ChAd mRNA Solid Tumors
ChAd mRNA Solid Tumors
ChAd mRNA Solid Tumors
https://doi.org/10.1038/s41591-022-01937-6
Checkpoint inhibitor (CPI) therapies provide limited benefit to patients with tumors of low immune reactivity. T cell-inducing
vaccines hold promise to exert long-lasting disease control in combination with CPI therapy. Safety, tolerability and recom-
mended phase 2 dose (RP2D) of an individualized, heterologous chimpanzee adenovirus (ChAd68) and self-amplifying mRNA
(samRNA)-based neoantigen vaccine in combination with nivolumab and ipilimumab were assessed as primary endpoints in an
ongoing phase 1/2 study in patients with advanced metastatic solid tumors (NCT03639714). The individualized vaccine regi-
men was safe and well tolerated, with no dose-limiting toxicities. Treatment-related adverse events (TRAEs) >10% included
pyrexia, fatigue, musculoskeletal and injection site pain and diarrhea. Serious TRAEs included one count each of pyrexia,
duodenitis, increased transaminases and hyperthyroidism. The RP2D was 1012 viral particles (VP) ChAd68 and 30 µg sam-
RNA. Secondary endpoints included immunogenicity, feasibility of manufacturing and overall survival (OS). Vaccine manu-
facturing was feasible, with vaccination inducing long-lasting neoantigen-specific CD8 T cell responses. Several patients with
microsatellite-stable colorectal cancer (MSS-CRC) had improved OS. Exploratory biomarker analyses showed decreased circu-
lating tumor DNA (ctDNA) in patients with prolonged OS. Although small study size limits statistical and translational analy-
ses, the increased OS observed in MSS-CRC warrants further exploration in larger randomized studies.
C
ancer immunotherapies have shown promise in harnessing Accordingly, peptide-based neoantigen vaccine platforms have
the immune system to target and destroy cancers, leading to to date failed to consistently induce robust neoantigen-specific
clinical benefit enriched in patients with a high mutational CD8 T cell responses in the majority of patients13–15. Although
burden1–5. Multiple studies indicate that cytotoxic CD8 T cells tar- more immunogenic, a homologous prime boost messenger RNA
geting tumor neoantigens are critical to tumor control and clear- (mRNA)-based vaccination approach elicited predominantly CD4
ance in response to immunotherapies targeting CTLA-4 or PD-16–10. T cell responses16–18. Cumulatively, previous findings suggest that a
Clinical responses to CPI therapy rely mostly on reinvigorating pre- successful cancer vaccine should (1) target tumor-specific neoanti-
existing tumor-specific T cell responses11, and active vaccination gens, (2) use highly immunogenic vaccine platform(s), (3) expand
to expand preexisting and prime de novo tumor-specific T cells is and prime T cells, (4) be combined with CPI therapy19 and (5) gen-
anticipated to overcome this limitation. erate long-term memory responses to ensure continuous tumor
The limited success of cancer vaccines in the past can be attrib- control for durable clinical benefit.
uted to a number of factors, including selection of poorly immuno- Viral vector-based vaccine platforms, such as recombinant ade-
genic self-antigens12, insufficiently immunogenic vaccine platforms novirus, are able to prime robust T cell responses20–24. Although high
and immunosuppressive milieus in patients with advanced cancers4. seroprevalence of anti-adenoviral antibodies in human populations
Gritstone bio, Inc., Emeryville, CA, USA. 2Virginia Cancer Specialists, Virginia Cancer Specialists, VA, Fairfax, USA. 3The Ohio State University Medical Center,
1
Columbus, OH, USA. 4Columbia University Medical Center, New York, NY, USA. 5Peter MacCallum Cancer Centre, Melbourne, VIC, Australia. 6Mayo Clinic,
Phoenix, AZ, USA. 7Mayo Clinic Cancer Center, Rochester, MN, USA. 8Memorial Sloan Kettering Cancer Center, New York, NY, USA. 9The University of Texas
MD Anderson Cancer Center, Houston, TX, USA. 10University of Chicago Comprehensive Cancer Center, Chicago, IL, USA. ✉e-mail: [email protected]
can lead to diminished immune responses to antigens delivered via memory T cells (Extended Data Fig. 1b). Accordingly, interferon-γ
adenoviral-based vaccines20,25, preexisting anti-vector immunity can (IFN-γ) ELISpot at week 106 revealed positive responses to four or
be circumvented by using non-human adenoviral species, such as more of the SIV antigens in all six animals, and samRNA adminis-
chimpanzee adenovirus (ChAd). RNA virus-based vaccines have tration at week 106 resulted in an ~8-fold and ~17-fold increase in
been tested in humans in a range of cancer and infectious disease antigen-specific T cell response detectable at week 107 ± s.c. ipilim-
indications26,27, and samRNA vectors are of particular interest due to umab, respectively (Fig. 1d and Extended Data Fig. 1c). Finally, the
their ability to express high and durable antigen levels28. ability of the boosted antigen-specific CD8 T cells to kill target cells
In this study, the quality of antigen-specific T cell responses presenting SIV epitopes was determined. CD8 T cells from all six
induced by a heterologous vaccine regimen consisting of a ChAd animals showed cytotoxic activity against their cognate targets, with
vector (ChAd68) and a fully synthetic Venezuelan equine encepha- CD8 T cells from four of six animals achieving killing of >70%, even
litis virus-based samRNA vector was assessed in non-human pri- at the lower effector-to-target ratio of 10:1 (Extended Data Fig. 1d).
mates (NHPs) and patients with advanced cancers in combination
with CPIs. The potency and longevity of the immune response Individualized vaccine regimen is safe and well tolerated. After
against simian immunodeficiency virus (SIV) model antigens29 establishing the potency and longevity of T cell responses induced
and the impact of subcutaneous (s.c.) anti-CTLA-4 administra- by this vaccine in NHPs, the vaccine was assessed as an individual-
tion were assessed in NHPs. Immunogenicity against individual- ized, heterologous regimen expressing patient-specific neoantigens
ized (patient-specific) predicted cancer neoantigens30 and in-depth in an ongoing phase 1/2 clinical study in patients with advanced
analysis of T cell responses elicited by vaccination are being assessed cancers (Extended Data Fig. 2a; NCT03639714). The primary
in an ongoing first-in-human phase 1/2 clinical trial (GRANITE, objectives for the phase 1 portion of this study were assessment of
NCT03639714) in patients with metastatic MSS-CRC, non-small safety, tolerability and RP2D. A single priming dose of ChAd68 was
cell lung cancer (NSCLC) or gastroesophageal adenocarcinoma administered i.m. followed by multiple i.m. samRNA boosts with
(GEA). Primary endpoints of the phase 1 part of the study were escalating doses of samRNA (Fig. 2a). Eligible patients included
safety and tolerability and RP2D. Secondary and exploratory end- those with metastatic MSS-CRC, NSCLC or GEA who were begin-
points include immunogenicity, OS, feasibility of manufactur- ning treatment with standard-of-care chemotherapy during vaccine
ing patient-specific vaccines and changes in ctDNA. Induction manufacturing and had tissue available for neoantigen prediction,
of neoantigen-specific CD8 T cell responses was observed in all normal organ function and no contraindications for treatment
patients, including patients with MSS-CRC whose tumors lacked with immunotherapy (Methods). As of data cutoff, enrollment in
immune reactivity before vaccination. Decreases in ctDNA and phase 1 had been completed, and the study is ongoing. For each
stabilization of disease and/or reduction in tumor burden in these patient meeting a threshold for cumulative predicted neoantigen
patients suggest early signs of potential therapeutic benefit in this score30 (Extended Data Fig. 2a), the 20 highest ranking mutations
CPI-refractory patient population31–33. were selected for inclusion in a codon-optimized vaccine expres-
sion cassette. The same expression cassette was inserted into both
Results patient-specific vaccine vectors for manufacturing. Patient-specific
Vaccination induces durable CD8+ T cell responses in NHPs. To vaccines were successfully manufactured for 15 patients enrolled
assess the potency and durability of the immune responses induced for vaccine manufacturing, confirming manufacturing feasibility
by this heterologous vaccine regimen preclinically in NHPs, a (a secondary endpoint) of this individualized vaccination approach
model antigen cassette encoding six well-defined NHP-specific (one patient subsequently elected not to receive study treatment).
MAMU-A*01-restricted SIV antigens29 was inserted into the Fourteen patients received vaccination and monthly intravenous
ChAd68 and samRNA vaccine platforms. MAMU-A*01-positive (i.v.) anti-PD-1 antibody nivolumab administration (480 mg every 4
rhesus macaques were immunized intramuscularly (i.m.) with weeks). Patients at dose level 3 (DL3) and DL4 also received s.c. ipi-
ChAd68 prime (n = 11), followed by i.m. samRNA boosts at 4, 12 limumab (30 mg) with each vaccination (Fig. 2a and Table 1). Four
and 20 weeks, with (n = 6) or without (n = 5) concomitant admin- patients (G1, G8, G9 and G10) received continued standard-of-care
istration of s.c. anti-CTLA-4 antibody ipilimumab (Fig. 1a and chemotherapy with study treatment, and eight patients received
Supplementary Table 1). ChAd68 induced immune responses a single ChAd68 boost ≥24 weeks after the prime vaccination
against all six antigens (mean 923 and 313 spot-forming units (Extended Data Table 1). Planned dosing intervals and timepoints
(SFU)/106 cells at week 4 ± s.c. ipilimumab, respectively; Fig. 1b). with blood draws for immunogenicity are shown in Extended Data
Primed T cell responses were boosted following administration of Fig. 2b. The median age of the 14 phase 1 patients was 59 years;
samRNA, with T cell responses still detectable 32 weeks after prime tumor types included metastatic MSS-CRC (n = 7), GEA (n = 6) or
in both groups. Concomitant administration of s.c. ipilimumab NSCLC (n = 1); and patients had received one or two prior lines of
resulted in maintenance of an overall higher T cell response (mean therapy (Table 1 and Extended Data Table 1). The individualized
843 and 277 SFU/106 cells ± s.c. ipilimumab, respectively; Fig. 1b). vaccine regimen was safe and well tolerated, with no dose-limiting
ChAd68 readministration at week 32 boosted T cell responses toxicities. The primary safety endpoint of this vaccine regimen was
~15-fold and ~12-fold ± concomitant ipilimumab, respectively reflective of a viral-based vaccine regimen with TRAEs consistent
(Fig. 1c). Immunization resulted in the generation and maintenance with a vaccine-induced immune response (Fig. 2b). TRAEs >10%
of antigen-specific effector memory T cells (Extended Data Fig. 1a), included pyrexia (86%), fatigue (43%), musculoskeletal and injec-
which have been demonstrated to be predictive of providing thera- tion site pain (both 36%) and diarrhea (29%). Serious TRAEs were
peutic benefit to patients with cancer34. largely consistent with the known safety profile of CPI therapy and
In both therapeutic and prophylactic vaccine settings, the gen- included one count each of pyrexia, duodenitis, increased transami-
eration and maintenance of long-lived memory responses is highly nases and hyperthyroidism (Fig. 2b). Based on the totality of the
desirable to maintain disease control or provide protection from safety and immunogenicity data (secondary endpoint; see next sec-
future infections. To investigate whether antigen-specific memory tion) from the phase 1 part of the study, the primary endpoint of
T cells were detectable in NHPs up to 2 years after the ChAd68 prime RP2D was determined to be 1012 VP ChAd68 and 30 µg samRNA.
vaccination, six animals that had not received additional treatment
and remained available from the initial study were assessed. Tetramer Vaccine induces neoantigen-specific CD8+ T cells. The
analysis of peripheral blood mononuclear cells (PBMCs) 80 weeks key secondary endpoint of immunogenicity was assessed for
after ChAd68 prime identified the presence of antigen-specific central neoantigen-specific T cells. To preferentially stimulate CD8 T cells
b c
ChAd68 boost
10,000 15,000
Pol SV9 ChAd68 prime + samRNA boosts + Ipilimumab
Gag LW9 – Ipilimumab
Env CL9
7,500 Env TL9
Gag CM9
10,000
Tat TL8
5,000
5,000
2,500
0 0
32 33
0
2
4
5
6
8
10
11
12
13
14
15
16
18
19
20
21
22
23
24
25
26
28
29
30
31
32
7
17
27
ChAd68 prime + samRNA boosts + ipilimumab d samRNA boost
10,000 30,000 + Ipilimumab
– Ipilimumab
7,500
IFN-γ (SFU per 106 cells)
5,000
10,000
2,500
0 0
106 107
0
2
4
5
6
8
10
11
12
13
14
15
16
18
19
20
21
22
23
24
25
26
28
29
30
31
32
7
17
27
Fig. 1 | Heterologous vaccination with ChAd68 and samRNA induces broad, durable CD8+ T cell responses in NHPs that are detectable long-term
and can be boosted ≥2 years after prime. a, Schematic of study treatment schedule. b, Antigen-specific T cell response (IFN-γ SFU per 106 cells) at
each measured timepoint for duration of study for each group (mean ± standard error of the mean for each antigen stacked), n = 6 per group up to week
20 (n = 5 for group without ipilimumab at weeks 24–107; Supplementary Table 1). c, Antigen-specific T cell response at weeks 32 and 33 (before and
after ChAd68 boost) for each group (n = 5 without ipilimumab, n = 6 with ipilimumab), average response per animal (SFU per 106 cells) for all antigens
stacked. d, Antigen-specific T cell response at weeks 106 and 107 (before and after samRNA boost) for each group (n = 5 without ipilimumab, n = 6 with
ipilimumab), average response per animal (SFU per 106 cells) for all antigens stacked.
via major histocompatibility complex (MHC) class I presenta- ELISpot following heterologous prime boost vaccination in 100% of
tion in immune assays30,35,36, individual patient minimal epitope patients (13/13), with a ChAd68 boost further increasing peripheral
(8–11mer) peptide pools were designed to include the 40 highest T cell levels in 67% of patients (4/6) with evaluable post-boost sam-
ranked predicted neoepitopes (CD8 pool; Supplementary Table 2). ples (Fig. 2c and Extended Data Fig. 3a). Only 15% of patients (2/13)
Because this exploratory minimal epitope pool design does not had responses above LOD to their cognate neoantigen peptide pool
include all potential neoantigens per mutation and can dispro- at baseline, which were increased following vaccine administra-
portionately favor some mutations with high prediction scores, tion (Fig. 2c). Similarly to observations in NHPs, vaccine-induced
additional patient-specific peptide pools consisting of overlapping T cell responses were long-lived, and T cell levels of 700–5,000
15mers spanning each vaccine cassette were also used to interrogate SFU/106 cells were maintained for >52 weeks in three patients
T cell responses more broadly (15mer pool; Supplementary Table 2). with available longitudinal samples (Extended Data Fig. 3a,b).
Posttreatment PBMC samples were available for 13 of 14 patients Notably, readministration of ChAd68 resulted in elevated T cell lev-
(all except patient G5). Stimulation of PBMCs with patient-specific els detectable 12–24 weeks after ChAd68 boost (patients G1, G3,
peptide pools resulted in IFN-γ responses detectable by ex vivo G8 and G11; Fig. 2b and Extended Data Fig. 3a). Dose escalation
a b
Treatment-related AEs >10%
Phase 1: dose selection based on safety data Hypotension Grade 1/2
Rash Grade 3/4
Adaptive design (n = 14); all patients received nivolumab
Injection site reaction
12
Arthralgia
Dose level 4: GRT-C901 (IM: 10 VP) + 30 mg ipilimumab Decreased appetite
GRT-R901 (IM: 300 µg) + 30 mg ipilimumab Diarrhea
12
Injection site pain
Dose level 3: GRT-C901 (IM: 10 VP) + 30 mg ipilimumab Muskoskeletal pain
GRT-R901 (IM: 100 µg) + 30 mg ipilimumab
Fatigue
12
Pyrexia
Dose level 2: GRT-C901 (IM: 10 VP) Treatment-related serious AE by patient
GRT-R901 (IM: 100 µg)
Hyperthyroidism
12 Transaminases increased
Dose level 2: GRT-C901 (IM: 10 VP) Duodenitis
GRT-R901 (IM: 30 µg)
Pyrexia
0 5 10 15
Number of events
c
G1
2,000 2,000 6,000
ULOQ
1,500 1,500 4,000
1,000 1,000
IFN-γ (SFU per 106 cells)
2,000
500 500
500 500
1,000
400 400 800
300 300 600
200 200 400
8
9
10
11
12
13
14
1
2
3
4
6
8
9
10
11
12
13
14
11
7
7
G
G
G
G
G
G
G
G
G
G
G
G
G
G
G
G
G
G
G
G
G
G
G
G
G
G
G
G
Patient ID
d 1,500
CD8enr (CD4 depl) CD4enr (CD8 depl) Total PBMC
10 6 6 CD8enr (CD4 depl)
70.1% 1.27% 10 5.71% 0.30% CD4enr (CD8 depl)
IFNγ (SFU per 106 cells)
105 105
1,000
104 104
G2
G8
Fig. 2 | First-in-human data show the vaccine regimen is safe and induces CD8+ T cell responses against predicted patient-specific cancer neoantigens.
a, Phase 1 study schematic outlining dose escalation. b, Safety data: TRAEs >10% and all treatment-related serious adverse events (AEs) are shown (n = 14
patients). Graph shows counts of grade 1/2 (blue bars) and 3/4 (orange bars) events. c, Patient PBMC ex vivo IFN-γ ELISpot responses to patient-specific
peptide pools (Supplementary Table 2) are shown for baseline (left panel), maximum response following ChAd prime and samRNA boosts (middle panel)
and maximum response post ChAd boost (right panel). Graphs show mean SFU per 106 PBMCs ± standard deviation (s.d.) for triplicate ELISpot wells.
Assay limit of detection (LOD) and upper limit of quantitation (ULOQ) are indicated by dotted lines. d, Flow plot showing relative CD8 and CD4 T cell
frequencies in CD4- or CD8-depleted (depl) PBMC populations. Corresponding ELISpot data (mean SFU of technical replicates ± s.d.) are shown for whole
PBMCs (black bars), CD8enr PBMCs (navy blue bars) and CD4enr PBMCs (orange bars) stimulated with dimethylsulfoxide (DMSO), patient-specific CD8 or
15mer pools for patients G2 and G8.
Mutated gene
OGT POLR1E KRAS
NKAIN1 PHF8 GLDN
60 60 MYH14 MYO5B GGA2
MUC2 MAP2K2 EXPH5
LMNB2 IGSF23 ERF
40 40 DYNC1H1 GIN1 EHBP1L1
DNAJC10 FKBP15 DNAJC11
CHP2 FILIP1 DNAH2
20 20 CCNH DHX37 CUL4A
C1orf50 CTR9 CSMD3
BNC2 CHD6 CHD9
BICD1 CENPM AP1B1
0 0 AHNAK ADAR ABI1
1
2
3
4
7
8
9
10
11
12
13
14
0 1 2 3 0 1 2 3 4 0 1 2 3
G
G
G
G
G
G
G
G
G
G
G
Patient ID G Number of positive epitopes per mutation
c d Patient G2 leukopak
Predicted HLA coverage
of positive epitopes Total CD8+ A*30:04 Tet+ A*02:01 Tet+ B*08:01 Tet+
105 21.5% 105 0.23% 105 105 0.01%
CD8-PerCP-Cy5.5
0.33%
3
3
10 3
4
10 4
5
10 5
0
0
0
10 K
15 K
20 K
25 K
0K
0
10
10
10
10
10
10
5
0
0
0
–1
–1
Total = 13 FSC-A Tet-APC Tet-PE Tet-PE
105 1.85% 14.5% 105 0.34% 0.68% 105 1.92% 6.00% 105 2.63% 1.32%
4
4 HLA-A*02:01 10
4 HLA-A*30:04
G2 1 HLA-B*08:01 103 103 103 103
8 HLA-B*57:01 0 0 0
2 HLA-C*06:02 0
40.1% 43.5% 41.8% 57.2% 30.7% 60.5%
–103 –103 –103 61.4%
–103 35.5%
3
3
4
4
5
5
0
0
0
10
10
10
10
10
10
10
10
10
10
10
10
–1
–1
–1
–1
Total = 19
CD45RA-SB702
Total = 14
CD8-PerCP-Cy5.5
33.9%
5
3
0
3
5
3
10
10
10
0
0
3
5
0
–1
0
10
10
10
0
10
10
10
0
10
10
10
–1
–1
–1
Fig. 3 | Vaccine induces polyclonal effector memory CD8 T cell responses across multiple neoantigens and HLAs. a, Patient PBMCs from available
posttreatment timepoints (G1 W12, G2 W5, G3 W8, G4 W6, G7 W4, G8 W12, G9 W8, G10 W16, G11 W16, G12 W12, G13 W12, G14 W13 and G15 W12)
were stimulated overnight in ex vivo or post-IVS IFN-γ ELISpot with patient-specific minipools or DMSO. Graphs show percentage of total response
of mean IFN-γ responses to minipool 1 (turquoise), minipool 2 (light blue), minipool 3 (medium blue) and minipool 4 (navy blue) for each patient.
b, Number of positive minimal epitopes detected (positive by ex vivo or post-IVS ELISpot) for each mutated gene. Undetectable responses against
epitopes tested are indicated by ‘x’. c, Pie charts showing patient-specific predicted HLA class I presentation of positive neoantigen responses shown
in panel b. d, Density plots showing recognition of pHLA (A*30:04, A*02:01, B*08:01) and corresponding T cell memory phenotypes for patient G2. Pie
charts showing frequencies of naive (CD45RA+CCR7+; purple), central memory (CM; CD45RA−CCR7+; blue), effector memory (EM; CD45RA−CCR7−;
green) and T effector memory RA+ (TEMRA; CD45RA+CCR7−; orange) cells in total CD8+ population (left-most chart) and CD8+tetramer+ populations
from corresponding dot plots. e, Density plots and corresponding pie charts showing recognition of pHLA (B*44:05) and corresponding T cell memory
phenotypes before and after ChAd68 boost for patient G1.
Fig. 4 | Neoantigen-reactive CD8 T cells are polyfunctional, kill targets presenting cognate peptides in multiple HLA alleles and are increased in
on-treatment tumor samples. a, Representative density plots for patient G2 ICS for CD8+ cells producing IFN-γ, CD107α, TNF-α and IL-2 in ex vivo or
IVS-expanded PBMCs stimulated overnight with DMSO or patient-specific CD8 pool. Cells were gated on PBMCs (forward scatter (FSC)-A versus side
scatter (SSC)-A), singlets (FSC-A versus FSC-H), live cells (FSC-A versus Live/Dead), CD8 or CD4 T cells (CD8 versus CD4) and cytokine+ cells.
b, Polyfunctionality was assessed via Boolean gating of CD8+cytokine+ populations (background subtracted). Pie charts show polyfunctionality of ex vivo
PBMCs and post-IVS PBMCs. Pie arcs depict individual cytokines in red (CD107α), lime green (IFN-γ), turquoise (TNF-α) and navy blue (IL-2). Pie sections
depict frequencies of cells expressing multiple cytokines as outlined in pie category legend. Main functional pie slices are colored red (quadruple positive
for CD107α, IFN-γ, TNF-α and IL-2), orange (triple positive for CD107α, IFN-γ and TNF-α) and yellow (double positive for CD107α and IFN-γ). c, Target
cell killing by IVS-expanded PBMCs from patients G2 (B*08:01, B*57:01, A*30:04). Graphs show relative viability based on NucRed count and normalized
to effector-to-target ratio of 10:1 DMSO control over time. Data are shown as mean ± s.d. from duplicate wells for targets alone stimulated with DMSO
(gray circles) or cognate peptide (black squares) and for 10:1 effector/target co-cultures stimulated with DMSO (light blue circles) or cognate peptide
(navy blue squares). d, DNA sequencing of TCR-β chains in tumor samples in baseline and on-treatment samples from patients G1 and G3. TCR-β chains
expanded in PBMCs, in both PBMCs and tumors, and contracted are shown.
Cy5-5-A :: CD8
Comp-PerCP-
4 4 + + +
10
4
10
4 10 10 CD107α IFN-γ TNF-α
3 3
10
3
10
3 10 10
0 0 0 0
0.0008% –10
3 0.58% –10
3 36.3%
–10
3
–10
3
0.20%
3 3 4 5 3 3 4 5
–10 0 10
3 3
10 10
4 5
–10 0 10
3 3 4
10 10
5 –10 0 10 10 10 –10 0 10 10 10
IFNγ-APC IFNγ-APC + +
CD107α IFNγ
5 5
10
5
10
5
10 10
Cy5-5-A :: CD8
Comp-PerCP-
4 4
10
4
10
4
10 10
3 3
3 3 10 10
10 10 Pie arcs
0 0 0 0
9.0% 47.7%
0.20% 0.32% 3 3
CD107α+
CD8-PerCP-Cy5.5
–10
CD8-PerCP-Cy5.5
–10
3
–10
3 –10
IFN-γ+
3 3 4 5 3 3 4 5
–10 0 10
3 3
10 10
4 5
–10 0 10
3 3 4
10 10
5 –10 0 10 10 10 –10 0 10 10 10
CD107α-BV421 CD107α-BV421
TNF-α+
5 5
5
10
10 10 IL-2+
5
Cy5-5-A :: CD8
10
Comp-PerCP-
4 4
4
10
4
10 10
10 Pie categories
3 3
3
10
3
10 10
10 CD107α IFN-γ TNF-α IL-2 CD107α IFN-γ TNF-α IL-2
0 0 0
0 0.30% 1.66% 32.6%
–10
3 0.04% –10
3
–10
3
–10
3
+ + + + – + + +
3 3 4 5 3 3 4 5
–10 0 10
3 3 4
10 10
5
–10 0 10
3 3 4
10 10
5
–10 0 10 10 10 –10 0 10 10 10 + + + – – + + –
TNFα-BV786 TNFα-BV786
+ + – + – + – +
5 5 5 5
10 10 10 10 + + – – – + – –
Cy5-5-A :: CD8
Comp-PerCP-
4 4 4 4
10 10 10 10 + – + + – + +
–
3
10
3
10
3
10
3
10 + – + – – – + –
0 0 0 0
3 0.016% 3 0.25% 3 0.61% –10
3 11.8% + – – + – – – +
–10 –10 –10
–10 0 10
3 3 4
10 10
5 3 3 4 5 3
–10 0 10
3 4
10 10
5 3
–10 0 10
3 4
10 10
5 + – – –
–10 0 10 10 10
IL-2-PE IL-2-PE
Cytokine
0 30 CD4
Undetermined
20
–3 Baseline On-
treatment
–4 Expanded TCR-β in tumor (n = 9)
0 8 16 24 32 40 48 56 64 72 8
G1
7
2 6
G2: HLA-B*57:01
5
0 10–3
4
Relative viability
3
–2 10–5 10–4 10–3 10–2 2
Baseline biopsy TCR-β frequency (%) 1
–4 VAF 10% 0
Baseline On-
treatment
–6 Expanded in tumor and IVS 5
Expanded in tumor 35 Expanded TCR-β in IVS (n = 27)
Contracted in tumor 11 30
On-treatment biopsy TCR-β frequency (%)
2 10
VAF 9%
Relative viability
1
0
0 Baseline On-
10–3 treatment
–1
Expanded TCR-β in tumor (n = 40)
–2 4
G3
–3 3
10–4
0 8 16 24 32 40 48 56 64 72
10–4 10–3 10–2 2
Time (h)
Baseline biopsy TCR-β frequency (%)
Targets only + DMSO Targets only + peptide VAF 17% 1
E:T 10:1 + DMSO E:T 10:1 + peptide
0
Baseline On-
treatment
IFN-γ signature
PD-L1 RNA-seq
Lung Ad.
Lung Sq.
0
100
–2
50
–4
MSS-CRC
GEA
NSCLC
MSS-CRC
MSI-CRC
Gastric
Lung Ad.
Lung Sq.
Melanoma
0
0.01 0.1 1 10 100 1,000 10,000
Tumor mutational burden
c d
1,500 100
PD per RECIST at <24 weeks
SD per RECIST for ≥24 weeks
Probability of survival
1,250
Percent change in ctDNA %
1,000
100 50
50
25
0
No ctDNA decrease (n = 4)
–50 0
0 20 40 60 80
–100
Weeks after prime
G4 G10 G6 G11 G9 G8 G14
ctDNA decrease (n = 3) No ctDNA decrease (n = 4)
PET scan
Lung
Pretreatment
Liver
On treatment
Liver
Fig. 5 | Early signals of clinical efficacy in patients with MSS-CRC in advanced disease setting. a, Scatter plot showing tumor sample RNA sequencing
(RNA-seq) for patients before study treatment. PD-L1 normalized RNA-sequencing expected counts (y axis) versus tumor mutational burden (x axis) for
patients and relevant TCGA cancer types. b, Patient RNA-sequencing IFN-γ signature average z-scores (x axis) calculated across patients and relevant
TCGA cancer types (y axis). TCGA samples: MSS-CRC (n = 286), MSI-CRC (n = 62), gastric (n = 406), lung adenocarcinoma (Lung Ad.)_ (n = 498),
lung squamous (Lung Sq.) (n = 463) and melanoma (n = 436). GRANITE patients: MSS-CRC (n = 7), GEA (n = 6) and NSCLC (n = 1). c, Percent change
in ctDNA following initiation of study treatment relative to baseline levels shown as best response and colored by RECIST-based response and time on
treatment. Patients in blue were all on study treatment >24 weeks with two patients treated beyond RECIST-based progressive disease (PD) that was
not confirmed by 24 weeks and continued on study treatment. d, Probability of survival in MSS-CRC patients with (n = 4) or without (n = 3) reduction in
ctDNA since ChAd68 prime. Data cutoff 31 January 2022. e, Radiological images showing tumor lesions in the lung and liver of patient G8 before initiating
study treatment (baseline) and following study treatment (16 and 48 weeks). Fluorodeoxyglucose-positron emission tomography imaging of tumor lesions
in the liver when patient was diagnosed in February 2018 and 7 months after starting study treatment. Arrows indicate relevant target lesions.
frequencies of vaccine-induced TCR clonotypes were found in 4. Lee, C. H., Yelensky, R., Jooss, K. & Chan, T. A. Update on tumor
on-treatment tumor samples, providing proof-of-concept for tumor neoantigens and their utility: why it is good to be different. Trends Immunol.
39, 536–548 (2018).
infiltration of vaccine-induced T cells. Patients with molecular 5. Tran, E., Robbins, P. F. & Rosenberg, S. A. ‘Final common pathway’ of human
responses following vaccination showed stabilization and/or reduc- cancer immunotherapy: targeting random somatic mutations. Nat. Immunol.
tion of target lesions and prolonged OS, indicating potential early 18, 255–262 (2017).
signs of clinical activity in this CPI-refractory patient population. A 6. DuPage, M., Mazumdar, C., Schmidt, L. M., Cheung, A. F. & Jacks, T.
Expression of tumour-specific antigens underlies cancer immunoediting.
randomized phase 2/3 study has been initiated to demonstrate the Nature 482, 405–409 (2012).
efficacy of this vaccine regimen in patients with MSS-CRC in the 7. Gubin, M. M. et al. Checkpoint blockade cancer immunotherapy targets
first-line metastatic setting (NCT05141721). tumour-specific mutant antigens. Nature 515, 577–581 (2014).
A vaccine that induces neoantigen-specific CD8 T cells provides 8. Lennerz, V. et al. The response of autologous T cells to a human melanoma is
a foundation for combination with CPIs, making their combined dominated by mutated neoantigens. Proc. Natl Acad. Sci. USA 102,
16013–16018 (2005).
administration highly desirable. Although the effects of s.c. ipi- 9. Matsushita, H. et al. Cancer exome analysis reveals a T-cell-dependent
limumab were difficult to discern in this small cohort of patients, mechanism of cancer immunoediting. Nature 482, 400–404 (2012).
administration of ipilimumab in addition to a vaccine in NHPs 10. Robbins, P. F. et al. Mining exomic sequencing data to identify mutated
resulted in increased breadth and magnitude of antigen-specific antigens recognized by adoptively transferred tumor-reactive T cells. Nat.
T cell responses in all animals. This may be due to a lower thresh- Med. 19, 747–752 (2013).
11. Tumeh, P. C. et al. PD-1 blockade induces responses by inhibiting adaptive
old for T cell activation against weaker epitopes with ipilimumab immune resistance. Nature 515, 568–571 (2014).
compared to vaccine alone. The lower threshold of T cell activation 12. McGranahan, N. & Swanton, C. Neoantigen quality, not quantity. Sci. Transl.
provided by the addition of ipilimumab might allow induction of an Med. 11, eaax7918 (2019).
overall broader immune response to vaccine-encoded neoantigens 13. Keskin, D. B. et al. Neoantigen vaccine generates intratumoral T cell
responses in phase Ib glioblastoma trial. Nature 565, 234–239 (2019).
and a more robust immune attack of the tumor. Driving broad T cell
14. Ott, P. A. et al. An immunogenic personal neoantigen vaccine for patients
responses against multiple tumor antigens across multiple HLA with melanoma. Nature 547, 217–221 (2017).
alleles, as observed in this study, may limit the impact of common 15. Ott, P. A. et al. A phase Ib trial of personalized neoantigen therapy plus
HLA-related immune escape mechanisms39–41. As a patient’s disease anti-PD-1 in patients with advanced melanoma, non-small cell lung cancer,
progresses, increased heterogeneity can lead to immune evasion. or bladder cancer. Cell 183, 347–362.e324 (2020).
16. Sahin, U. et al. Personalized RNA mutanome vaccines mobilize poly-specific
Treating patients earlier in their disease, such as in the adjuvant set- therapeutic immunity against cancer. Nature 547, 222–226 (2017).
ting or early in the treatment of metastatic disease, has the potential 17. Sahin, U. et al. An RNA vaccine drives immunity in
benefit of completely eradicating tumor cells by vaccine-induced checkpoint-inhibitor-treated melanoma. Nature 585, 107–112 (2020).
T cells before resistance mutations arise. Also, given the time lag 18. Hilf, N. et al. Actively personalized vaccination trial for newly diagnosed
between vaccine administration and T cell expansion, patients glioblastoma. Nature 565, 240–245 (2019).
19. Riaz, N. et al. Tumor and microenvironment evolution during
with lower initial disease burden and more time to generate a immunotherapy with nivolumab. Cell 171, 934–949 e916 (2017).
T cell response before disease progression are more likely to receive 20. Shaw, A. R. & Suzuki, M. Immunology of adenoviral vectors in cancer
long-term benefit and durable disease control from our vaccination therapy. Mol. Ther. Methods Clin. Dev. 15, 418–429 (2019).
approach. Increasing evidence suggests that a clear survival benefit 21. Lopez-Camacho, C. et al. Rational Zika vaccine design via the modulation of
(even without a radiologic response) may be a hallmark of immu- antigen membrane anchors in chimpanzee adenoviral vectors. Nat. Commun.
9, 2441 (2018).
notherapies42. Follow-up of this ongoing phase 1/2 study is ongoing, 22. Ogwang, C. et al. Prime-boost vaccination with chimpanzee adenovirus and
and one phase 2/3 and one phase 2 study are assessing potential clini- modified vaccinia Ankara encoding TRAP provides partial protection against
cal activity of this vaccine regimen in patients with MSS-CRC in the Plasmodium falciparum infection in Kenyan adults. Sci. Transl. Med. 7,
metastatic and adjuvant treatment settings, respectively. In addition 286re285 (2015).
23. Folegatti, P. M. et al. Safety and immunogenicity of the ChAdOx1 nCoV-19
to benefiting patients with cancer, this versatile and potent vaccine vaccine against SARS-CoV-2: a preliminary report of a phase 1/2,
can be applied to other indications such as prophylactic vaccination single-blind, randomised controlled trial.Lancet 396, 467–478 (2020).
against SARS-CoV-2 (ref. 43) (NCT05148962, NCT04776317). In 24. & Sadoff, J. et al. Interim results of a phase 1-2a trial of Ad26.COV2.S
sum, this versatile heterologous vaccine regimen generates strong, covid-19 vaccine.N. Engl. J. Med. 384, 1824–1835 (2021).
persistent and functional immune responses that have broad appli- 25. Zhao, H. et al. Seroprevalence of neutralizing antibodies against human
adenovirus type-5 and chimpanzee adenovirus type-68 in cancer patients.
cability to a wide range of disease settings and support continued Front Immunol. 9, 335 (2018).
exploration in the clinical setting to bring better treatment options 26. Lundstrom, K. RNA viruses as tools in gene therapy and vaccine
to patients with cancer and people at risk of infection. development. Genes (Basel) 10, 189 (2019).
27. Mulligan, M. J. et al. Phase I/II study of COVID-19 RNA vaccine BNT162b1
Online content in adults. Nature 586, 589–593 (2020).
28. McNamara, M. A., Nair, S. K. & Holl, E. K. RNA-based vaccines in cancer
Any methods, additional references, Nature Research report- immunotherapy. J. Immunol. Res 2015, 794528 (2015).
ing summaries, source data, extended data, supplementary infor- 29. Allen, T. M. et al. CD8+ Lymphocytes from simian immunodeficiency
mation, acknowledgements, peer review information; details of virus-infected rhesus macaques recognize 14 different epitopes bound by the
author contributions and competing interests; and statements of major histocompatibility complex class i molecule Mamu-A*01: implications
for vaccine design and testing. J. Virol. 75, 738–749 (2001).
data and code availability are available at https://doi.org/10.1038/
30. Bulik-Sullivan, B. et al. Deep learning using tumor HLA peptide mass
s41591-022-01937-6. spectrometry datasets improves neoantigen identification. Nat. Biotechnol.,
https://doi.org/10.1038/nbt.4313 (2018).
Received: 4 March 2022; Accepted: 6 July 2022; 31. Chen, E. X. et al. Effect of combined immune checkpoint inhibition vs best
Published online: 15 August 2022 supportive care alone in patients with advanced colorectal cancer: the
Canadian Cancer Trials Group CO.26 Study. JAMA Oncol. 6, 831–838 (2020).
32. Bendell, J. et al. Efficacy and safety results from IMblaze370, a randomised
References phase III study comparing atezolizumab1cobimetinib and atezolizumab
1. Lawrence, M. S. et al. Mutational heterogeneity in cancer and the search for monotherapy vs regorafenib in chemotherapy-refractory metastatic colorectal
new cancer-associated genes. Nature 499, 214–218 (2013). cancer. Ann. Oncol. 29, v123 (2018). 2018.
2. Rizvi, N. A. et al. Cancer immunology. Mutational landscape determines 33. Grothey, A. et al. Fluoropyrimidine (FP) 1 bevacizumab (BEV) 1
sensitivity to PD-1 blockade in non-small cell lung cancer. Science 348, atezolizumab vs FP/BEV in BRAFwt metastatic colorectal cancer (mCRC):
124–128 (2015). findings from cohort 2 of MODUL–a multicentre, randomized trial of
3. Snyder, A. et al. Genetic basis for clinical response to CTLA-4 blockade in biomarkerdriven maintenance treatment following first-line induction
melanoma. N. Engl. J. Med. 371, 2189–2199 (2014). therapy. Ann. Oncol. 29, viii714–viii715 (2018).
IFN-γ ELISpot assay (NHP study). IFN-γ enzyme-linked immunospot (ELISpot) Anti-ChAd68 neutralizing antibody assay (NHP and clinical study). Dilutions of
assays were performed using precoated 96-well plates (MAbtech, Monkey IFNγ heat-inactivated human and NHP sera were evaluated for the ability to neutralize
ELISpot PLUS, ALP) following the manufacturer’s protocol. Then, 105 PBMCs a ChAd virus expressing the reporter protein beta-galactosidase (ChAd-LacZ).
per well were plated in triplicate with 10 µg ml−1 peptide stimuli (GenScript) and The reciprocal of the dilution that gives 50% or greater neutralization is used to
incubated overnight in complete RPMI (RPMI + 10% FBS). A human hepatitis B assign a neutralization titer. In brief, pre- and post-vaccination serum samples
virus S-antigen peptide not contained in the cassette (WLSLLVPFV, GenScript) were heat inactivated at 56˚C for 30 min. Sera samples were then diluted twofold
was used as a negative control for each sample. Plates were washed with PBS and in serum-free media (DMEM) starting at 1:20 dilution down to 1:1,280. If needed,
incubated with anti-monkey IFN-γ mAb biotin (MAbtech) for 2 h, followed by an additional dilutions were made to get a titer within the range of the assay. The
additional wash and incubation with Streptavidin-ALP (MAbtech) for 1 h. After diluted samples were then mixed with an equal volume of media containing diluted
final wash, plates were incubated for ten minutes with BCIP/NBT (MAbtech) to virus sufficient to infect 293 A cells at an approximate multiplicity of infection
develop the immunospots and dried overnight at 37°C. Spots were imaged and (m.o.i.) of 1. The diluted sera and virus were incubated for 1 h at 37˚C before
enumerated using AID reader (Autoimmun Diagnostika). For data processing and adding onto 293 A cells plated at 2.5 ×104 cells/well on a 96-well tissue culture
analysis, samples with replicate well variability (variability = variance/(median + 1)) plate 1 day earlier. The cells and neutralizing mix were incubated for 1 h at 37˚C
greater than 10 and median greater than 10 were excluded44. Spot values were and then an equal volume of media with 20% FBS was added. The cells and virus
adjusted based on the well saturation according to the formula45: were then incubated for an additional 18–20 h before lysis and measurement of
cfDNA collection and isolation. Whole blood was collected in two 10-ml cell-free IncuCyte killing assays. Single HLA-expressing A375 NucLight Red cell lines
DNA (cfDNA) BCT (Streck) starting at the time of the prime, and subsequent were seeded in 96- or 48-well plates at concentrations of 2.5 × 104 or 3.5 ×104 cells
draws were collected at dosing visits. Due to COVID-19 restrictions, several per well in DMEM with 10% heat-inactivated FBS. The plates were placed in the
blood draws were missed. Whole blood underwent a double-spin protocol to first IncuCyte S3 (Essen Biosciences) and 24 h after the seeding, the effector cells were
separate plasma from WBCs and red blood cells before a second spin to remove plated at a concentration of 2.5 × 105 or 3.5 × 105 cells per well in a 96- or 48-well
any remaining cellular debris. The separated plasma was frozen and shipped to plate, respectively, for an effector to target ratio of 10:1. Individual peptides
Gritstone Bio and stored at −80°C until extraction. cfDNA was extracted from the (GenScript; Supplementary Table 2) were added to the treated wells for a final
entire plasma volume of a single draw using the Apostle MiniMax cfDNA Isolation concentration of 10 µg ml−1, and DMSO was used for the control wells. The plates
kit (ApostleBio) and quantified using the Qubit 1x dsDNA High Sensitivity Assay were imaged with the IncuCyte for up to 72 h, after which the data were analyzed
(Thermo Fisher Scientific). using the IncuCyte S3 2018 analysis software. Viability of A375 was assessed by
red cell count, and relative viability was calculated relative to DMSO co-culture
control wells.
ctDNA sequencing and analysis. Custom hybrid capture panels were designed
for each patient enrolled in the study. Each patient panel was designed to capture
Statistics and reproducibility. For the clinical study, the sample size is not
the EDGE-predicted neoantigens and all predicted coding (nonsynonymous)
driven by a statistical power due to the nature of an adaptive dose-escalation
mutations present in the original screening biopsy specimen. Target mutations
method. Therefore, the sample size of the phase 1 portion of the study ranges
for each patient were curated and sent for customized design to IDT as xGen
from 11 to maximum of 24 patients, which is dependent on occurrence of
Lockdown Probes. Each capture pool contained up to nine patients per pool.
DLTs. No data were excluded from the analyses, and experiments were not
Patient-matched genomic DNA was fragmented before library preparation using
randomized. The investigators were not blinded to allocation during experiments
the NEB FS module (NEB). Shotgun libraries for cfDNA (up to 30 ng) and the
and outcome assessment. For the NHP study, the number of animals per group
fragmented, patient-matched genomic DNA (20–30 ng) were prepared using
is n = 3/sex, which is the minimum number of animals needed to do simple
the KAPA HyperPrep (KAPA Biosystems) kit using a customized pool of duplex
statistics. The total number of animals used in this study is considered the
adapters containing unique molecular identifiers for duplex sequencing (IDT).
minimum required to properly characterize the effect of the test articles (i.e.,
Shotgun libraries were captured with patient-specific probes overnight using the
vaccine and anti-CTLA-4) and compare their route of administration (i.e., s.c.
IDT xGen Hybridization and Wash kit. Enriched libraries were sequenced on
versus i.v.) and has been designed such that it does not require an unnecessary
an Illumina NovaSeq to a minimum mean raw depth of 80,000×. Briefly, unique
number of animals to accomplish its objectives. For both the NHP and clinical
molecular identifiers were clipped from the raw sequencing reads before alignment
study, descriptive statistics are described throughout. Where applicable,
to hg38 using BWA-MEM. Using fgbio, aligned reads were grouped by position
statistical analyses applied for specific methods are outlined in the relevant
and duplex identity. Consensus reads were created using a duplex of 3× (three
methods sections.
supporting reads from each strand) and realigned to hg38. Variant calling was
performed using FreeBayes and filtered for the patient-specific variants. Variant
Reporting summary. Further information on research design is available in the
allele frequency was converted to mutated haploid genomic equivalents per
Nature Research Reporting Summary linked to this article.
milliliter plasma (hGE ml−1) using the extracted plasma volume and total cfDNA
yield from the extraction. Percent change in ctDNA was calculated as the change of
the median mutated hGE ml−1 from the baseline sample. Data availability
Deidentified individual participant clinical data that underlie the results reported
Patient tumor biopsy whole-exome and transcriptome analysis and TCGA in this article are available for transfer. Interested investigators can obtain and
data comparison. Pretreatment GRANITE patient FFPE biopsy-derived DNA certify the data transfer agreement and submit requests to the principal investigator
and RNA and matched whole blood-derived DNA were whole-exome sequenced (K.J.). Investigators and institutions who consent to the terms of the data transfer
(IDT xGen V1) and analyzed as described previously38. Relevant tumor types agreement form, including, but not limited to, the use of these data for the
for genomic comparison from TCGA Pan-Cancer Atlas dataset included gastric purpose of a specific project and only for research purposes, and to protect the
(n = 405 stomach adenocarcinoma), colon adenocarcinoma (n = 332, MSS-CRC confidentiality of the data and limit the possibility of identification of participants
(281) and MSI-CRC (51)), lung adenocarcinoma (n = 498), lung squamous cell in any way whatsoever for the duration of the agreement, will be granted access.
carcinoma (n = 463, LUSC) and melanoma (n = 436, skin cutaneous melanoma). Gritstone bio will then facilitate the transfer of the requested deidentified data.
For RNA expression analyses, all GRANITE and TCGA sample RNA-seq by This mechanism is expected to be via a Gritstone Secure File Transfer Service, but
Expectation-Maximization expected counts were normalized utilizing DESeq2 Gritstone bio reserves the right to change the specific transfer method at any time,
(ref. 49). Normalized counts were used to determine patient PD-L1 RNA expression provided appropriate levels of access authorization and control can be maintained.
and the IFN-γ RNA expression signatures, which were calculated as the average of Source data are provided with this paper. Pan-Cancer Atlas data sets were obtained
28 IFN-γ-associated gene specific z-scores across all GRANITE and TCGA samples from the cBioPortal (https://www.cbioportal.org/) and the UCSC Xena TCGA
(n = 2,494)50. Tumor mutational burden was calculated for each GRANITE patient Pan-Cancer Atlas data hub (https://pancanatlas.xenahubs.net), and raw data are
as the total of all somatic variants called >4% variant allele frequency divided by available through the Genomic Data Commons (https://gdc.cancer.gov/). Epitope
the exome bait panel size (39 Mb). For TCGA samples, total mutations called were selection and patient-specific neoantigen selection used a previously published
divided by exome bait panel size used (38 Mb)51 (Supplementary Table 4). algorithm30. Source data are provided with this paper.
Author contributions
C.D.P., A.R.R., M.D., M.S., G.G., W.B., R.R., R.Y., A.A., A.F. and K.J. designed the study Additional information
and wrote the manuscript. C.D.P., A.R.R., M.D., M.G.H., C.D.S., L.D.K., S.K., A.Y., L.S., Extended data is available for this paper at https://doi.org/10.1038/s41591-022-01937-6.
D.S., M.S., K.T., M.M., J.R.J., C.N.N., E.M., R.Z., D.N.G., A.C.G., R.G., K.B., M.D.C. Supplementary information The online version contains supplementary material
and R.Y. contributed to experimental design, execution and data analysis. A.I.S., S.R., available at https://doi.org/10.1038/s41591-022-01937-6.
D.C., B.S.H., C.G.D., B.J.S., D.A., A.M., S.B.M., B.J., R.R., C.-Y.L., D.V.T.C. and A.R.F.
contributed to clinical oversight, patient recruitment, enrollment and treatment. C.D.S., Correspondence and requests for materials should be addressed to Karin Jooss.
L.G., S.-J.H., W.B., M.D.C., S.C. and K.J. contributed to vaccine design and production. Peer review information Nature Medicine thanks Robert Seder and the other,
anonymous, reviewer(s) for their contribution to the peer review of this work. Primary
Handling editors: Saheli Sadanand and Joao Monteiro, in collaboration with the Nature
Competing interests
Medicine team.
C.D.P., A.R.R., M.G.H., C.D.S., L.G., S-.J.H., L.D.K., S.K., D.S., M.D., M.M., J.R.J.,
C.N.N., R.Z., D.N.G., A.C.G., M.D.C., S.C., K.B., R.R., A.A., A.R.F., K.J., A.Y., L.S., M.S., Reprints and permissions information is available at www.nature.com/reprints.
Extended Data Fig. 2 | (A) CONSORT flow diagram of patient enrollment. (B) Schematic outlining personalized vaccine production, planned dosing
schedule, and immune monitoring.
Extended Data Fig. 4 | (A) ELISpot data (mean SFU per 106 PBMCs +/- SD for triplicate wells) are shown for whole PBMCs (black bars), CD4-depleted
(CD8enr) PBMCs (navy blue bars), and CD8-depleted (CD4enr) PBMCs (orange bars) stimulated with DMSO, CD8 Pool, or 15mer pool for patients G1, G11,
and G14 (pooled posttreatment timepoints). (B) Post-IVS ELISpot data for baseline and posttreatment sample PBMCs stimulated with patient-specific
CD8 and minipools are shown for patients G1, G2, G3, G8, G9, G12, G13 and G4. Graphs show mean of technical duplicates (SFU per 106 PBMCs) for all
except G2 CD8 Pool, where enough cells to test triplicates were available, with DMSO background subtracted. ULOQ for post-IVS assay is indicated by
dotted line.
Extended Data Table 1 | Listing of patient baseline characteristics, Disposition, and Efficacy by DL (as of 31 January 2022)
GEA: Gastroesophageal adenocarcinoma; NSCLC: Non-Small Cell Lung Cancer; CRC: Colorectal Cancer; 1Dose Level 1=GRT-C901/GRT-R902 30μg+ Nivolumab; Dose Level 2= GRT-C901/GRT-R902
100μg+ Nivolumab; Dose Level 3= GRT-C901/GRT-R902 100μg+ Nivolumab+ SC Ipilimumab; Dose Level 4= GRT-C901/GRT-R902 300μg+ Nivolumab+ SC Ipilimumab 2Weeks from the prime ChAd68
dosing 3Best of response as per RECIST v 1.1; CR=confirmed response; PR=partial response; SD=stable disease (≥ 16 weeks on treatment); PD=progressive disease; NE=no evidence of disease 4Time from
the first dose with GRT-C901 (or GRT-R902 if patient does not receive GRT-C901) to the earliest date of PD or death by any cause 5Time to death (died) or last contact/assessment (alive) from the 1st
study treatment 6This is a confirmed complete response defined as disappearance of target liver lesions and no visible disease on imaging; biopsy at original primary tumor site showed microscopic invasive
adenocarcinoma at 4 months; response duration of 6 months followed by worsening of primary esophageal lesion with dysphagia *Concurrent chemotherapy with study treatment: Patient G1 received 3
doses of 5-FU over the first 8 weeks. Patient G8 received 5-FU/bevacizumab (had been receiving for 21 months prior to study treatment). Patient G9 received FOLFIRI/cetuximab (receiving for 11 months
prior to study treatment). Patient G10 received FOLFIRI (receiving for 11 months prior to study treatment). Patient G12 received Paclitaxel/ramucirumab started after 12 weeks of study treatment.
≥
μ μ μ
μ
μ