Anda di halaman 1dari 12

articles

Eradication of large established tumors in mice by


combination immunotherapy that engages innate and
adaptive immune responses
Kelly D Moynihan1–3,14, Cary F Opel1,4,14, Gregory L Szeto1–3,13, Alice Tzeng1,2, Eric F Zhu1,4, Jesse M Engreitz5,6,
© 2016 Nature America, Inc., part of Springer Nature. All rights reserved.

Robert T Williams7, Kavya Rakhra1, Michael H Zhang1,13, Adrienne M Rothschilds1,2, Sudha Kumari1,
Ryan L Kelly1,2, Byron H Kwan1,2, Wuhbet Abraham1, Kevin Hu2, Naveen K Mehta1,2, Monique J Kauke1,4,
Heikyung Suh1, Jennifer R Cochran8–10, Douglas A Lauffenburger1–3, K Dane Wittrup1,2,4 & Darrell J Irvine1–3,11,12

Checkpoint blockade with antibodies specific for cytotoxic T lymphocyte–associated protein (CTLA)-4 or programmed cell death
1 (PDCD1; also known as PD-1) elicits durable tumor regression in metastatic cancer, but these dramatic responses are confined
to a minority of patients. This suboptimal outcome is probably due in part to the complex network of immunosuppressive
pathways present in advanced tumors, which are unlikely to be overcome by intervention at a single signaling checkpoint. Here
we describe a combination immunotherapy that recruits a variety of innate and adaptive immune cells to eliminate large tumor
burdens in syngeneic tumor models and a genetically engineered mouse model of melanoma; to our knowledge tumors of this
size have not previously been curable by treatments relying on endogenous immunity. Maximal antitumor efficacy required
four components: a tumor-antigen-targeting antibody, a recombinant interleukin-2 with an extended half-life, anti-PD-1 and a
powerful T cell vaccine. Depletion experiments revealed that CD8+ T cells, cross-presenting dendritic cells and several other
innate immune cell subsets were required for tumor regression. Effective treatment induced infiltration of immune cells and
production of inflammatory cytokines in the tumor, enhanced antibody-mediated tumor antigen uptake and promoted antigen
spreading. These results demonstrate the capacity of an elicited endogenous immune response to destroy large, established
tumors and elucidate essential characteristics of combination immunotherapies that are capable of curing a majority of tumors
in experimental settings typically viewed as intractable.

Checkpoint blockade therapies prove that an endogenous immune which may generate immunosuppressive networks more akin to
response can regress human tumors, but significant responses those of human tumors, are also generally refractory to immuno-
remain restricted to a minority of patients1–5. To avoid immune-medi- therapy19,20. As such, the general characteristics of an endogenous
ated clearance, advanced tumors co-opt an array of immunosuppres- immune response capable of reliably eradicating large immuno­
sive pathways, which are unlikely to be overcome by intervention at suppressive tumor burdens remain unclear.
a single signaling checkpoint4,6–9. Combining checkpoint blockade We recently reported that therapies combining tumor-specific
with other agents may improve response rates10–12, and immuno- antibodies and an interleukin (IL)-2 molecule with an extended
therapies that use three or more agents in tandem are already in half-life (IL-2 fused with either mouse serum albumin (MSA–IL-2)
clinical testing13–16. However, even in preclinical models, com- or the antibody Fc fragment) showed synergistic therapeutic effects
plete tumor rejection is usually achieved only by treating very small in mouse models of cancer21. This combination was curative when
tumors and/or by treating the tumors at very early times preceding administered with adoptive T cell therapy. We hypothesized that simi-
the establishment of a fully developed tumor microenvironment, lar therapeutic results might be achieved in the setting of a wholly
unless adoptive transfer of ex-vivo-expanded T cells is used17,18. endogenous immune response by replacing T cell transfer with a pow-
Autochthonous tumors induced in genetically engineered animals, erful vaccine. To this end, we used a potent lymph-node (LN)-targeted

1Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology (MIT), Cambridge, Massachusetts, USA. 2Department of Biological
Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA. 3Ragon Institute of Massachusetts General Hospital, Massachusetts Institute
of Technology and Harvard University, Cambridge, Massachusetts, USA. 4Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge,
Massachusetts, USA. 5Division of Health, Science and Technology, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA. 6The Broad Institute
of MIT and Harvard University, Cambridge, Massachusetts, USA. 7Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA.
8Department of Bioengineering, Stanford University, Stanford, California, USA. 9Department of Chemical Engineering, Stanford University, Stanford, California,

USA. 10Stanford Cancer Institute, Stanford, California, USA. 11Department of Materials Science and Engineering, Massachusetts Institute of Technology, Cambridge,
Massachusetts, USA. 12Howard Hughes Medical Institute, Chevy Chase, Maryland, USA. 13Present address: Department of Chemical, Biochemical and Environmental
Engineering, University of Maryland Baltimore, Baltimore, Maryland, USA. 14These authors contributed equally to this work. Correspondence should be addressed
to D.J.I. (djirvine@mit.edu) or K.D.W. (wittrup@mit.edu).

Received 20 March; accepted 14 September; published online 24 October 2016; doi:10.1038/nm.4200

nature medicine  advance online publication 


Articles

vaccine composed of peptide antigens and CpG DNA conjugated to epitopes were synthesized (Supplementary Fig. 1d–g and ref. 22).
albumin-binding lipids. These amphiphile–vaccines reversibly bind Tumor cells (106) were injected subcutaneously and allowed to grow for
to the albumin present in interstitial fluid and efficiently traffic to 8 d before initiation of therapy, resulting in tumor sizes of ~40–60 mm2
LNs, leading to robust T cell responses22. We also included systemic at the initial time of treatment, depending on the tumor model. The
anti-PD-1 administration because of its capacity to enhance vaccine protein components were injected intraperitoneally (i.p.), and the
responses and further modulate the tumor microenvironment by vaccine was administered subcutaneously (s.c.) at the base of the tail.
blocking suppressive signals that dampen antitumor immunity5,8. In Therapies were given once per week, with P and V being administered
multiple syngeneic tumor models, as well as in the Braf/Pten geneti- three times, and A and I being given five times (Fig. 1a).
cally engineered mouse model of melanoma, this quaternary com- This AIPV regimen induced robust tumor regression and durable
bination immunotherapy cured a majority of mice with established cures in 75% of the mice bearing B16F10 melanoma tumors (Fig. 1b),
tumors and elicited long-lived protective T cell memory responses. whereas combinations of three or fewer of the AIPV components
elicited weaker therapeutic responses, ranging from modestly reduced
RESULTS efficacy (AIP) to outcomes barely different from those of untreated
Tumor regression by combination therapy tumors (APV) (Fig. 1b and Supplementary Fig. 2a,b,g). Greater than
Motivated by the synergy observed between extended-half-life IL-2 and 80% of AIPV-treated long-term survivors rejected a re-challenge with
tumor-specific antibodies21 and by our recent discovery of a strategy to 105 B16F10 cells 2 months after cessation of therapy, suggesting for-
© 2016 Nature America, Inc., part of Springer Nature. All rights reserved.

efficiently target peptide vaccines to LNs22, we explored combination mation of effective immunological memory (Fig. 1c) Similarly, in
treatments bringing these reagents together with checkpoint blockade. the DD-Her2/neu and TC-1 tumor models, >70% of mice achieved
To simplify reference to this four-component therapy, we designate complete tumor rejection after treatment with AIPV, and 100% of the
combinations of these components by concatenated single initials A long-term survivors rejected tumors after re-challenge (Fig. 1d–g
(tumor-antigen-specific antibody), I (MSA–IL-2), P (anti-PD-1) and V and Supplementary Fig. 2c–f,h,i). Triple combinations of AIPV
(amphiphile–vaccine). We first evaluated this combination treatment components demonstrated substantially lower efficacy in primary
in three subcutaneous syngeneic tumor models: B16F10 melanoma, tumor rejection, although each of the three tumor models showed
DD-Her2/neu (a mouse breast cancer line that expresses the transform- a different hierarchy of efficacy with the ternary combinations. A
ing rat Erbb2 (also known as Her2/neu) oncogene) and TC-1 tumors, limited exploration of alternate four-agent combinations revealed that
which express the human papilloma virus (HPV) onco-antigens E6 and anti-PD-1 could be replaced by an antibody specific for CTLA-4, but
E7. For each tumor, appropriate A and V components were selected several other combinations failed to replicate AIPV in eliminating
(Fig. 1a). Antibodies to melanoma-associated antigen tyrosinase- B16F10 tumors (Supplementary Fig. 3). Notably, despite high rates of
related protein 1 (Trp1; targeted by the TA99 antibody) and Her2/neu response, AIPV therapy was associated with minimal systemic toxic-
(targeted by 7.16.4) were used to treat mice bearing the B16F10 and ity, as mice did not show weight loss or substantial elevation in the
DD-Her2/neu tumors, respectively, whereas an antibody surrogate amounts of liver enzymes in the blood (Supplementary Fig. 4a–c).
2.5F–Fc (Fc fused with an engineered binding protein that recognizes
the tumor-expressed αvβ3, αvβ5 and α5β1 integrins23) was used to treat Combination therapy affects immune cell infiltration into the tumor
mice with TC-1 tumors. Amphiphile–peptide vaccines encoding the To understand the cellular and molecular mechanisms underlying the
well-characterized immunodominant Trp2, Her2/neu and HPV E7 observed therapeutic effect of the quaternary combination therapy, we

a 0 8 15 22 29 36 75 or 125
b c
AIPV
% rejecting secondary

100 IPV*** 100


106 tumor cells, AIPV AIPV AIPV Al Al 105 tumor cells, 80
AIV**
Survival (%)

75
challenge

s.c. right flank s.c. left flank


60
AIP*
Antibody (target) Vaccine (sequence) 50 40
APV***
Trp-2180–188 (SVYDFFVWL) 25 20
B16F10 TA99 (TRP1) Untreated***
0
DD-Her2/Neu 7.16.4 (HER2) p66, Her2/Neu66–74 (TYVPANASL) 0
P
AI V
PV
AI
AI

0 15 30 45 60 75
TC-1 2.5–Fc (αvβ3, αvβ5, α5β1 integrins) HPV E743–62 (GQAEPDRAHYNIVTFCCKCD)
Time (d)

d e f AIPV
g
100 AIPV 100
% rejecting secondary

100
% rejecting secondary

100 IPV*
IPV 75
Survival (%)

75
Survival (%)

80 80
AIV**
challenge

AIV
challenge

50 60 50 60
AIP* AIP***
40 40
25 APV* 25 APV*
20 20
0 Untreated*** 0 Untreated*** 0
0
0 20 40 60 80 100 120 0 25 50 75 100 125 150
AI V
V
AI V
PV
V
AI V
PV

AP

IP
AI
IP

Time (d) Time (d)

Figure 1  AIPV immunotherapy cures large established tumors and establishes protective memory in multiple tumor models. (a) Components of AIPV
therapy and timeline of treatment. (b–g) Survival over time for mice inoculated with 106 B16F10 (b), DD-Her2/neu (d) or TC-1 (f) tumor cells s.c. in the
flank and treated 8 d later with AIPV or the indicated ternary combinations, and the fraction of long-term survivors that rejected a re-challenge with 10 5
tumor cells on day 75 for B16F10 (c) and day 125 for DD-Her2/neu cells (e) or TC-1 (g) cells. In b,c, n = 10 C57Bl/6 mice per group for APV, and
n = 20 for all other groups. In d,e, n = 7 Balb/c mice for untreated and AIPV groups, and n = 5 for all other groups. In f,g, n = 8 C57Bl/6 mice per
group. Data were compiled from two (DD-Her2/neu and TC-1) or three (B16F10) independent experiments. Arrows indicate treatment time points.
*P < 0.05; **P < 0.01; ***P < 0.001; versus AIPV by log-rank (Mantel–Cox) test.

 advance online publication  nature medicine


articles

a AIPV AIP
c Euclidean
distance
–8 –4 0 4 8
4 IFN-γ+ 4 +
10 10 IFN-γ
log2 fold change
3
13.8%
3
0.53%
+ Peptide 20
10 10 relative to untreated
0 0 10
3 4 4 3 AIPV
0 10 10 0 10 10
IFN-γ (PE)

4 IFN-γ+ 4 IFN-γ+
10 10 IPV
3 0.51% 3 0.73% – Peptide
10 10
0 0
AIV
3 4 3 4
0 10 10 0 10 10
CD8 (APC) AIP

b 15
APV
% lFN-γ+ of CD8+

10 AIPV
AIV Untreated
5
© 2016 Nature America, Inc., part of Springer Nature. All rights reserved.

IPV
IP10
VEGF
IL-12p40
IL-9
IL-15
IL-17
IL-13
IL-10
GMCSF
IL-3
IL-12p70
IL-1β
MIP-1β
LIF
TNF-α
IL-17A
IL-2
IL-4
IL-1α
Eotaxin
MCP1
MIG
MCSF
RANTES
MIP-1α
LIX
KC
GCSF
IL-6
MIP2

IL-5
IFN-γ
AIP
0
0 14 28 42 56 70
Time (d) 5 4 3 2 1 Clusters

d 80
e 4
f
variance captured (%)

3 4
log10 tumor mass

score
VIP

60 2
2 2
1 R = 0.69
AIPV
40 0
0 IPV
0.0
LV1

20 AIV
loading

–0.2 –2
LV1

AIP
–0.4
0 APV
–0.6 –4
1 2 3 4 5
Cytokine clusters
–6
Eo ES
n
M -4
a
a
M IG
M -1
SF

-2
xi

-1
-1

by heat map
IL

IL
M
ta

-C
T

IP
IL

C
AN

0.5 1.0 1.5 2.0 2.5 3.0


R

log10 tumor mass

Figure 2  AIPV therapy primes sustained vaccine-specific T cell responses and remodels the microenvironment of established tumors. C57Bl/6 mice
with B16F10 tumors were treated with AIPV or the indicated ternary combinations of the therapy components as in Figure 1a. (a,b) Peripheral blood
cells were stimulated with Trp2 peptide for 6 h in the presence of brefeldin A. Shown are representative flow cytometry plots for intracellular cytokine
staining to detect Trp2-specific CD8+ T cells in peripheral blood on day 21 (a) and quantification of Trp2-specific IFN-γ+ cells among CD8+ T cells
at the indicated time points (n = 5 mice per group) (b). Data are mean + s.e.m. (c–f) Mice bearing B16F10 tumors were treated with the regimens
indicated, and on day 17 following tumor cell inoculation, tumors were isolated, and cytokine and chemokine levels were measured by Luminex
(n = 9 mice per group). Five co-regulated clusters of cytokines and chemokines were identified by hierarchical cluster analysis and then pruning
clusters by applying a threshold to linkage distance (c and Supplementary Fig. 5). These clusters were independently tested for their ability to predict
log-transformed tumor size in treated mice on day 17 using a single latent variable (LV) in an orthogonalized partial least-squares regression (oPLSR)
model (d). For the best-performing cluster 3 (red bar), VIP scores (a measure of each cytokine’s contribution to the LV score and overall regression
performance) were used to assess the contribution of individual proteins from the cluster to the oPLSR model (e); VIP scores >1 (dashed line) were
considered significant (top, red bars), and the loading factors for LV1 of the oPLSR model are shown (bottom, red bars). A scatter plot shows the
oPLSR cluster 3 model performance for individual mice from all treatment groups and the best fit linear regression (dashed line) (f).

further analyzed responses to AIPV therapy in the B16F10 model as Analysis of the variance in tumor mass explained by each model
representative of an immunosuppressive, poorly immunogenic tumor. revealed that cluster 3 cytokines captured the most variance (Fig. 2d)
AIPV treatment induced expansion of an interferon (IFN)-γ-producing and, therefore, provided the most predictive model. Variable impor-
Trp2-specific CD8+ T cell population in the blood of mice (Fig. 2a,b); tance in projection (VIP) scores identified four predictive biomarkers
abundance of these cells in treated animals correlated with vitiligo, from cluster 3: macrophage inflammatory protein (MIP)-1α; regu-
which is associated with favorable responses to immunotherapy in lated on activation, normal T cell expressed and secreted (RANTES);
the clinic24 (Supplementary Fig. 4d–f). However, similar Trp2-spe- eotaxin; and IL-4; all of whose levels were inversely correlated
cific T cell responses were elicited by AIV and IPV treatments, which with tumor size and predicted tumor mass across all five treatment
failed to induce comparable therapeutic outcomes (Fig. 2b). We thus combinations (R2 = 0.69; Fig. 2e,f).
looked for changes within tumors that correlated more closely with Motivated by this chemoattractant-rich protective signature,
survival. Luminex-based quantification of intratumoral cytokines we analyzed cellular infiltrates in treated tumors. AIPV therapy
and chemokines revealed five clusters of co-regulated proteins, with induced tumor infiltration by effector CD4+ and CD8+ T cells,
many pro-inflammatory factors increased in tumors after treatment CD11b+Ly6G+Ly6Clow neutrophils25,26, NK cells and other myeloid
with effective therapies (Fig. 2c and Supplementary Fig. 5). We used cells (Fig. 3a–d and Supplementary Fig. 6a–d). PD-1+TIM-3+CD8+
partial least-squares regression (PLSR) to construct models using T cells were infrequent in all treatment groups, but AIPV treatment
cytokines from each cluster to predict log10(tumor mass) values. induced a large increase in the ratio of CD8+ T cells to regulatory T

nature medicine  advance online publication 


Articles

a d e

PMNs (×105) per g tumor


CD11b+ Ly6G+ Ly6Clow
50 25
CD8 Untreated IPV
CD8s (×105) per
* *
g tumor 40
*** ** ** ** 15
* * Actin
30 10
* ** ** * DAPI
20 ** *
5
10
* *** 0
0

d
V
PV
V
AI I
P
V
AIPV
PV

AI I
AIP
V
AI V
PV
d
V
APV
V
AI I
P
V
AI V
PV

AI I
P
V
AI V
PV

A
A

te

AP

AI

IP
te

AI
IP

AI
IP

I
a
a

re
re

nt
nt

U
Day 14 Day 21 Day 14 Day 21
U

b 12 *
APV AIV
(×105) per g tumor

f
CD4 effectors

9 ** ** **
** 100
6 * **

Survival (%)
3 75

0 50
d
V
AP V
V
AI I
P
V
AI V
PV

AI I
P
V
AI V
PV
A

25
te

AI
IP

AI
IP
a
re

AIP AIPV
nt

Day 14 Day 21 0
U

c
© 2016 Nature America, Inc., part of Springer Nature. All rights reserved.

0 15 30 45 60 75
125 ** *
Time (d)
CD8/Treg ratio

50
** AIPV AIPV (CD8 dep)***
40 * **
** ** AIPV (CSF1R dep)* AIPV (Ly6G dep)**
20 ** ***
** ** AIPV (NK1.1 dep)** Untreated ***
0
ed
V
PV
V
AI I
P
V
AI V
PV

AI I
P
V
AI V
PV
A

A
AP

AI
IP

AI
IP
at
re
nt

Day 14 Day 21
U

Figure 3  AIPV therapy induces pronounced immune infiltration of tumors with efficacy dependent on innate and adaptive immune cells. (a–e) Mice
with B16F10 tumors were treated as indicated; tumors were isolated on days 14 and 21 after tumor cell inoculation, and quantification was performed
by flow cytometry (day 14, n = 15 mice per group; day 21, n = 9 mice per group). Data are from at least two independent experiments. Shown are
box plots (whiskers 5–95 percentile) for tumor-infiltrating CD8 + T cells (a), CD4+FoxP3− T cells (b), the ratio of CD8+ T cells to CD4+CD25hiFoxp3+
Treg cells (c), CD11b+LyG+LyClow polymorphonuclear (PMN) cells (d), and representative immunofluorescence images from tumors isolated from mice
6 d after the initiation of the indicated treatment (e). Scale bar, 100 µm. (f) Survival over time with depleting antibodies specific for the indicated
surface markers administered i.p. beginning 1 d before initiation of AIPV therapy to deplete macrophages (CSF1R), NK cells (NK1.1), CD8 + T cells
(CD8) or neutrophils (Ly6G) (n = 5 mice per group for CD8 depletion (dep); n = 10 mice per group for all other groups). Data were compiled from two
independent experiments. Arrows indicate treatment time points. *P < 0.05; **P < 0.01; ***P < 0.001; by Welch’s t-test with Bonferroni correction
(versus untreated) (a–d) or by log-rank (Mantel–Cox) test (versus AIPV) (f).

(Treg) cells (Fig. 3c and Supplementary Fig. 6g,h), a notable outcome dendritic cells (DCs) in mice27, and AIPV-treated Batf3−/− mice failed
given the potential for IL-2 treatment to increase Treg cell numbers. to reject tumors, indicating a requirement for cross-presenting DCs for
Of interest, however, was that no single leukocyte population exam- the response to therapy (Fig. 4a). Tumor-specific antibodies promote
ined correlated exactly with therapeutic outcome, as several treatment cross-presentation of tumor antigens28. To further explore the role
subcombinations elicited infiltration of individual cell populations of tumor-specific antibodies in this process, we treated mice bearing
equivalent to that with AIPV treatment. Immunohistochemical analy- GFP-expressing B16F10 tumors with AIPV or with subcombinations
sis of tumors from treated mice revealed that CD8+ T cells were present of the four components and analyzed the uptake of GFP and fluores-
at high density throughout the AIPV-treated tumors, with less effective cently labeled TA99 in DCs from TDLNs (Fig. 4b,c). We examined
triple-combination therapies eliciting progressively fewer CD8+ cells in two Batf3-dependent cross-presenting DC populations that may have
the bulk tumor mass (Fig. 3e and Supplementary Fig. 6e,f). Antibody distinct roles in tumors, CD8-α+ DCs and CD103+ DCs29–31. Labeled
depletion experiments showed that CD8+ T cells were critical to tumor TA99 accumulated in these DC populations, and treatment combina-
rejection (Fig. 3f and Supplementary Fig. 7a). Macrophage, NK cell tions that included TA99 significantly increased GFP uptake by both
or neutrophil depletion also led to significant reductions in overall CD8-α+ and CD103+ DCs over that of untreated tumors (Fig. 4d–g).
survival rates, whereas IL-5 blockade or CD4+ T cell depletion did Furthermore, ovalbumin (OVA)-expressing B16F10 tumors from mice
not affect efficacy (Fig. 3f and Supplementary Fig. 7a,b). Thus, AIPV treated with AIPV (vaccinating against Trp2) induced T cell responses
treatment elicited substantial remodeling of the tumor microenviron- specific for the SIINFEKL OVA peptide that were substantially greater
ment with contributions from diverse effector cells. than responses observed in untreated or IPV-treated tumors, indicat-
ing a role for the antibody in priming T cell responses to new tumor
Therapy promotes cross-presentation to generate de novo antigens (Fig. 4h,i). Further supporting the conclusion that AIPV
T cell responses treatment induced a broad T cell response to multiple tumor anti-
We next assessed whether de novo adaptive immune responses gens, splenic T cells from B16F10-tumor-bearing mice treated with
specific for antigens that were not encoded by the vaccine were primed AIPV produced IFN-γ after restimulation with either parental B16F10
by AIPV treatment. Given the critical role of CD8 + T cells in the cells or a Trp2-deleted B16F10 cell line (Supplementary Fig. 8a,b)
therapeutic effect, we first analyzed the effect of AIPV treatment (Fig. 4j,k). In addition, 50% of AIPV-treated mice that were cured
on antigen presentation in the tumor-draining LNs (TDLNs). Batf3 of primary tumors also rejected a re-challenge with Trp2-deleted
is a transcription factor required for development of cross-presenting B16F10 cells on day 125 (Supplementary Fig. 8c). These data suggest

 advance online publication  nature medicine


articles

a b c AIPV IPV
d e
100 1,200 *** 150
*** *** ***

TA99 CD8-α DCs


5 53.2% 13.5% 0.1% 0%
10 10
5

per 10 LN cells
** ***

Alexa647 TA99
Survival (%)

75 0 8 10 4 900 100

GFP MFI
10 4

+
AIPV (WT) 10
50 AIPV (Batf3–/–)*** 3
10 10
3 600
Untreated 50

6
6 – AIPV, TDLN
25 10 B16 0 0 300

+
GFP labeled A analysis
0 2 0

0
0
2

3
0 0 – – – + – + – + TA99
–1

10

10

0
10

10
0 15 30 45 60 75

–1

ed
V
P
AI V
PV
AIP AIV AIPV

ed
V
Time (d)

IP
AI
AI
GFP

IP
at

at
re

re
nt

nt
U

U
f 1,500 g 250 h i 4
* j AIPV +
k 1,800
DCs per 10 LN cells

*** *** *** ***

+
200 B16F10

SFU per 10 splenocytes


% teramer of CD8
+

** 2.99% 3
TA99 CD103

1,000
GFP MFI

150 600
***
6

100 2 AIPV + **

+
+

500 B16–Trp2-KO 500


50

6
1
0 0 250
1.03% AIPV +
– – – + – + – + TA99

SIINFEKL tetramer (PE)


ed

V
P
V
PV

0 TC-1
IP
AI
AI
at

AI

ed

AIP AIV AIPV


re

0
IP

ed

V
PV
at
nt

IP
re
U

at

AI

Tr -1
+ -KO

B1 + 0
-1
+ -KO

10
nt
© 2016 Nature America, Inc., part of Springer Nature. All rights reserved.

re

1
U

6– TC

6– TC
nt

PV AI 16F

6F
v e p2

PV rp2
U

B1
B1 +

T
ve

PV
ai
0.47%

ai

AI
+

+
N
ve
ai

AI
N
CD8 (APC)

Figure 4  Combination therapy elicits antibody-enhanced antigen spreading and de novo T cell responses. (a) Survival over time after AIPV treatment
was carried out in wild-type (WT) or Batf3−/− mice (n = 7 mice per group) bearing established B16F10 tumors as in Figure 1a; mice were euthanized
after the tumor area exceeded 100 mm2. Arrows indicate treatment time points. (b–g) B16–GFP cells (106) were injected s.c. (n = 10 mice per group),
and AIPV or the indicated ternary combination treatments were applied with Alexa-Fluor-647-labeled TA99 antibody, followed by TDLN isolation,
digestion and analysis by flow cytometry (b). Shown are representative scatter plots of CD11c + DCs (c), enumeration of TA99+CD11c+CD8-α+CD11b−
(d) and CD11c+CD103+ DCs (f), and the mean fluorescence intensity (MFI) of GFP within CD11c +CD8-α+CD11b− (e) and CD11c+CD103+ DCs (g) that
were gated on either TA99+ or TA99− cells. In d–g, box plots (whiskers 5–95 percentile) are shown. (h,i) B16-OVA cells (106) were injected s.c., and
mice were treated as indicated (with vaccine to Trp2). Staining with SIINFEKL–H-2K b tetramers on peripheral blood mononuclear cells was performed
on day 15. Shown are representative flow plots of TDLNs from untreated (bottom), IPV-treated (middle), or AIPV-treated (top) mice (h) and box plots
(whiskers 5–95 percentile) (i) from one of two independent experiments (n = 10 mice per group). (j,k) Representative images of an ELISPOT assay (j)
and quantification of B16F10-reactive splenocytes (box plots, with whiskers showing 5–95 percentile; n = 6 mice per group) (k) to a Trp2-knockout
(KO) B16F10 line generated using CRISPR–Cas9 technology (Supplementary Fig. 8), parental B16F10 cells or control TC-1 cells 6 d after AIPV-treated
or naive mice were challenged with 105 B16F10 tumor cells 75 d following primary tumor injection. SFU, spot-forming unit. Throughout, *P < 0.05,
**P < 0.01, ***P < 0.001; by Welch’s t-test versus untreated with Bonferroni correction (d,f), Welch’s t-test versus the TA99− fraction within the same
treatment group (e,g), analysis of variance (ANOVA) with Bonferroni’s post hoc test (i) or Student’s t-test (k).

that the antibody component of AIPV promoted epitope spreading, mice, suggesting that the endogenous antibody response was not
leading to functional de novo T cell responses to tumor antigens that essential for the therapeutic effect of the AIPV regimen (Fig. 5f,g).
were not encoded by the T cell vaccine. Taken together, these experiments indicate that AIPV broadly pro-
moted both antibody and T cell responses to epitopes that were not
Therapy-induced endogenous antibody responses directly targeted by the therapy components.
In parallel, we evaluated endogenous antibody responses in AIPV-
treated mice. In all three transplanted tumor models, AIPV therapy Efficacy in an autochthonous genetically engineered mouse model
elicited antibodies that bound to the tumor cells (Fig. 5a). To analyze Transplanted tumors do not closely mimic the histology of human
the endogenous IgG response over time, we developed an approach to cancers, and they often have a high mutational burden that gen-
deplete the injected antibody from sera recovered from the mice under- erates potential neoantigens. To assess the efficacy of AIPV treat-
going treatment (Supplementary Fig. 9a–f). By using this strategy, we ment in a model that more closely mimics the pathophysiology of
found that both AIPV and the less effective triple subcombinations human disease and to simultaneously determine the effect of a low
elicited tumor-specific antibodies, which were detectable as early as frequency of neoantigens, we used the BrafCA PtenloxPTyr::CreERT2
7 d following the start of treatment and that increased in binding signal inducible melanoma model32 (which we hereafter refer to as Braf/Pten
over 30 d for combinations that promoted better survival (Fig. 5b and mice). In this model, topical application of tamoxifen results in
Supplementary Fig. 9f). An immunoblot with sera from AIPV-treated expression of oncogenic BrafV600E and deletion of tumor suppres-
mice against B16F10 lysate revealed that these endogenous antibod- sor gene Pten in melanocytes, resulting in malignant melanoma
ies recognized numerous antigens (Fig. 5c). These therapy-induced formation 21–28 d later. We first enhanced the potency of AIPV
antibodies were functional, as serum transferred from AIPV-treated by increasing the number of antigens targeted by the vaccine com-
mice protected naive mice against intravenous B16F10 challenge ponent33. AIPV therapy using a trivalent amphiphile–vaccine tar-
(Fig. 5d,e). However, administration of AIPV therapy to B16F10- geting gp100, Trp1 and Trp2 cured 100% of mice bearing B16F10
tumor-bearing µMT mice, which lack mature B cells due to a tar- tumors with mean initial tumor sizes of 50 mm2 (Fig. 6a,b). To test
geted mutation in immunoglobulin heavy constant mu (Ighm), led to this combination in genetically engineered mice, tumors were
a similar frequency of tumor regressions as that observed in wild-type induced by painting tamoxifen on the ears of Braf/Pten mice,

nature medicine  advance online publication 


Articles

a Secondary
only
Naive
serum
AIPV
serum b c d f 150
PBS AIPV serum Naive µMT untreated
B16F10 serum µMT AIPV
serum TA99

Tumor area
10 100
Mr (kDa)

(mm2)
WT untreaed

Fold MFi
increase
245 AIPV WT AIPV
50
190 serum
135 0
0 10
3
10
4
10
5
1 TA99
100 control 0 10 20 30 40
DD-Her2/Neu
14 21 28 35 42
e
80 Time (d)
Time (d)
58 g

Number of tumor foci


AIP 75 100
*

Survival (%)
3 4
APV 46 75
0 10 10 AIV 50 µMT untreated
TC-1 IPV 32 50 µMT AIPV
AIPV 25 WT untreaed
25
Untreated 25 WT AIPV
22 0 0
17 0 20 40 60 80

m
m
99
PV ru
ru
TA
Time (d)

AI se
se
3 4 5
0 10 10 10 46 β-actin

ve
Anti–mouse IgG

ai
N
(Alexa Fluor 647 conjugated)
© 2016 Nature America, Inc., part of Springer Nature. All rights reserved.

Figure 5  AIPV therapy induces de novo endogenous tumor-specific antibody responses. (a) The indicated tumor cells were incubated with 5% serum
collected on day 150 from AIPV-treated mice that had previously rejected B16F10 (top), DD-Her2/neu (middle) or TC-1 (bottom) tumors, or from
age-matched naive mice. The cells were then washed, stained with an Alexa-Fluor-647-conjugated mouse-IgG-specific secondary antibody and
analyzed by flow cytometry. Shown are results for sera from individual treated animals, representative of two independent experiments of n = 4 (for
DD-Her2/neu and TC-1) and n = 8 (for B16F10) mice. (b) C57Bl/6 mice bearing B16F10 tumors were treated with the indicated combination therapies,
as in Figure 1a, using biotinylated TA99. Mice were bled weekly, and serum was depleted of TA99–biotin using streptavidin resin (Supplementary
Fig. 9). The binding of the remaining endogenous IgG to B16F10 cells was then measured by flow cytometry, as in a. Plotted is the fold change in MFI
of anti–mouse IgG over time + s.e.m. (n = 5 mice per group). (c) Western blot analysis of B16F10 tumor cell lysate with serum from AIPV-treated mice,
PBS-treated mice or TA99 with detection using an IRDye800-conjugated mouse-IgG-specific secondary antibody. β-actin was used as a loading control.
Shown is one representative of two independent experiments. (d,e) 350 µl serum from AIPV-treated mice (day 150) or age-matched naive mice (n = 4
mice per group) was incubated at 57 °C for 1 h to inactivate complement and then transferred into naive recipients; mice were then challenged with
2.5 × 105 B16F10 cells intravenously. Shown are representative images of lungs (d) and quantification of tumor foci (e) in lungs isolated 17 d after
transfer and tumor challenge. In e, data are mean counts ± s.e.m. Data shown are representative of two independent experiments. (f,g) B cell–deficient
µMT or WT C57Bl/6 mice were inoculated with B16F10 tumors and left untreated or treated with AIPV therapy as in Figure 1a. Shown are mean +
s.e.m. tumor area curves (f) and overall survival (g) (n = 8 mice per group for the AIPV-treated groups; n = 4 mice per group for the untreated groups).
Arrows indicate time points of treatment. *P < 0.05 by one-way ANOVA with Bonferroni post hoc test.

and after visible lesions were present (~4 weeks), treatment was initiated. CT26 and MC-38) could be cured by double- or triple-component
Melanomas grew progressively, and by week 10 they completely subcombinations of AIPV (unpublished data).
covered the ears of untreated mice (Fig. 6c). By contrast, trivalent- Many immunotherapy studies focus on CD8+ T cell responses to
vaccine AIPV promoted regression of induced tumors, leading to tumors, but important roles for innate immune effector cells have also
clearance of pigmented lesions in the majority of animals and a been described. Especially in the presence of tumor-opsonizing anti-
significant improvement in overall survival as compared to that in bodies, NK cells, neutrophils and macrophages can all contribute to
untreated mice (Fig. 6c,d). This result was coincident with the expan- direct antibody-dependent phagocytosis, reactive-oxygen-mediated
sion of T cells recognizing the three vaccine-encoded antigens in cytotoxicity against tumors, and secretion of inflammatory cytokines
peripheral blood (Fig. 6e) and with the infiltration of CD8+ T cells and chemokines35–37. This innate attack creates intertwined posi-
throughout tumors of AIPV-treated Braf/Pten mice (Fig. 6f). tive feedback loops—opsonized tumor antigen is released, which is
efficiently cross-presented by DCs11,38,39; inflammatory cytokines
DISCUSSION and immune complexes are released that activate DCs, stimulate
To date, complete elimination of large, poorly immunogenic tumors in their migration to draining LNs and promote adaptive effector func-
immunocompetent animal models has only been reliably achieved by tions21,40,41; and additional innate effectors and T cells are recruited
combination therapies using large numbers of adoptively transferred to the tumor21,40,42. Consistent with the importance of the innate
T cells12,17,34. Thus, it has remained an outstanding question whether immune response, maximal efficacy of AIPV therapy was depend-
an endogenous immune response can be stimulated to overcome ent not only on CD8+ T cells but also neutrophils, macrophages, NK
advanced tumors in a majority of animals. Here we demonstrate the cells and cross-presenting DCs. AIPV therapy completely failed in
capacity of endogenous immunity, elicited by a safe four-component Batf3-deficient mice and was, in fact, even less effective than AIP
immunotherapy, to regress large established tumors in the majority treatment, suggesting that the loss in efficacy in these knockout ani-
of mice in several transplanted models and a genetically engineered mals reflected more than loss of responsiveness to the vaccine compo-
model of melanoma. All four components incorporated in AIPV were nent (unpublished data). Although models of infectious disease and
required for treatment of several difficult-to-treat tumor models, systemic immunization have implicated CD8-α+ DCs as key cross-
and other treatment combinations were often less effective or very presenting cells for T cell immunity43,44, recent data has suggested that
toxic; for example, using anti-CD40 in place of the tumor-specific in growing tumors, a second Batf3-dependent DC lineage, CD103+
antibody caused inflammatory toxicity, resulting in death of treated DCs, has a critical role in tumor antigen presentation29,30. Consistent
mice (unpublished data). Notably, the models studied here were with these reports, we found that both CD8-α+ and CD103+ DCs
selected in part due to their resistance to immunotherapy treatment; acquired tumor antigen in a manner enhanced by the antibody
other commonly used immunogenic tumors (for example, EG7.OVA, component of AIPV.

 advance online publication  nature medicine


articles

a 120
b 100 f Untreated AIPV

Tumor area (mm2) 90 75 CD8

Survival (%)
AIPV, trivalent AIPV, trivalent
vaccine vaccine ***
60 50
Untreated Untreated
30 25

0 0
0 15 30 45 60 75 0 20 40 60 80
Time (d) Time (d)

c d
100

75 AIPV, trivalent
Untreated

vaccine ***

Survival (%)
50
Untreated
25

0 Actin
0 30 60 90
© 2016 Nature America, Inc., part of Springer Nature. All rights reserved.

Time after start of treatment (d)

e +
40 Trp-1
*** Trp-2
% IFN-γ of CD8

30
gp100
20 ***
AIPV

***
10

0
PV

ed
at
AI

re
nt
U

At treatment Week 3 Week 6

Figure 6  AIPV with a trivalent vaccine is curative for established B16F10 tumors and induces regression in Braf/Pten autochthonous melanoma.
(a,b) Mice with established B16F10 tumors were treated with AIPV, as outlined in Figure 1a, using a trivalent vaccine targeting Trp2, Trp1 and gp100.
Shown are mean + s.e.m tumor area measurements (a) and survival (b) (n = 10 mice per group). (c,d) BrafCAPtenloxPTyr::CreERT2 mice were painted
with 4-hydroxytamoxifen on one ear and treated with AIPV (including trivalent vaccine) beginning 28 d later, when visible lesions were apparent.
Shown are representative photographs of AIPV-treated or untreated ears at treatment (left), 3 weeks after treatment initiation (middle) and 6 weeks
after treatment initiation (right) (c) and survival over time (d) (n = 16 mice per group, compiled from four independent experiments). Mice were
euthanized when tumor coverage exceeded 90% of the ear. (e) On day 48 after tumor induction, peripheral blood from untreated or AIPV-treated
BrafCAPtenloxPTyr::CreERT2 mice was stimulated with peptide antigens for 6 h in the presence of brefeldin A, and intracellular cytokine staining
was performed to assess CD8+ T cell responses to each vaccine antigen. Shown are box plots (whiskers 5–95 percentile) (quantified as a fraction
of total CD8+ T cells; n = 7 mice per group). (f) BrafCAPtenloxPTyr::CreERT2 mice were painted with 4-hydroxytamoxifen on the left flank and treated
with AIPV (including trivalent vaccine) beginning on day 36 after tumor induction. Shown are representative immunofluorescence images of tumors
harvested on day 70 (n = 8 tumors per condition). Bottom panel is a composite image. Scale bar, 200 µm. In a,b,d, arrows indicate treatment
time points. ***P < 0.001 by Welch’s t-test (e) or log-rank (Mantel–Cox) test (b,d).

The treatment of multiple tumor models with AIPV therapy on tumors46. Despite the potential of IL-2 to expand Treg cells, we
revealed distinct hierarchies of importance for the four components. found that intratumoral CD8:Treg cell ratios were amplified relative
For example, the monoclonal antibody component was a critical con- to those in untreated tumors during AIPV therapy.
tributor to efficacy in the B16F10 model, but it was the least important Although it remains an understudied area, prior work has demon-
component in the DD-Her2/neu model. A key aspect of AIPV treat- strated that the number of antigens targeted by cancer vaccines can
ment is that this set of four agents collectively mounts an integrated be an important variable, with vaccines targeting more antigens lead-
response that overcomes tumor resistance mechanisms in all of the ing to greater antitumor efficacy33. In agreement with these data, we
models evaluated here, suggesting that the appropriate combination of found that the efficacy of AIPV therapy was sensitive to the valency
immune effectors can overcome a range of obstacles present in tumor of the vaccine; targeting a single epitope in the B16F10 model led to a
microenvironments. Notably, therapy excluding the extended-half- 75% cure rate (Fig. 1c), whereas a modest increase in vaccine valency
life IL-2 (APV treatment) showed greatly reduced efficacy in all three to target three melanocyte antigens led to cures in 100% of treated
transplanted tumor models, correlating with reduced abundance of mice (Fig. 6a,b). There is currently much enthusiasm for targeting
inflammatory cytokines and chemokines in tumors and fewer infil- neoantigens formed by mutations in tumors, but our data demonstrate
trating CD4+ and CD8+ T cells. The cell types that IL-2 must act that combination immunotherapy targeting only tumor-associated
on for therapeutic efficacy remain to be defined; however, besides self-antigens can elicit pronounced tumor regressions, even in the
supporting the proliferation and effector functions of T cells, IL-2 setting of a genetically induced mouse model bearing an intrinsically
is known to also drive expansion and enhance the responsiveness of low burden of mutations. However, AIPV treatment also promoted
NK cells to target cells45, promote neutrophilia and eosinophilia, and responses to additional antigens not present in the vaccine, which
induce the production of myriad additional cytokines and chemok- could protect against challenge with tumor cells lacking the vaccine-
ines that may support a combined innate and adaptive immune attack targeted antigens. We attempted to evaluate whether AIPV could

nature medicine  advance online publication 


Articles

be protective in the absence of antigen spreading by treating T cell Note: Any Supplementary Information and Source Data files are available in the
receptor (TCR)-transgenic pmel-1 mice17, which lack T cells capable online version of the paper.
of responding to peptides other than gp100; however, AIPV treat- Acknowledgments
ment of these mice using a vaccine specific for gp100 elicits severe This work was supported in part by the Koch Institute Support (core) grant P30-
inflammatory toxicity (unpublished observation). Antigen spreading CA14051 from the National Cancer Institute, the US National Institutes of Health
promoted by immunotherapy has also been observed in recent clini- (NIH) grant CA174795 (K.D.W.), the Bridge Project partnership between the
Koch Institute for Integrative Cancer Research and the Dana Farber–Harvard
cal studies of patients with melanoma47, and it may be important for Cancer Center (DF–HCC) (D.J.I.), the V Foundation (D.J.I.) and the Ragon
dealing with tumor heterogeneity and preventing escape by tumor Institute (D.J.I.). K.D.M. and J.M.E. are supported by the Fannie and John Hertz
antigen loss variants48. Foundation Fellowship; K.D.M., C.F.O., J.M.E., B.H.K. and E.F.Z. are supported
Endogenous antibody responses were primed by AIPV and other by NSF Graduate Research Fellowships; A.M.R. is supported by the NIGMS–NIH
Interdepartmental Biotechnology Training Program (NIH #T32GM008334); A.T.
combinations of the four monotherapies, and AIPV treatment induced is supported by the Siebel Scholarship; and G.L.S. was supported by the NIH with a
antibodies that were protective when transferred to naive recipients. Ruth L. Kirschstein National Research Service Award (CA180586). We thank T.C.
Despite these findings, B cells were not required for AIPV treatment Wu (Johns Hopkins University) for kindly providing the TC-1 tumor cells,
efficacy. We speculate that because the therapy itself includes a potent D. Sabatini (Whitehead Institute) for providing the Cas9 constructs and G. Dranoff
(Dana-Farber Cancer Institute) for providing the DD-Her2/neu and B16-OVA
monoclonal tumor-opsonizing antibody, the endogenous response
cells. We thank M. Ghebremichael (Ragon Institute of MGH, MIT and Harvard)
may be dispensable. An interesting question for future work will be for helpful statistical advice. We thank the Koch Institute Swanson Biotechnology
© 2016 Nature America, Inc., part of Springer Nature. All rights reserved.

whether the monoclonal antibody component can be removed from Center for technical support, specifically the applied therapeutics and whole-
the treatment regimen after the endogenous antibody response devel- animal imaging core facility, the histology and the flow cytometry core facilities.
ops, i.e., by the second or third treatment. D.J.I. is an investigator of the Howard Hughes Medical Institute.
AIPV incorporates two tumor-specific reagents, the LN-targeted AUTHOR CONTRIBUTIONS
peptide vaccine and a monoclonal opsonizing antibody. Although K.D.M., C.F.O., D.J.I. and K.D.W. designed the studies and wrote the manuscript;
it is still a challenge in some diseases, identification of T cell target K.D.M. and C.F.O. carried out experiments; E.F.Z., A.T., B.H.K., M.J.K. and H.S.
antigens that are shared by a proportion of tumors of a given type has assisted with protein production; J.M.E. assisted in generating the Trp2-knockout
line using CRISPR–Cas9; K.R., A.T., E.F.Z., W.A., R.T.W., A.M.R., R.L.K., N.K.M.
been well delineated for many cancers over the past 20 years by the and K.H. assisted with experiments; G.L.S. designed and analyzed intratumoral
cancer vaccine field49, and a number of pipelines for finding unique Luminex, and assisted in flow cytometry design and writing of the manuscript;
mutant neoantigens have recently been developed50,51. In contrast, D.A.L. aided with multivariate analysis of intratumoral Luminex data; G.L.S. and
identifying an effective opsonizing monoclonal antibody presents M.H.Z. conducted intratumoral Luminex and assisted with Braf/Pten mouse
experiments; S.K. assisted with immunofluorescence microscopy; and J.R.C.
a development hurdle for applying AIPV therapy to an arbitrary supplied 2.5F–Fc reagents.
given tumor. However, the demonstration that the integrin-binding
protein 2.5F–Fc, which contains an Fc domain for antibody effector COMPETING FINANCIAL INTERESTS
functions and which recognizes the αvβ3, αvβ5, and α5β1 integrins The authors declare competing financial interests: details are available in the online
version of the paper.
(which are overexpressed in a broad range of cancers52), suggests
that the 2.5F–Fc protein may be applicable to many tumor types. Reprints and permissions information is available online at http://www.nature.com/
Careful consideration of potential side effects is needed in the choice reprints/index.html.
of target antigens for translation of this approach. Targeting tumor-
1. Hodi, F.S. et al. Improved survival with ipilimumab in patients with metastatic
associated antigens carries the risk of autoimmunity; for example melanoma. N. Engl. J. Med. 363, 711–723 (2010).
immunotherapy targeting melanocyte self-antigens is known to 2. Topalian, S.L. et al. Safety, activity and immune correlates of anti-PD-1 antibody
induce vitiligo, uveitis and hearing loss in some patients due to in cancer. N. Engl. J. Med. 366, 2443–2454 (2012).
3. Larkin, J. et al. Combined nivolumab and ipilimumab or monotherapy in untreated
loss of normal melanocytes53,54. Sequencing of patient tumors for melanoma. N. Engl. J. Med. 373, 23–34 (2015).
neoantigen identification is an attractive alternative, and the pep- 4. Gajewski, T.F. The next hurdle in cancer immunotherapy: overcoming the non-T-
cell-inflamed tumor microenvironment. Semin. Oncol. 42, 663–671 (2015).
tide vaccine platform used here is well suited to such an approach. 5. Zou, W., Wolchok, J.D. & Chen, L. PD-L1 (B7-H1) and PD-1 pathway blockade for
Similarly, although antigen spreading as observed here may promote cancer therapy: mechanisms, response biomarkers and combinations. Sci. Transl.
tumor rejection, an important issue for future work is understanding Med. 8, 328rv4 (2016).
6. Mellman, I., Coukos, G. & Dranoff, G. Cancer immunotherapy comes of age. Nature
whether these de novo T cell responses are focused on tumor-specific 480, 480–489 (2011).
mutations or on self-antigens. 7. Drake, C.G. Combination immunotherapy approaches. Ann. Oncol. 23 (Suppl. 8),
41–46 (2012).
In summary, we have demonstrated that a rational combination 8. Sharma, P. & Allison, J.P. The future of immune checkpoint therapy. Science 348,
of four complementary immunomodulatory agents, each of which 56–61 (2015).
have modest antitumor efficacy individually, can lead to robust com- 9. Smyth, M.J., Ngiow, S.F., Ribas, A. & Teng, M.W.L. Combination cancer
immunotherapies tailored to the tumor microenvironment. Nat. Rev. Clin. Oncol.
plete responses in the setting of large, immunosuppressive tumors. 13, 143–158 (2016).
Rejection of established tumors relied on an orchestrated response 10. Curran, M.A., Kim, M., Montalvo, W., Al-Shamkhani, A. & Allison, J.P. Combination
CTLA-4 blockade and 4-1BB activation enhances tumor rejection by increasing T cell
invoking diverse effector mechanisms of the immune system. infiltration, proliferation and cytokine production. PLoS One 6, e19499 (2011).
Although using a quaternary treatment regimen will be challenging 11. Carmi, Y. et al. Allogeneic IgG combined with dendritic cell stimuli induce antitumor
for direct clinical translation because of the complexity of identifying T cell immunity. Nature 521, 99–104 (2015).
12. Hu-Lieskovan, S. et al. Improved antitumor activity of immunotherapy with BRAF and
optimal dosing levels and schedules for each component, insights MEK inhibitors in BRAFV600E melanoma. Sci. Transl. Med. 7, 279ra41 (2015).
from this successful protocol provide a framework for devising sim- 13. Rapoport, A.P. et al. Combination immunotherapy after ASCT for multiple myeloma
using MAGE-A3–poly-ICLC immunizations followed by adoptive transfer of vaccine-
pler regimens and extensions to other treatment modalities.
primed and co-stimulated autologous T cells. Clin. Cancer Res. 20, 1355–1365
(2014).
Methods 14. Lum, L.G. et al. Targeted T cell therapy in stage IV breast cancer: a phase 1 clinical
trial. Clin. Cancer Res. 21, 2305–2314 (2015).
Methods and any associated references are available in the online 15. Ribas, A. et al. Phase 1 study combining anti-PD-L1 (MEDI4736) with BRAF
version of the paper. (dabrafenib) and/or MEK (trametinib) inhibitors in advanced melanoma. ASCO

 advance online publication  nature medicine


articles

Annual Meeting Proceedings 33, abstract 3003 (American Society of Clinical 34. Nelson, M.H. et al. Toll-like receptor agonist therapy can profoundly augment the
Oncology, Alexandria, Virginia, USA, 2015). antitumor activity of adoptively transferred CD8+ T cells without host preconditioning.
16. Chen, G. et al. A feasibility study of cyclophosphamide, trastuzumab and an J. Immunother. Cancer 4, 6 (2016).
allogeneic GM-CSF-secreting breast tumor vaccine for HER2+ metastatic breast 35. Woo, S.R., Corrales, L. & Gajewski, T.F. Innate immune recognition of cancer.
cancer. Cancer Immunol. Res. 2, 949–961 (2014). Annu. Rev. Immunol. 33, 445–474 (2015).
17. Overwijk, W.W. et al. Tumor regression and autoimmunity after reversal of a 36. van Egmond, M. & Bakema, J.E. Neutrophils as effector cells for antibody-based
functionally tolerant state of self-reactive CD8+ T cells. J. Exp. Med. 198, 569–580 immunotherapy of cancer. Semin. Cancer Biol. 23, 190–199 (2013).
(2003). 37. Weiskopf, K. & Weissman, I.L. Macrophages are critical effectors of antibody
18. Klebanoff, C.A. et al. Determinants of successful CD8+ T cell adoptive immunotherapy therapies for cancer. MAbs 7, 303–310 (2015).
for large established tumors in mice. Clin. Cancer Res. 17, 5343–5352 38. DiLillo, D.J. & Ravetch, J.V. Differential Fc receptor engagement drives an antitumor
(2011). vaccinal effect. Cell 161, 1035–1045 (2015).
19. DuPage, M. et al. Endogenous T cell responses to antigens expressed in lung 39. van Montfoort, N. et al. Antigen storage compartments in mature dendritic cells
adenocarcinomas delay malignant tumor progression. Cancer Cell 19, 72–85 facilitate prolonged cytotoxic T lymphocyte cross-priming capacity. Proc. Natl. Acad.
(2011). Sci. USA 106, 6730–6735 (2009).
20. Stromnes, I.M. et al. T cells engineered against a native antigen can surmount 40. Guilliams, M., Bruhns, P., Saeys, Y., Hammad, H. & Lambrecht, B.N. The function
immunologic and physical barriers to treat pancreatic ductal adenocarcinoma. of Fc-γ receptors in dendritic cells and macrophages. Nat. Rev. Immunol. 14,
Cancer Cell 28, 638–652 (2015). 94–108 (2014).
21. Zhu, E.F. et al. Synergistic innate and adaptive immune response to combination 41. Clatworthy, M.R. et al. Immune complexes stimulate CCR7-dependent dendritic cell
immunotherapy with anti-tumor-antigen antibodies and extended serum half-life migration to lymph nodes. Nat. Med. 20, 1458–1463 (2014).
IL-2. Cancer Cell 27, 489–501 (2015). 42. Albanesi, M. et al. Neutrophils mediate antibody-induced antitumor effects in mice.
22. Liu, H. et al. Structure-based programming of lymph node targeting in molecular Blood 122, 3160–3164 (2013).
vaccines. Nature 507, 519–522 (2014). 43. Allan, R.S. et al. Migratory dendritic cells transfer antigen to a lymph-node-resident
© 2016 Nature America, Inc., part of Springer Nature. All rights reserved.

23. Moore, S.J. et al. Engineered knottin peptide enables non-invasive optical imaging dendritic cell population for efficient CTL priming. Immunity 25, 153–162 (2006).
of intracranial medulloblastoma. Proc. Natl. Acad. Sci. USA 110, 14598–14603 44. den Haan, J.M., Lehar, S.M. & Bevan, M.J. CD8+ but not CD8− dendritic cells
(2013). cross-prime cytotoxic T cells in vivo. J. Exp. Med. 192, 1685–1696 (2000).
24. Phan, G.Q., Attia, P., Steinberg, S.M., White, D.E. & Rosenberg, S.A. Factors 45. Gasteiger, G. et al. IL-2-dependent tuning of NK cell sensitivity for target cells is
associated with response to high-dose interleukin-2 in patients with metastatic controlled by regulatory T cells. J. Exp. Med. 210, 1167–1178 (2013).
melanoma. J. Clin. Oncol. 19, 3477–3482 (2001). 46. List, J. et al. Cytokine responses to intraventricular injection of interleukin-2 into
25. Rose, S., Misharin, A. & Perlman, H. A novel Ly6C- and Ly6G-based strategy to patients with leptomeningeal carcinomatosis: rapid induction of tumor necrosis
analyze the mouse splenic myeloid compartment. Cytometry A 81, 343–350 factor–α, interleukin 1–β, interleukin-6, γ-interferon and soluble interleukin-2
(2012). receptor (Mr 55,000 protein). Cancer Res. 52, 1123–1128 (1992).
26. Ostrand-Rosenberg, S. & Sinha, P. Myeloid-derived suppressor cells: linking 47. Carreno, B.M. et al. Cancer immunotherapy. A dendritic cell vaccine increases the
inflammation and cancer. J. Immunol. 182, 4499–4506 (2009). breadth and diversity of melanoma neoantigen-specific T cells. Science 348,
27. Hildner, K. et al. Batf3 deficiency reveals a critical role for CD8-α+ dendritic cells 803–808 (2015).
in cytotoxic T cell immunity. Science 322, 1097–1100 (2008). 48. DuPage, M., Mazumdar, C., Schmidt, L.M., Cheung, A.F. & Jacks, T. Expression of
28. Rafiq, K., Bergtold, A. & Clynes, R. Immune-complex-mediated antigen presentation tumor-specific antigens underlies cancer immuno-editing. Nature 482, 405–409
induces tumor immunity. J. Clin. Invest. 110, 71–79 (2002). (2012).
29. Broz, M.L. et al. Dissecting the tumor myeloid compartment reveals rare activating 49. Vigneron, N., Stroobant, V., Van den Eynde, B.J. & van der Bruggen, P. Database
antigen-presenting cells critical for T cell immunity. Cancer Cell 26, 638–652 of T cell–defined human tumor antigens: the 2013 update. Cancer Immun. 13, 15
(2014). (2013).
30. Roberts, E.W. et al. Critical role for CD103+/CD141+ dendritic cells bearing CCR7 50. Yadav, M. et al. Predicting immunogenic tumor mutations by combining mass
for tumor antigen trafficking and priming of T cell immunity in melanoma. Cancer spectrometry and exome sequencing. Nature 515, 572–576 (2014).
Cell 30, 324–336 (2016). 51. Gubin, M.M. et al. Checkpoint blockade cancer immunotherapy targets tumor-
31. Salmon, H. et al. Expansion and activation of CD103+ dendritic cell progenitors at specific mutant antigens. Nature 515, 577–581 (2014).
the tumor site enhances tumor responses to therapeutic PD-L1 and BRAF inhibition. 52. Desgrosellier, J.S. & Cheresh, D.A. Integrins in cancer: biological implications and
Immunity 44, 924–938 (2016). therapeutic opportunities. Nat. Rev. Cancer 10, 9–22 (2010).
32. Dankort, D. et al. BrafV600E cooperates with Pten loss to induce metastatic 53. Morgan, R.A. et al. Cancer regression in patients after transfer of genetically
melanoma. Nat. Genet. 41, 544–552 (2009). engineered lymphocytes. Science 314, 126–129 (2006).
33. Mendiratta, S.K. et al. Therapeutic tumor immunity induced by polyimmunization 54. Johnson, L.A. et al. Gene therapy with human and mouse T cell receptors mediates
with melanoma antigens gp100 and TRP-2. Cancer Res. 61, 859–863 cancer regression and targets normal tissues expressing cognate antigen. Blood
(2001). 114, 535–546 (2009).

nature medicine  advance online publication 


ONLINE METHODS as measured by the QCL-1000 chromogenic LAL assay (Lonza). The IL-15
Mice. B6 mice (C57BL/6NTac) were purchased from Taconic. Balb/c mice superagonist60 was produced by transiently transfecting HEK293 cells with a
(BALB/cJ), Batf3−/− mice (B6.129S(C)-Batf3tm1Kmm/J), Braf/Pten mice plasmid encoding histidine (His)-tagged IL-15 superagonist and purified using
(B6.Cg-Braftm1MmcmPtentm1HwuTg(Tyr-CreERT2)13Bos/BosJ), and mT/mG Ni–NTA agarose (Qiagen) on a PD-10 column (Bio-Rad) followed by further
mice (B6.129(Cg)-Gt(ROSA)26Sor tm4(ACTB-tdTomato,-EGFP)Luo/J) were pur- purification using a His-Spin Protein Miniprep (Zymo Research). Anti-TGF-β,
chased from the Jackson Laboratory. Mice used in the inducible cancer model a homodimeric fusion between murine IgG2a Fc and the extracellular domain
(Braf/Pten-TG) were crosses of Braf/Pten and mT/mG mice that were bred of the type II TGF-β receptor, was produced by transient transfection of HEK293
in-house, and they had the following genotype: Braftm1Mmcm +/−Ptentm1Hwu cells and purified by protein A.
+/+Tg(Tyr-CreERT2)13Bos+Gt(ROSA)26Sortm4(ACTB-tdTomato,-EGFP)Luo+/+, where

‘+’ indicates presence of the mutant or transgenic allele. All Braf/Pten mice Tumor inoculation and subcutaneous tumor therapy. An inoculum of 106
were genotyped via Transnetyx. Mice were used in studies when 6–8 weeks tumor cells was injected s.c. on the flank of mice in 50 µl sterile PBS. Eight days
old. All animal work was conducted under the approval of the Massachusetts following injection, treatment was initiated as indicated (Fig. 1a). The tumor-
Institute of Technology (MIT) Division of Comparative Medicine in accord- targeting antibody (A) was administered at 100 µg per dose (i.p.) for the B16F10
ance with federal, state and local guidelines. and DD-Her2/neu models. 2.5–Fc was administered at 500 µg per dose (i.p.).
MSA–IL2 (I) was administered i.p. at 30 µg (6 µg molar equivalent IL-2) per dose.
Cells. B16F10 cells were purchased from ATCC. TC-1 cells were kindly provided Anti-PD-1 (P) (clone RMP1-14, BioXCell) was administered at 200 µg (i.p.).
by T.C. Wu (Johns Hopkins University). B16-OVA and DD-Her2/neu cells were The vaccine, composed of 1.24 nmol amph–CpG and 20 µg of amph–peptide,
kindly provided by G. Dranoff (Dana-Farber Cancer Center). B16-GFP-Luc cells was administered s.c. at the base of the tail, with half of the dose given on each
© 2016 Nature America, Inc., part of Springer Nature. All rights reserved.

were generated as previously described55. DD-Her-2/neu breast tumor cells were side. For experiments exploring other quaternary combos, the following doses
derived from a spontaneous breast tumor in a Balb/c Her-2/neu (Balb-NeuT) were used as indicated in Supplementary Figure 3: anti-CTLA-4 (clone 9D9,
transgenic mouse, in which mouse mammary tumor virus (MMTV)-promoter- BioXCell) was administered at 200 µg per dose i.p.; IFN-α was administered at
driven breast-epithelium-specific expression of the activated oncogenic form 50 µg per dose i.p.; IL-15 superagonist was administered at 4 µg per dose i.p.;
of rat Her-2/neu drives progressive development of breast tumors in transgenic and anti-TGF-β was administered at 100 µg per dose i.p. Mice were randomized
Her-2/neu female mice56. The DD cell line was found to lose Her-2/neu expres- into treatment groups on day 8 following tumor inoculation, immediately before
sion in vitro, and thus cells were infected with the pMFG-Her-2/neu virus to treatment. Tumor size was measured as an area (longest dimension × perpendic-
achieve stable expression of high levels of oncogenic rat Her-2/neu both in vitro ular dimension) three times weekly, and mice were euthanized when tumor area
and in vivo. HEK293 cells were purchased from Life Technologies. exceeded 100 mm2. Mice that rejected tumors were re-challenged as indicated
Tumor cell lines were cultured in complete Dulbecco’s modified Eagle’s with 105 tumor cells by subcutaneous injection on the opposite flank.
medium (DMEM, GE Healthcare Life Sciences; supplemented with 10%
FBS, 100 units/ml penicillin, 100 µg/ml streptomycin, and 4 mM l-alanyl-l- Flow cytometry. Antibodies to CD8-α (53-6.7), IFN-γ (XMG1.2), TNF-α
glutamine) with the exception of TC-1 cells, which were cultured in complete (MP6-XT22), CD3-ε (500A2), IgM (RMM-1), CD19 (6D5), NK1.1 (PK136),
RPMI medium (GE Healthcare Life Sciences). T cells and splenocytes were Ly-6G (1A8), F4/80 (BM8), CD11b (M1/70), CD25 (PC61), PD-1 (29F.1A12),
cultured in RPMI with 10% heat-inactivated FBS, 20 mM HEPES, 1 mM sodium Tim-3 (B8.2C12), Ly-6C (HK1.4) and CD64 (X54-5/7.1) were purchased
pyruvate, 0.05 mM β-mercaptoethanol, 100 units/ml penicillin, 100 µg/ml strep- from BioLegend. Antibodies to CD4 (RM4-5) and Foxp3 (FJK-16s) were pur-
tomycin, 2 mM l-alanyl-l-glutamine, and 1× minimal essential medium (MEM) chased from eBioscience. Trp2 tetramer (iTAg Tetramer/PE H-2Kb TRP2)
non-essential amino acids. Hybridomas were cultured in CD Hybridoma AGT and OVA tetramer (iTAg Tetramer/PE H-2Kb OVA) were purchased from
Medium (Life Technologies). HEK293 cells were cultured in Freestyle medium MBL. Tetramer staining was performed in buffer containing 50 nM dasat-
(Life Technologies). All cell lines and assay cultures were maintained at 37 °C inib to enhance staining. Viability was assessed by LIVE/DEAD Fixable
and 5% CO2. All cells were tested regularly for mycoplasma contamination and Aqua (Life Technologies). TA99 was labeled with Alexa Fluor 647 NHS Ester
for rodent pathogens, and none used tested positive at any point. (Life Technologies) to generate TA99–647. Foxp3 was stained using the
Transcription Factor Buffer Set from BD.
Vaccine and protein production. Amphiphile–CpG (amph–CpG) was pro- Intracellular cytokine staining (ICS) was performed as described previ-
duced, as previously described, by solid-phase synthesis of a class B CpG 1826 ously10. Peptides used for restimulation were 10 µg/ml of the relevant antigen:
oligonucleotide with G2 spacer (5′-diacyl lipid–*G*G*T*C*C*A*T*G*A* C* Trp2180–188 (SVYDFFVWL), Her2/Neu66–74 (TYVPANASL), Tyrp-1455–463native
G*T*T*C*C*T*G*A*C*G*T*T-3′) conjugated via the 5′ end to an 18-carbon (TAPDNLGYA), or gp10025–33native (EGSRNQDWL). Immune cell infiltrates
diacyl tail10. Amphiphile–peptides (amph–peptides) were produced as previ- of tumors were analyzed as previously described9; briefly, tumors were resected
ously described10, via conjugation of cysteine residues of peptide antigens to from mice on day 14 or 21, weighed, and mechanically disrupted to generate a
1,2-distearoyl-sn-glycero-3-phosphoethanolamine-N-[maleimide(polyethylene single-cell suspension. Cells were stained to identify infiltrating immune cells
glycol)-2000] (Avanti Polar Lipids). Vaccine sequences are as follows: and quantified using flow cytometry. Shown are cell counts normalized by tumor
Trp2180–188 (CSVYDFFVWL); Her2/Neu66–74 (p66, CTYVPANASL), HPV mass. For antibody- and GFP-uptake experiments, lymph nodes were resected
E743–62 (GQAEPDRAHYNIVTFCCKCD), Tyrp1 opt455–463 (optimized from tumor-bearing mice on day 10, digested with collagenase and dispase in
A463M57, CTAPDNLGYM), gp100opt20–39 (optimized S27P, EGP long58, the medium at 37 °C for 20 min, and mechanical disruption was used to gener-
CAVGALEGPRNQDWLGVPRQL). TA99 and MSA–IL-2 were produced ate a single-cell suspension. Cells were stained to identify DC populations and
in HEK293 cells as previously described9. 2.5–Fc was generated by fusing an run on the cytometer. Cells were analyzed using BD FACS LSR II, BD FACS
integrin (αVβ3, αVβ5, and α5β1)-binding domain to a mouse IgG2a Fc region LSR Fortessa, and BD FACSCanto flow cytometers, and data were analyzed
to create an antibody-like tumor-targeting molecule23. 2.5–Fc was produced by using FlowJo.
transient transfection of HEK293 cells and purified using protein A. The 7.16.4
monoclonal antibody to rat Her2/neu was generated from hybridoma 7.16.4 Depletions. Cellular subsets were depleted by administering 400 µg of deplet-
(purchased from ATCC) and purified by using protein A. IFN-α was expressed ing antibody i.p. twice weekly beginning 1 d before therapy as indicated: CD8
as a SUMO fusion using the pE-SUMOpro vector (LifeSensors) in Rosetta-gami T cells with anti-CD8-α (clone 2.43, BioXCell), CD4 T cells with anti-CD4
2 (DE3) competent cells (Novagen). After immobilized metal-affinity column (clone GK1.5, BioXCell), NK cells with anti-NK1.1 (clone PK136, BioXCell),
purification using TALON metal-affinity resin according to the manufacturer’s neutrophils with anti-Ly-6G (clone 1A8, BioXCell). Macrophages or eosinophils
instructions (Clontech Laboratories), the SUMO tag was removed by incuba- were depleted using 300 µg anti-CSF1R (clone AFS98, BioXCell) every other
tion with SUMO protease and reapplication on TALON resin as detailed previ- day or 1 mg anti-IL-5 (clone TRFK5, BioXCell) weekly, respectively9. Cellular
ously59. Finally, the protein was passed through Detoxi-Gel endotoxin removal depletions of CD8 T cells, CD4 T cells, neutrophils and NK cells were confirmed
resin (Thermo Scientific) until endotoxin levels were below 0.1 total EU/dose by flow cytometry of PBMC (Supplementary Fig. 7).

nature medicine doi:10.1038/nm.4200


Autochthonous tumor model induction and therapy. BrafCAPtenloxPTyr::Cre and sequencing confirmed deletion of the 283-bp fragment containing the Trp2
ERT2 mice27 on the C57BL/6 background61 were crossed with mT/mG mice62 peptide sequence (data not shown).
to generate Braf/Pten-TG mice. To induce tumors, 2 µl of 5 mg/ml tamoxifen
was administered to the left ear on three consecutive days. Tumors were allowed Enzyme-linked immunospot assay. Target B16F10, TC-1, or B16-Trp2-KO
to develop for 24–26 d, at which time visible pigmented tumors were present. cells were treated with 500 U/ml mIFN-γ (Peprotech) for 12 h, then irradiated
Treatment schedule and doses were the same as for the B16F10 subcutaneous (120 Gy). Effector cells were splenocytes isolated from AIPV-treated mice that
model, except that the vaccine used was a combination of three amph–peptides had rejected tumors 6 d after re-challenge with 106 B16F10 cells. A mouse IFN-γ
(15 µg amph–gp100, 15 µg amph–Trp1, and 15 µg amph–Trp2) and 1.24 nmol ELISPOT Kit (BD) was used. Targets cells were seeded at 25,000 cells per well.
amph–CpG. Mice were euthanized when pigmented lesions covered greater Effector cells were seeded at 106 cells per well. Plates were wrapped in foil and
than 90% of the ear. cultured for 24 h, then developed according to manufacturer’s protocol. Plates
were scanned using a CTL-ImmunoSpot Plate Reader, and data were analyzed
Liver enzyme measurements. Serum was isolated from mice 36 h after using CTL ImmunoSpot Software.
treatment, and the liver enzymes AST and ALT were quantified by using a
colorimetric Aspartate Aminotransferase Activity Assay Kit (Sigma Aldrich) Immunoblot analysis. B16F10 cell lysate was run on an SDS–PAGE gel,
or Alanine Aminotransferase Activity Assay Kit (Sigma Aldrich), respectively, transferred to nitrocellulose membranes, blocked for 1 h at 25 °C, and then
according to the manufacturer’s protocol. stained with serum from treated mice and rabbit anti-β-actin overnight at 4 °C.
Membranes were then washed and stained with goat anti-mouse-IRDye800 and
Endogenous antibody detection and serum transfer. For endogenous anti-rabbit-IRDye680 for 1 h at 25 °C. Imaging was performed on an Odyssey
© 2016 Nature America, Inc., part of Springer Nature. All rights reserved.

antibody-binding measurements following therapy, B16F10 cells, TC-1 cells, scanner (Licor). As a control, TA99 was used in place of mouse serum in the
or DD-Her2/neu cells were incubated with 5% serum from AIPV-treated protocol above to show the presence of Trp1 in the B16F10 lysate.
mice (day 150) or age-matched naive mice in PBS for 30 min at 4 °C, washed
twice with PBS, 1% BSA, and 5 mM EDTA, and then incubated with Alexa- Tumor cytokine Luminex assay. Tumors were harvested at day 17 (2 d after
Fluor-647-labeled anti-mouse-IgG for 20 min at 4 °C. Cells were washed in second treatment). All tumors from a treatment group were collected, up to
PBS, 1% BSA, and 5 mM EDTA, and DAPI was included to exclude dead cells. a maximum of ~250 mg, for processing in a bead beater tube. Tissue samples
The cells were subsequently run on the BD FACSCanto for analysis. To measure were lysed in lysis buffer (5 µl/mg tumor): 50 mM Tris-HCl (pH 7.5), 10% glyc-
endogenous antibody responses during therapy in the B16F10 model, the TA99 erol, 150 mM NaCl, 1% NP-40, and freshly supplemented with Halt Protease
antibody, which was used as part of the therapy, needed to be depleted from and Phosphatase Inhibitor Cocktail (Life Technologies). Lysates were aliquoted
serum samples. To this end, biotinylated TA99 was used for therapy, which and stored at −80 °C until analysis. Samples were diluted 1:1 with Assay Buffer
could be removed from the recovered sera using streptavidin resin. TA99 was and assayed using a Luminex bead-based ELISA (MILLIPLEX MAP Mouse
biotinylated using an N-hydroxy-succinimide-activated biotin according to Cytokine/Chemokine Magnetic Bead Panel, Millipore), following manufac-
the manufacturer’s protocol (Thermo Fisher Scientific), and excess biotin was turer’s instructions with the following modifications: magnetic capture beads
removed using desalting columns (Zeba). Biotin labeling was quantified by using and detection antibodies were both used at 0.2× the recommended amounts.
the colorimetric Biotin Quantitation Kit (Pierce), and a degree of labeling of 5.1 Concentrations were normalized to total protein input. All samples and analytes
biotins per TA99 molecule was achieved for the experiments shown. For serum were above the limit of detection.
antibody measurement during therapy, serum was isolated from mice once a
week and incubated with an excess of streptavidin resin (GenScript) for 1 h at Histology. Tumor inoculation and treatment were performed following the
25 °C. Serum was frozen, stored at −80 °C, and thawed on the day of the binding timeline in Figure 1a. For hematoxylin and eosin (H&E) staining, mice were
assay, which was performed as described above. euthanized 3 d after the second treatment, and tumors were fixed in 10% forma-
For serum transfer studies, serum was obtained from AIPV-treated mice (day lin, embedded in paraffin, and stained with hematoxylin and eosin. For immun-
150) or age-matched naive mice, and complement was inactivated by incuba- ofluorescence, tumors were isolated on day 14 and processed as previously
tion for 30 min at 57 °C. Serum (350 µl) or TA99 (200 µg) was injected i.p. into detailed64. Flank tumors were used to analyze infiltrate by immunofluorescence
naive recipients 6 h before injection of 250,000 B16F10 cells intravenously (i.v.) in the BrafCAPtenloxPTyr::CreERT2 model to allow for higher quality sectioning.
through the tail vein. Lungs were isolated 17 d later, and lung nodules were For staining, anti-CD8 (CT-CD8a, Cedar Lane), DAPI, and phalloidin were
counted in a blinded fashion. used. The images were acquired using an Olympus Fluoview FV1200 microscope
equipped with 10× (NA 0.40) and 30× (NA 1.05) objectives, and optimum lasers
Generation of the Trp2-knockout cell line. B16-Trp2-KO cells were gener- and filter sets. The images were acquired under identical acquisition settings and
ated using CRISPR–Cas9. B16-GFP-Cas9 cells were generated by transduc- subsequently processed using Fiji image analysis software.
tion of B16F10 cells with lentivirus expressing humanized SpCas9-P2A-EGFP
from the promoter of the EEF1A1 gene (which encodes elongation factor–1α) Multivariate analysis of cytokine data. Cytokine levels for each tumor were
(unpublished data; kindly provided by Tim Wang in Dr. David Sabatini’s Lab normalized to the median of the ‘untreated’ controls and log2-transformed. Heat
at the Whitehead Institute). Clones were isolated by single-cell flow cytometry maps and clustering trees were generated using the clustergram function in
for GFP-positive cells. A clone stably expressing Cas9 was designated B16- Matlab (Mathworks). Euclidean distance was used for hierarchical clustering of
GFP-Cas9. Guide RNA (sgRNA) expression vectors were created by cloning cytokines only. The resulting dendrogram was pruned by collapsing leaves with
a human U6 promoter and sgRNA sequences into a minimal vector with an Euclidean distance <95% confidence interval for all 32 potential leaves.
ampicillin-selectable marker and a ColE1 replication origin. The optimized Partial least-squares regression was applied using raw cytokine concen-
sgRNA sequences described previously63 were used. Sequences for the sgRNAs trations (pg/µg total protein) regressed against a vector of total tumor mass
(Supplementary Table 1) were designed using tools provided by the Zhang Lab using PLS_Toolbox R7 (Eigenvector Research). Orthogonalization was used
at the Broad Institute (available at http://crispr.mit.edu/). B16-GFP-Cas9 cells to maximize tumor mass variance captured in the first latent variable; model
were cotransfected with two sgRNA expression vectors targeting Trp2 and a performance was assessed by random subsets cross-validation with maximum
plasmid expressing EGFP and a puromycin selectable marker from a CAG pro- splits and iterations performed.
moter using Xfect (Clontech) according to manufacturer’s instructions. Stable
cells were selected for using 2 µg/ml puromycin followed by single-cell FACS. Statistical analysis. Statistical methods were not used to predetermine
Clones were grown for 3 weeks before being analyzed for knockout by PCR necessary sample size, but sample sizes were chosen based on estimates from
amplification of the Trp2 gene (primers shown in Supplementary Table 2). pilot experiments and previously published results such that appropriate sta-
The final clone was then selected and designated B16-Trp2-KO. PCR amplifi- tistical tests could yield significant results. Experiments were not performed
cation of genomic DNA showed a smaller fragment (Supplementary Fig. 8), in a blinded fashion. Intratumoral infiltrates, serum transfer foci quantitation,

doi:10.1038/nm.4200 nature medicine


tetramer staining, AST levels, and ALT levels were analyzed by using one-way 57. Guevara-Patiño, J.A. et al. Optimization of a self-antigen for presentation of multiple
ANOVA with a Bonferroni post hoc test using GraphPad Prism software. Where epitopes in cancer immunity. J. Clin. Invest. 116, 1382–1390 (2006).
58. van Stipdonk, M.J.B. et al. Design of agonistic altered peptides for the robust
ANOVA was used, variance between groups was found to be similar by Bartlett’s
induction of CTL directed towards H-2Db in complex with the melanoma-associated
test. Survival curves were analyzed by using the log-rank (Mantel–Cox) test, and epitope gp100. Cancer Res. 69, 7784–7792 (2009).
a two-tailed Student’s t-test was used for the antigen spreading ELISPOT. For the 59. Malakhov, M.P. et al. SUMO fusions and SUMO-specific protease for efficient
tumor-infiltrate data and the DC-uptake data, Welch’s t-test (with Bonferroni expression and purification of proteins. J. Struct. Funct. Genomics 5, 75–86
(2004).
correction) was used for each condition versus untreated due to unequal
60. Rubinstein, M.P. et al. Converting IL-15 to a superagonist by binding to soluble
variance between groups. No samples were excluded from analysis. IL-15Rα. Proc. Natl. Acad. Sci. USA 103, 9166–9171 (2006).
61. Hooijkaas, A.I., Gadiot, J., van der Valk, M., Mooi, W.J. & Blank, C.U. Targeting
BRAFV600E in an inducible murine model of melanoma. Am. J. Pathol. 181,
785–794 (2012).
55. Tzeng, A., Kwan, B.H., Opel, C.F., Navaratna, T. & Wittrup, K.D. Antigen specificity 62. Muzumdar, M.D., Tasic, B., Miyamichi, K., Li, L. & Luo, L. A global double-
can be irrelevant to immunocytokine efficacy and biodistribution. Proc. Natl. Acad. fluorescent Cre reporter mouse. Genesis 45, 593–605 (2007).
Sci. USA 112, 3320–3325 (2015). 63. Chen, B. et al. Dynamic imaging of genomic loci in living human cells by an
56. Boggio, K. et al. Interleukin-12-mediated prevention of spontaneous mammary optimized CRISPR–Cas system. Cell 155, 1479–1491 (2013).
adenocarcinomas in two lines of Her-2/neu transgenic mice. J. Exp. Med. 188, 64. Joshi, N.S. et al. Regulatory T cells in tumor-associated tertiary lymphoid structures
589–596 (1998). suppress antitumor T cell responses. Immunity 43, 579–590 (2015).
© 2016 Nature America, Inc., part of Springer Nature. All rights reserved.

nature medicine doi:10.1038/nm.4200

Anda mungkin juga menyukai