pre-vax

TCR Peptide MHC

D1‐TCR(111105)‐TCR(139954) CDR1 Ser30 hb‐Thr163, hb‐Glu166 CDR1 Ser31 (bb) hb‐Thr163 CDR1 Thr32 np‐Leu4

CDR1 Tyr33 hb‐Trp3 hb‐Glu154 (bb)

CDR2 Tyr50 hb‐Glu166

CDR2 Phe52 np‐Ala158

CDR2 Ser53 hb‐Thr163

CDR3 Asp96 io‐Arg65, io‐Lys66

CDR1 His29 ‐Phe8 CDR1 Ala30 np‐Phe8 CDR2 Gln50 (bb) hb‐Gln72 CDR2 Asn51 hb‐Gln72 (bb) CDR2 Asn52 hb‐Gln72 CDR2 Val55 np‐Ala69, np‐Arg65 CDR2 Asp56 io‐Arg65 CDR3 Phe99 np‐Val5

D1‐TCR(79822)‐TCR(139954) CDR1 Ser29 hb‐Glu58 CDR1 Glu30 io‐Lys1 io‐Arg65, io‐Lys66 CDR1 Ser31 hb‐Trp167 CDR1 Asp32 np‐Leu4 hb‐Thr163 CDR1 Tyr34 hb‐Glu154, hb‐Gln155 CDR2 Tyr55 np‐Ala158, hb‐Glu166 or hb‐Glu154 + hb‐

Gln155 CDR2 Lys56 io‐Glu154

CDR3 Ser97 np‐Leu4 hb‐Arg65 CDR3 His98 np‐Leu4, np‐Val5 np‐Ala69 CDR3 Asp99 io‐Arg65 CDR1 His29 ‐Phe8 CDR1 Ala30 np‐Phe8

CDR2 Gln50 (bb) hb‐Gln72 CDR2 Asn51 hb‐Gln72(bb) CDR2 Asp56 io‐Arg65

CDR2 Val55 np‐Ala69, np‐Arg65

CDR3 Phe99 np‐Val5

Post-vax

TCR Peptide MHC

D83‐TCR(102501)‐TCR(311993) CDR1 Ser29 hb‐Glu58 CDR1 Glu30 io‐Lys1 io‐Arg65, io‐Lys66 CDR1 Ser31 hb‐Trp167 CDR1 Asp32 np‐Leu4 hb‐Thr163 CDR1 Tyr34 hb‐Glu154, hb‐Gln155 CDR2 Tyr55 np‐Ala158, hb‐Glu166 or hb‐Glu154 + hb‐

Gln155 CDR2 Lys56 io‐Glu154 CDR3 Ile97 np‐Leu4 CDR3 Asn98 hb‐Leu4(bb), hb‐Val5(bb), hb‐Trp6(bb) hb‐Arg65 CDR3 Asp99 io‐Arg65 CDR1 Thr32 np‐Phe8 hb‐Gln72 CDR2 Glu53 (bb+sc) io‐Arg75 CDR2 Glu60 io‐Arg65 CDR3 Val98 np‐Phe8 CDR3 Val100 np‐Phe8, np‐Leu7 np‐Ala150 CDR3 Asn101 np‐Ala150

D83‐TCR(102501)‐TCR(142889) CDR1 Ser29 hb‐Glu58 CDR1 Glu30 io‐Lys1 io‐Arg65, io‐Lys66 CDR1 Ser31 hb‐Trp167 CDR1 Asp32 np‐Leu4 hb‐Thr163 CDR1 Tyr34 hb‐Glu154, hb‐Gln155 CDR2 Tyr55 np‐Ala158, hb‐Glu166 or hb‐Glu154 + hb‐

Gln155 CDR2 Lys56 io‐Glu154 CDR3 Ile97 np‐Leu4 CDR3 Asn98 hb‐Leu4(bb), hb‐Val5(bb) hb‐Arg65 CDR3 Asp99 io‐Arg65 CDR1 Asp26 io‐Arg82 CDR1 Met27 np‐Val76, np‐Gly79 CDR1 His29 ‐Phe8 np‐Gln72 CDR2 Tyr48 io‐Glu19, np‐Gln72, hb‐Arg75 CDR3 Ser94 np‐Phe8 CDR3 Pro95 np‐Phe8 CDR3 Ser96 Hb‐Trp6(bb) CDR3 Phe97 Np‐Leu7 np‐Ala150