+ All Categories
Home > Documents > IMMUNOTHERAPY Enhanced CAR T cell activityagainst solid … · Ma et al., Science 365, 162–168...

IMMUNOTHERAPY Enhanced CAR T cell activityagainst solid … · Ma et al., Science 365, 162–168...

Date post: 10-Jul-2020
Category:
Upload: others
View: 0 times
Download: 0 times
Share this document with a friend
8
IMMUNOTHERAPY Enhanced CAR T cell activity against solid tumors by vaccine boosting through the chimeric receptor Leyuan Ma 1,2 , Tanmay Dichwalkar 1 , Jason Y. H. Chang 1 , Benjamin Cossette 1 , Daniel Garafola 1 , Angela Q. Zhang 1 , Michael Fichter 1 , Chensu Wang 1 , Simon Liang 1 , Murillo Silva 1 , Sudha Kumari 1 , Naveen K. Mehta 1,3 , Wuhbet Abraham 1 , Nikki Thai 1 , Na Li 1 , K. Dane Wittrup 1,3,4 , Darrell J. Irvine 1,2,3,5,6 * Chimeric antigen receptorTcell (CAR-T) therapy has been effective in the treatment of hematologic malignancies, but it has shown limited efficacy against solid tumors. Here we demonstrate an approach to enhancing CAR-T function in solid tumors by directly vaccine-boosting donor cells through their chimeric receptor in vivo. We designed amphiphile CAR-T ligands (amph-ligands) that, upon injection, trafficked to lymph nodes and decorated the surfaces of antigen-presenting cells, thereby priming CAR-Ts in the native lymph node microenvironment. Amph-ligand boosting triggered massive CAR-T expansion, increased donor cell polyfunctionality, and enhanced antitumor efficacy in multiple immunocompetent mouse tumor models. We demonstrate two approaches to generalizing this strategy to any chimeric antigen receptor, enabling this simple nonhuman leukocyte antigenrestricted approach to enhanced CAR-T functionality to be applied to existing CAR-Tdesigns. C himeric antigen receptorT cell (CAR-T) immunotherapy targeting the CD19 anti- gen has produced some marked clinical responses in patients with leukemia and lymphoma, including a high proportion of durable complete remissions (1, 2). However, poor functional persistence of CAR-Ts in some patients results in disease progression (3). De- spite the success of CAR-T therapy in hemato- logical cancers, it has to date been much less effective for solid tumors, and strategies to en- hance efficacy in this setting remain an impor- tant goal (4, 5). Therapeutic vaccination is one well-established approach to enhance endoge- nous T cell responses against cancer (6). Several groups have demonstrated the concept of pre- paring CAR-Ts from virus-specific endogenous lymphocytes or introducing a CAR together with a second antigen receptor specific for a target peptide and then vaccinating recipients against the viral or secondary antigen to boost CAR-T therapy (79). However, these approaches suf- fer from being human leukocyte antigen (HLA) restricted, and the use of endogenous T cell receptors (TCRs) may be superseded by recent advances where CARs genetically targeted to the native TCR locus (thereby deleting the native TCR) have significantly enhanced acti- vity (10). We recently developed a strategy to target vac- cines to lymph nodes by linking peptide antigens to albumin-binding phospholipid polymers (11). Small peptides are normally rapidly dispersed into the blood after parenteral injection, but binding of amphiphile peptides to endogenous albumin, which constitutively traffics from blood to lymph, retargets these molecules to lymph nodes (LNs). In addition to exhibiting efficient lymph trafficking, these lipid-tailed molecules can also insert into cell membranes (12). We therefore hypothesized that by attaching a small molecule, peptide, or protein ligand for a CAR to the same polymer-lipid tail, CAR ligands could be delivered by albumin to LNs and subsequently partition into membranes of resident antigen- presenting cells (APCs), thereby codisplaying the amphiphile ligand (amph-ligand) from the APC surface together with native cytokine-receptor costimulation (Fig. 1A). Here we show how the dual properties of amph-ligands, lymph node targeting and membrane insertion, combine to create a booster vaccine for CAR-Ts. This amph- ligand strategy safely expands CAR-Ts in vivo, while increasing their functionality and enhanc- ing antitumor activity in multiple models of solid tumors. To test the ability of amph-ligands to func- tionally decorate APCs in vivo, we first employed a recently described retargetableCAR recogniz- ing the small molecule fluorescein isothiocyanate (FITC), which is directed against tumors by co- administration of a FITC-conjugated antitumor antibody (13). The anti-FITC scFv 4m5.3 peptide (14) was fused to the CD8a transmembrane do- main followed by CD28 and CD3z intracellular domains; the cognate amph-ligand for this mu- rine CAR is FITC-poly(ethylene glycol) (PEG)1,2-distearoyl-sn-glycero-3-phosphoethanolamine (amph-FITC; Fig. 1B). When incubated with model APCs in vitro, amph-FITC was absorbed into the plasma membrane in a dose-dependent manner, and despite ongoing endocytosis, many mole- cules remained accessible to surface staining with an anti-FITC antibody (Fig. 1, C and D). Amph-FITCcoated cells stimulated FITCCAR- Ts in a dose-dependent manner and were killed by FITCCAR-Ts (Fig. 1, E and F). On the basis of these findings, we next tested whether amph-FITC molecules could decorate APCs in LNs to prime FITC-CAR-Ts in vivo. Sub- cutaneous (s.c.) immunization of mice with free FITC did not result in accumulation in the drain- ing LNs, whereas 10 nmol of amph-FITC was detectable for 21 days (fig. S1A). Amph-FITC primarily accumulated in draining LNs, with low to negligible levels detectable in the liver, spleen, and other organs (fig. S1B). Confocal imaging of LNs showed that amph-FITC ini- tially accumulated in interfollicular regions but partitioned onto CD11c + dendritic cells (DCs) in T cell areas over time (Fig. 2, A and B, and fig. S1C). Surface-displayed FITC could be detected on sorted FITC + CD11c + cells stained with an anti- body against FITC (Fig. 2C and fig. S1D). In con- trast to the efficient amph-FITC insertion into the membranes of many LN cell types in vitro, surface-accessible FITC was present primarily on macrophages and CD11c + CD11b + DCs in vivo (Fig. 2D and fig. S2, A and C). DCs line colla- gen conduits that carry lymph fluid into the LN, and we hypothesize that the anatomic struc- ture of LNs in part dictates preferential access of these cells to amph-vax molecules entering LNs (15). This is supported by the observation that amph-FITC coinjected with a low-molecular- weight dextran [which is known to be trans- ported through the LN conduit system (16)] showed substantial colocalization in fiber-like structures extending from the sinuses (fig. S2D). Immunization using amph-FITC together with the STING agonist adjuvant cyclic-di-GMP increased the duration of amph-FITC display on multiple APCs and, as expected, led to up-regulation of costimulatory molecules on amph-FITC + DCs (Fig. 2E and fig. S2E). Notably, however, surface- accessible FITC decayed quickly and persisted on only a small fraction of cells. To test the ability of amph-ligand immuniza- tion to expand CAR-Ts in vivo, we transferred CD45.1 + FITC-CAR-Ts into lymphocyte (lympho)- depleted congenic CD45.2 + recipient mice and subsequently vaccinated twice with amph-FITC and adjuvant. The CAR-Ts expanded substan- tially after amph-FITC vaccination, and expan- sion was increased by coadministering adjuvant (Fig. 2F). For example, transfer of 5 × 10 4 FITC- CAR-T followed by amph-FITC vaccination with adjuvant expanded these cells to a peak of ~70% of the total CD8 + T cell compartment, yielding a CAR-T population nearly double the size achieved by administering a 200-fold-greater RESEARCH Ma et al., Science 365, 162168 (2019) 12 July 2019 1 of 7 1 David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA. 2 Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA. 3 Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA. 4 Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA. 5 Department of Materials Science and Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA. 6 Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, Cambridge, MA 02139, USA. *Corresponding author. Email: [email protected] on August 15, 2020 http://science.sciencemag.org/ Downloaded from
Transcript
Page 1: IMMUNOTHERAPY Enhanced CAR T cell activityagainst solid … · Ma et al., Science 365, 162–168 (2019) 12 July 2019 3of7 Day: -2 -1 0 7 LD CAR-T Vax Vax Day 7 nucleus mergedamph-FITC

IMMUNOTHERAPY

Enhanced CAR–T cell activity againstsolid tumors by vaccine boostingthrough the chimeric receptorLeyuan Ma1,2, Tanmay Dichwalkar1, Jason Y. H. Chang1, Benjamin Cossette1,Daniel Garafola1, Angela Q. Zhang1, Michael Fichter1, Chensu Wang1, Simon Liang1,Murillo Silva1, Sudha Kumari1, Naveen K. Mehta1,3, Wuhbet Abraham1,Nikki Thai1, Na Li1, K. Dane Wittrup1,3,4, Darrell J. Irvine1,2,3,5,6*

Chimeric antigen receptor–Tcell (CAR-T) therapy has been effective in the treatment ofhematologic malignancies, but it has shown limited efficacy against solid tumors. Herewe demonstrate an approach to enhancing CAR-T function in solid tumors by directlyvaccine-boosting donor cells through their chimeric receptor in vivo.We designedamphiphile CAR-T ligands (amph-ligands) that, upon injection, trafficked to lymph nodesand decorated the surfaces of antigen-presenting cells, thereby priming CAR-Ts inthe native lymph node microenvironment. Amph-ligand boosting triggered massiveCAR-Texpansion, increased donor cell polyfunctionality, and enhanced antitumor efficacyin multiple immunocompetent mouse tumor models.We demonstrate two approachesto generalizing this strategy to any chimeric antigen receptor, enabling this simplenon–human leukocyte antigen–restricted approach to enhanced CAR-T functionality tobe applied to existing CAR-Tdesigns.

Chimeric antigen receptor–T cell (CAR-T)immunotherapy targeting the CD19 anti-gen has produced some marked clinicalresponses in patients with leukemia andlymphoma, including a high proportion of

durable complete remissions (1, 2). However,poor functional persistence of CAR-Ts in somepatients results in disease progression (3). De-spite the success of CAR-T therapy in hemato-logical cancers, it has to date been much lesseffective for solid tumors, and strategies to en-hance efficacy in this setting remain an impor-tant goal (4, 5). Therapeutic vaccination is onewell-established approach to enhance endoge-nous T cell responses against cancer (6). Severalgroups have demonstrated the concept of pre-paring CAR-Ts from virus-specific endogenouslymphocytes or introducing a CAR together witha second antigen receptor specific for a targetpeptide and then vaccinating recipients againstthe viral or secondary antigen to boost CAR-Ttherapy (7–9). However, these approaches suf-fer from being human leukocyte antigen (HLA)restricted, and the use of endogenous T cellreceptors (TCRs) may be superseded by recentadvances where CARs genetically targeted to

the native TCR locus (thereby deleting thenative TCR) have significantly enhanced acti-vity (10).We recently developed a strategy to target vac-

cines to lymph nodes by linking peptide antigensto albumin-binding phospholipid polymers (11).Small peptides are normally rapidly dispersedinto the blood after parenteral injection, butbinding of amphiphile peptides to endogenousalbumin, which constitutively traffics from bloodto lymph, retargets these molecules to lymphnodes (LNs). In addition to exhibiting efficientlymph trafficking, these lipid-tailed moleculescan also insert into cell membranes (12). Wetherefore hypothesized that by attaching a smallmolecule, peptide, or protein ligand for a CAR tothe same polymer-lipid tail, CAR ligands couldbe delivered by albumin to LNs and subsequentlypartition into membranes of resident antigen-presenting cells (APCs), thereby codisplaying theamphiphile ligand (amph-ligand) from the APCsurface together with native cytokine-receptorcostimulation (Fig. 1A). Here we show how thedual properties of amph-ligands, lymph nodetargeting and membrane insertion, combine tocreate a booster vaccine for CAR-Ts. This amph-ligand strategy safely expands CAR-Ts in vivo,while increasing their functionality and enhanc-ing antitumor activity inmultiplemodels of solidtumors.To test the ability of amph-ligands to func-

tionally decorate APCs in vivo, we first employeda recently described “retargetable”CAR recogniz-ing the smallmolecule fluorescein isothiocyanate(FITC), which is directed against tumors by co-administration of a FITC-conjugated antitumorantibody (13). The anti-FITC scFv 4m5.3 peptide(14) was fused to the CD8a transmembrane do-

main followed by CD28 and CD3z intracellulardomains; the cognate amph-ligand for this mu-rine CAR is FITC-poly(ethylene glycol) (PEG)–1,2-distearoyl-sn-glycero-3-phosphoethanolamine(amph-FITC; Fig. 1B).When incubatedwithmodelAPCs in vitro, amph-FITC was absorbed into theplasmamembrane in a dose-dependent manner,and despite ongoing endocytosis, many mole-cules remained accessible to surface stainingwith an anti-FITC antibody (Fig. 1, C and D).Amph-FITC–coated cells stimulated FITC–CAR-Ts in a dose-dependent manner and were killedby FITC–CAR-Ts (Fig. 1, E and F).On the basis of these findings, we next tested

whether amph-FITC molecules could decorateAPCs in LNs to prime FITC-CAR-Ts in vivo. Sub-cutaneous (s.c.) immunization of mice with freeFITC did not result in accumulation in the drain-ing LNs, whereas 10 nmol of amph-FITC wasdetectable for 21 days (fig. S1A). Amph-FITCprimarily accumulated in draining LNs, withlow to negligible levels detectable in the liver,spleen, and other organs (fig. S1B). Confocalimaging of LNs showed that amph-FITC ini-tially accumulated in interfollicular regions butpartitioned onto CD11c+ dendritic cells (DCs) inT cell areas over time (Fig. 2, A and B, and fig.S1C). Surface-displayed FITC could be detectedon sorted FITC+ CD11c+ cells stained with an anti-body against FITC (Fig. 2C and fig. S1D). In con-trast to the efficient amph-FITC insertion intothe membranes of many LN cell types in vitro,surface-accessible FITC was present primarilyonmacrophages and CD11c+ CD11b+ DCs in vivo(Fig. 2D and fig. S2, A and C). DCs line colla-gen conduits that carry lymph fluid into theLN, and we hypothesize that the anatomic struc-ture of LNs in part dictates preferential accessof these cells to amph-vax molecules enteringLNs (15). This is supported by the observationthat amph-FITC coinjectedwith a low-molecular-weight dextran [which is known to be trans-ported through the LN conduit system (16)]showed substantial colocalization in fiber-likestructures extending from the sinuses (fig. S2D).Immunizationusing amph-FITC togetherwith theSTING agonist adjuvant cyclic-di-GMP increasedthe duration of amph-FITC display on multipleAPCs and, as expected, led to up-regulation ofcostimulatory molecules on amph-FITC+ DCs(Fig. 2E and fig. S2E). Notably, however, surface-accessible FITC decayed quickly and persisted ononly a small fraction of cells.To test the ability of amph-ligand immuniza-

tion to expand CAR-Ts in vivo, we transferredCD45.1+ FITC-CAR-Ts into lymphocyte (lympho)-depleted congenic CD45.2+ recipient mice andsubsequently vaccinated twice with amph-FITCand adjuvant. The CAR-Ts expanded substan-tially after amph-FITC vaccination, and expan-sion was increased by coadministering adjuvant(Fig. 2F). For example, transfer of 5 × 104 FITC-CAR-T followed by amph-FITC vaccination withadjuvant expanded these cells to a peak of~70% of the total CD8+ T cell compartment,yielding a CAR-T population nearly double thesize achieved by administering a 200-fold-greater

RESEARCH

Ma et al., Science 365, 162–168 (2019) 12 July 2019 1 of 7

1David H. Koch Institute for Integrative Cancer Research,Massachusetts Institute of Technology, Cambridge, MA02139, USA. 2Howard Hughes Medical Institute, ChevyChase, MD 20815, USA. 3Department of BiologicalEngineering, Massachusetts Institute of Technology,Cambridge, MA 02139, USA. 4Department of ChemicalEngineering, Massachusetts Institute of Technology,Cambridge, MA 02139, USA. 5Department of MaterialsScience and Engineering, Massachusetts Institute ofTechnology, Cambridge, MA 02139, USA. 6Ragon Institute ofMassachusetts General Hospital, Massachusetts Institute ofTechnology, Cambridge, MA 02139, USA.*Corresponding author. Email: [email protected]

on August 15, 2020

http://science.sciencem

ag.org/D

ownloaded from

Page 2: IMMUNOTHERAPY Enhanced CAR T cell activityagainst solid … · Ma et al., Science 365, 162–168 (2019) 12 July 2019 3of7 Day: -2 -1 0 7 LD CAR-T Vax Vax Day 7 nucleus mergedamph-FITC

number of CAR-Tswithout vaccination (Fig. 2F).By 3 weeks after boost, the persisting CAR-Tswere a mixture of effector/effector memory andcentral memory cells (Fig. 2G). Amph-vax boost-ing also expandedCAR-Ts in lympho-repletemice;

in this setting, two immunizations could expand106 transferred cells from undetectable levels to~20% of the total CD8 compartment (Fig. 2H).To determine whether professional APCs playedan important role in CAR-T priming by amph-

ligand immunization, we depleted different celltypes in LNs. CAR-T expansion in response toamph-FITC immunization was not impairedin Batf3−/− mice lacking cross-presenting DCs,but depletion of total DCs in CD11c-diphtheria

Ma et al., Science 365, 162–168 (2019) 12 July 2019 2 of 7

Fig. 1. Design of anamph-ligand vaccine toboost CAR-Ts. (A) Sche-matic of the generalchemical structure ofamph-ligands (top) andthe steps in amph-ligandvaccine boosting in vivo(bottom). Upon injection,amph-ligands associatewith albumin at theinjection site and aresubsequently trafficked tothe draining LNs.Theamphiphiles then transferto themembrane of lymphnode–resident cells,including APCs. CAR-Tsthat encounter decoratedAPCs in the LNs arestimulated by the surface-displayed amph-ligandas well as costimulatoryreceptors and cytokinesproduced by the APCs.(B) Structures of amph-FITC and cognate FITC-CAR and a representativeflow cytometry analysisof Tcell surface expressionfor FITC-CAR. (C andD) Flow cytometry analysisat 24 hours (C) andconfocal imaging after30 min (D) of amph-FITCinsertion into DC2.4 cellmembranes, by directFITC fluorescence orstaining with an anti-FITCantibody. (E and F) IFN-gsecretion (in picogramspermilliliter) (E) and killing(the percentage of targetcell death) (F) of amph-FITC–coated DC2.4 cellsafter 6 hours coculturewith FITC–CAR-Tor con-trol untransduced Tcellsat a 10:1 effector:target(E:T) ratio. Shown in (E)and (F) are representativeexperiments with techni-cal triplicates. P valueswere determined byunpaired Student’s t test.Error bars represent95% confidence intervals(CI). ***P < 0.0001;**P < 0.01; *P < 0.05.

albumin binding/membrane-inserting domain PEG2000

(i) peptide

CAR ligand

(ii) small molecule

(iii) protein domain

lymph node

cytokines

H

O

O

O

O

P

O-

O

OO

O

(OCH2CH2)45NH

NH

O

NH4+

R

CAR-T

DC

AP

C

cell

surf

ace

CAR signaling

CA

R-T

cell

surf

ace

co-stimulatorymolecule 1

co-stimulatorymolecule 2

CAR-T activationTransfer to APC cell surfaceAlbumin binding and trafficking to LN

amph-ligand

amph-ligand

P

O-

O

OO

(OCH2CH2)45NH

NH4+

OH

O

O

O

OO

OH

O

ONH

NH4+

O- O

Myc tag4m5.3scFv

CD28

CD3ζ

UntransducedFITC-CAR

100

80

60

40

20

0

Nor

mal

ized

to m

ode

CAR expression

FITC

amph-FITC (nM)

anti-FITC

FBS

10%

1%

0%

amph-ligand:

cognate CAR:

500100

25

0

500100

25500100

25

A

B

FC

Dnucleus amph-FITC anti-FITC merged

E

albumin

10 µm

100nM amph-FITC

0 25 100 5000

1000

2000

3000

4000

amph-FITC (nM)

IFN

sec

retio

n (p

g/m

l)

******

****

**

DC2.4

amph

-FIT

C DC2.

4

DC2.4

amph

-FIT

C DC2.

40

10

20

30

40

50

60

Cel

l dea

th %

******

***FITC-CAR T cellsUntransduced T cells

RESEARCH | REPORTon A

ugust 15, 2020

http://science.sciencemag.org/

Dow

nloaded from

Page 3: IMMUNOTHERAPY Enhanced CAR T cell activityagainst solid … · Ma et al., Science 365, 162–168 (2019) 12 July 2019 3of7 Day: -2 -1 0 7 LD CAR-T Vax Vax Day 7 nucleus mergedamph-FITC

Ma et al., Science 365, 162–168 (2019) 12 July 2019 3 of 7

Day: -2 -1 0 7

LD CAR-T Vax Vax

Day 7

amph-FITC anti-FITCnucleus merged

10 µm

B220CD3 anti-FITC

Day 1

Day 7

500 µm 100 µm 25 µm

CD3/anti-FITC CD11c/anti-FITC

Day 3

Day 1

Control

Medullary MΦ CD11c+CD8+ DCs CD11c+CD11b+ DCs

Surface FITC

Day 1 Day 3amph-FITC amph-FITC + adjuvant

Day: -1 0 7

CAR-T Vax Vax

10x106 CAR-T5x104 CAR-T5x104 CAR-T (amph-FITC)5x104 CAR-T (amph-FITC + adjuvant)1x104 CAR-T

1x104 CAR-T (amph-FITC)1x104 CAR-T (amph-FITC + adjuvant)

******

***

*** ******

A B

C

D

E

F

G

H

0 7 14 21 280

20

40

60

80

Days post vaccination

% o

f CD

3+C

D8+

T c

ells

in

per

iphe

ral b

lood

0 7 14 21 280

5

10

15

20

25

Days post vaccination

% o

f CD

3+C

D8+

T c

ells

in

per

iphe

ral b

lood

10x106 CAR-T1x106 CAR-T1x106 CAR-T (amph-FITC)

1x106 CAR-T (amph-FITC + adjuvant)5x104 CAR-T

5x104 CAR-T (amph-FITC)5x104 CAR-T (amph-FITC + adjuvant)

n.s

******

p=0.12 *

***

n.s

Effecto

r

Effecto

r Memory

Central m

emory0

10

20

30

40

% o

f CA

R-T

cel

ls **

Med

ullar

y M

CD11c C

D8 D

Cs

+

+

CD11c+ CD11

b DCs

+ T ce

lls

B cells

0

10

20

30

40

50

% o

f cel

ls w

ith s

urfa

ce F

ITC

***

n.s n.s

***

n.s

Med

ullar

y M

CD11c+ CD8

+ DCs

CD11c+ CD11

b+ DCs

T cells

B cells

0

2

4

6

8

% o

f cel

ls w

ith s

urfa

ce F

ITC

***

n.s n.s

***

n.s

+ IL7R-)- KLRG1- IL7R+)

amph-FITCamph-FITC + adjuvant

Effector (CD62L- KLRG1Effector Memory (CD62LCentral memory (CD62L+ KLRG1- IL7R+)

Fig. 2. Amph-ligands accumulate on LN APCs and primeCAR-Ts in vivo. (A to E) C57BL/6 mice [n = 3 animalsper group for (A) to (C) or n = 5 animals per group for (D)and (E)] were immunized s.c. with amph-FITC and cyclicdi-GMP adjuvant [(A) to (C) and (E)] or amph-FITC alone[(D) and (E)]. Shown are histological images of LNs[(A) and (B)], confocal imaging of sorted amph-FITC–coated CD11c+ cells isolated from LNs at 24 hours (C),and flow cytometry analysis of the cellular biodistributionof amph-FITC 1 or 3 days after injection [(D) and (E)].mf, macrophages. (F to H) CD45.2+ C57BL/6 mice(n = 7 animals per group) with [(F) and (G)] or without (H)prior lympho-depletion (LD) were adoptively transferredwith CD45.1+ FITC–CAR-Ts and then vaccinated withamph-FITC. Shown are frequencies of peripheral bloodCAR-Ts [(F) and (H)] and cellular phenotypes at day 30(G). P values were determined by unpaired Student’st test [(E) and (G)] and by an RM (repeated measures)two-way analysis of variance (ANOVA) with Tukey’smultiple-comparisons test [(F) and (H)]. Error barsrepresent 95% CI. ***P < 0.001; **P < 0.01; *P < 0.05;n.s., not significant.

RESEARCH | REPORTon A

ugust 15, 2020

http://science.sciencemag.org/

Dow

nloaded from

Page 4: IMMUNOTHERAPY Enhanced CAR T cell activityagainst solid … · Ma et al., Science 365, 162–168 (2019) 12 July 2019 3of7 Day: -2 -1 0 7 LD CAR-T Vax Vax Day 7 nucleus mergedamph-FITC

toxin receptor (DTR)mice or macrophages usingchlodronate liposomes led to significant reduc-tions in CAR–T cell numbers (fig. S3, A to C). Inaddition, the cytokine functionality of respond-ing CAR-Ts was reduced in all three settings (fig.S3, A to C). In vivo blockade of a collection of co-stimulatory molecules expressed by APCs alsomarkedly suppressed both FITC–CAR-T expan-sion and cytokine functionality in response toamph-FITC immunization (fig. S3D).A key concern with amph-ligand delivery is

the potential for toxicity from CAR-T–mediatedkilling of decorated cells in LNs or other tissues.Consistent with the low fraction of any cell typewith detectable surface FITC ligand, no signifi-cant changes in viable LN cell populations weredetectable 1 day, 3 days, or 14 days after amph-FITC immunization (fig. S4, A to C). No changesin systemic liver enzymes, liver histopathology orCAR-T infiltration, or serum cytokine levels wereobserved after amph-FITC boosting (fig. S4, D toI). We further evaluated the functional integrityof vaccinated LNs by administering an amph-FITC boost in the presence or absence of trans-ferredFITC-CAR-Ts and then immunizing animalswith ovalbumin at the same site 5, 7, or 14 dayslater (fig. S4J). We observed decreased expan-sion and functionality of endogenous SIINFEKL-specific T cells when animals were immunized5 days—but not 7 or 14 days—after amph-FITCboost, suggesting that the combination of CAR-Ttransfer and amph-FITC vaccination has a short-term effect on priming of endogenous T cellresponses [which recovers rapidly (fig. S4K)].Owing to the lack of T cell help, repeated amph-FITC immunization with adjuvant elicited noantibody response against the amph-ligand it-self (fig. S5).We next evaluated if amph-ligands could be

used to prime a bona fide tumor antigen–specificCAR. The EGFRvIII-specific 139scFv CAR recog-nizes a short linear epitopederived fromEGFRvIII(17). We prepared murine T cells expressing thisCAR and synthesized an amph-vax moleculecomposed of PEG-DSPE linked to the peptideligand with or without an N-terminal FITC label(amph-pepvIII; Fig. 3A). Similar to amph-FITC,amph-pepvIII inserted in cellmembranes in vitroand the amph-pepvIII–coated cells stimulatedEGFRvIII-CAR-Ts (fig. S6, A andB). Immunizationof mice with amph-pepvIII triggered EGFRvIII-CAR-T proliferation in vivo (Fig. 3B). To test thetherapeutic impact of vaccine boosting, we trans-ducedmurine CT-2A glioma cells with EGFRvIII;these cells were efficiently killed by EGFRvIII-CAR-Ts in vitro (fig. S6, C and D). Transfer ofEGFRvIII–CAR-T into lympho-depleted CT-2A-mEGFRvIII tumor-bearing mice that were thenimmunized with amph-pepvIII expanded theCAR-Ts substantially in the periphery (Fig. 3C).Vaccination induced significant increases inthe proportion of cells with an effector phenotype(fig. S6E) and 5- to 10-fold increases in CAR–T cell polyfunctionality (Fig. 3D). Amph-vax boost-ing greatly increasedCAR-T infiltration into tumors,and these tumor-infiltrating lymphocytes expressedhigher levels of granzyme B and Ki67 than un-

boosted CAR-Ts (Fig. 3E). In therapeutic studies,animals receiving both CAR-T and repeatedamph-vax boosting had significantly delayedtumor growth and prolonged survival (Fig. 3, FandG). Treatmentwith 1 × 106 CAR-Ts alone ledto no long-term survivors, while this same CAR-Tdose boosted by amph-vaccination eliminatedtumors in amajority of animals (Fig. 3, F andG).Administration of amph-pepvIII with adjuvantin the absence of CAR-Ts had no therapeuticimpact (fig. S6F). EGFRvIII–CAR-Ts from vacci-nated animals persisted over time, and surviv-ing animals rejected tumor rechallenge at day75 (fig. S6, G and H). Notably, animals that re-jected primary tumors after CAR-T plus amph-vax boosting therapy also rejected rechallengewith parental CT-2A tumor cells lacking the lig-and for the CAR-Ts , suggesting induction of anendogenous T cell response against other tumorantigens (fig. S6I). Motivated by this finding, weevaluated the reactivity of splenocytes from CT-2A-mEGFvIII tumor-bearing mice that receivedCAR-Tswith orwithout two amph-pepvIII boosts.Enzyme-linked immunosorbent spot (ELISPOT)analysis of interferon-g (IFN-g) production bysplenocytes cultured with parental CT-2A cellsrevealed a strong endogenous T cell responseagainst parental tumors (Fig. 3H). Similar toamph-FITC–vaccinated mice, no antibody re-sponse was elicited against pepvIII after threerounds of weekly vaccination (fig. S6J). We alsoevaluated the therapeutic efficacy of CAR-T plusamph vaccination in tumor-bearing mice with-out lympho-depletion preconditioning. Tumorprogression in animals receiving CAR-T alonewas indistinguishable from that in animals re-ceiving control untransduced T cells, whereasCAR-T transfer combined with amph-pepvIIIimmunization delayed tumor growth and pro-longed animal survival (Fig. 3, I and J). In boththe lympho-depleted and non–lympho-depletedsettings, amph-vax boosting was accompaniedby small transient alterations in animal bodyweight and minimal alterations in serum cyto-kine levels (fig. S6, K and L). To assess the util-ity of amph-vax boosting with a more potent“third-generation”CAR design, we generated anEGFRvIII-targeting CAR containing both CD28and 41BB co-stimulatory domains. This CARwaswell-expressed and functional in vitro (fig. S7, Aand C). We then treated large (~50-mm2) estab-lished CT-2A-mEGFRvIII tumorswith EGFRvIII-28BBzCAR-T cells, with or without amph-pepvIIIboosting. In this high tumor burden setting, theCAR-Ts alone had a modest impact on tumorprogression, and amph-ligand boosting greatlyimproved tumor control and enhanced overallsurvival (fig. S7, D and E).Although use of a peptide ligand for CAR-Ts

waseffective, someCARsrecognize three-dimensionalstructural epitopes (18). As an alternative strategyto amph-ligand boost with any CAR regardlessof the nature of its binding domain or specific-ity, we devised a tandem scFv-based bispecificCAR based on recently reported designs (19).The anti-FITC scFv was fused to the N-terminalextracellular domain of a tumor-targeting CAR

(TA99) that recognized themelanoma-associatedantigen TRP1 (Fig. 4, A and B). FITC/TRP1-CAR-Ts were activated both by amph-FITC-coatedtarget cells and by TRP1-expressing B16F10 cells(fig. S8A), and killed TRP1+ target cells at levelsequivalent to those cells expressing mono-specific TRP1-CAR (Fig. 4C). In vivo, amph-FITC vaccination stimulated FITC/TRP1-bispecificCAR-T proliferation (fig. S8B). Similar to obser-vations in the EGFRvIII system, amph-vax boost-ing of FITC/TRP1–CAR-T in B16F10 tumor-bearinganimals led to pronounced CAR-T expansion inthe periphery and increased tumor infiltration(fig. S8, C and D), withminimal serum cytokineelevation and transient fluctuations in bodyweight after each vaccination (fig. S8, E and F).Whereas adoptive therapy with FITC/TRP1–CAR-T alone had almost no effect on B16F10 tumorprogression, repeated boosting after transfer withamph-FITC led to pronounced slowing in tumorgrowth and extended survival (Fig. 4, D and E).One resistance mechanism to CAR-T therapy isloss of surface antigen (20), but we did not ob-serve apparent Trp1 loss upon tumor outgrowthin this model (fig. S8, G and H). To assess poten-tial autoimmune toxicity induced by amph-vaxboosting, we examined thymus and skin tis-sues (which naturally express Trp1) from treatedanimals, butwe foundno changes inhistopathologyor CAR-T infiltration into the thymus with amph-vax boosting (fig. S8, I to K). We also assessedwhether CAR-T therapy with vaccine boostingwould be more effective if mixed CD4/CD8 CAR-Tswereused. In vitro, bothCD4+andCD8+CAR-Tswere activated by culture with amph-ligand–coated target cells (fig. S8L), and similar ther-apeutic efficacywas observedwhenB16F10 tumorswere treated with CD8 as with mixed CD4/CD8FITC/Trp1–CAR-Ts boosted by amph-FITC vac-cination (fig. S8, M and N).To assess the broad applicability of this bi-

specific CAR platform irrespective of animalstrain or haplotype and to evaluate treatmentof metastatic disease, we prepared 4T1 tumorcells transduced to express mEGFRvIII andluciferase, modeling EGFRvIII+ breast cancer(21) on the BALB/c background. A cognate FITC/EGFRvIII-bispecific CAR was generated, whichwas well-expressed in BALB/c T cells and wasfunctional in vitro and in vivo (fig. S9, A andD). 4T1-mEGFRvIII tumor cells were injectedintravenously (i.v.) into BALB/c mice to inducelung metastases and then treated with FITC/EGFRvIII–CAR-T with or without amph-FITCboosting. Tumor progression as assessed bybioluminescence imaging was significantly im-pacted only when CAR-Ts were supplementedwith amph-ligand boosting (fig. S9E), leadingto prolonged survival and clearance of tumorsin two of five animals (fig. S9F). In the CAR-Tplus amph-vax–treated animals that relapsed,EGFRvIII surface levels weremarkedly reduced,suggesting selection of low-antigen–expressingor null tumor cells during therapy (fig. S9G).Finally, to verify that this bispecific CAR ap-proach could also be used to boost human CAR-T,we constructed a FITC/hCD19-bispecific human

Ma et al., Science 365, 162–168 (2019) 12 July 2019 4 of 7

RESEARCH | REPORTon A

ugust 15, 2020

http://science.sciencemag.org/

Dow

nloaded from

Page 5: IMMUNOTHERAPY Enhanced CAR T cell activityagainst solid … · Ma et al., Science 365, 162–168 (2019) 12 July 2019 3of7 Day: -2 -1 0 7 LD CAR-T Vax Vax Day 7 nucleus mergedamph-FITC

Ma et al., Science 365, 162–168 (2019) 12 July 2019 5 of 7

Cell trace dilution

100

80

60

40

20

0

Nor

mal

ized

to m

ode

CAR+ T cellsCAR- T cells

D

E

H

J

F

I

100

80

60

40

20

0

Nor

mal

ized

to m

ode

CAR expression

amph-ligand:LEEKKGNYVVTDHC-PEG-DSPE

UntransducedEGFRvIII CAR

10x106 Control T10x106 CAR-T10 x106 CAR-T + Vax1x106 CAR-T1x106 CAR-T + Vax2.5x105 CAR-T2.5x105 CAR-T + Vax

*

**

**

p=0.16**

**

0 20 40 600

50

100

Days post tumor inoculation

Per

cent

sur

viva

l

Control T 10x106 CAR-T10x106 CAR-T + Vax

n.s

******

10x106

Day: 7 8 9 13 16 23

LD CAR-T Vax

30

Peripheral blood analysis4x106

0

tumor cells

Day: 7 8 9 16 23

LD CAR-T Vax Vax Vax

0

tumor cells4x106

Day: 8 9 16 23

CAR-T Vax Vax Vax

0

tumor cells4x106

Day: 0 4 5 6 13

tumor cellsLD

CAR-TVax TIL analysis10x106 1x106

6

61x101x10

Control T

CAR-T CAR-T + Vax

2.5x105 CAR-T2.5x105 CAR-T + Vax

***

*

Day: 0 4 5 6 20

tumor cellsLD

CAR-TVax ELISPOT

13

Vax10x106 10x106

CAR-T + Vax

CAR-T

Control T

No targetCT-2A

PBS

Vax

IFNγ

TNFα

9.1 12.1

3.090.9

6.1 69.5

0.838.9

A B C

G

0 5 10 15 20 250

10

20

30

40

50

60

Days post vaccination

% o

f CD

3+C

D8+

T c

ells

in

per

iphe

ral b

lood

Contro

l

1x10

6 CAR-T

1x10

6 CAR-T

+ V

ax

2.5x

105 C

AR-T

2.5x

105 C

AR-T +

Vax

0

20

40

60

80

100

Cyt

okin

e se

cret

ion

(% o

f CA

R-T

cel

ls)

IFN +IFN +TNF +

******

******

PBS

pepv

III V

ax0

10000

20000

30000

40000

# of

CA

R-T

cel

ls p

er m

g tu

mor **

PBS

pepv

III V

ax0

2000

4000

6000

8000

Gra

nzym

e B

MF

I

**

PBS

pepv

III V

ax6000

8000

10000

12000

Ki6

7 M

FI

*** Control T

10x106 CAR-T10x106 CAR-T + Vax

1x106 CAR-T

1x106 CAR-T + Vax

2.5x105 CAR-T

2.5x105 CAR-T + Vax ****** ***

***

n.s

p=0.088

10x106

0 7 10 20 300

50

100

150

200

250

Days post tumor inoculation

Tum

or a

rea

(mm

2)

0 10 20 30 400

50

100

150

200

Days post tumor inoculation

Tum

or a

rea

(mm

2)

Control T 10x106 CAR-T10x106 CAR-T + Vax

n.s

******

10x106

Contro

l T

CAR-T

CAR-T +

Vax

0

20

40

60

80

# of

Spo

ts

per

105

CD

3 T

cel

ls***

1500 50 1000

50

100

Days post tumor inoculation

Per

cent

sur

viva

l

Fig. 3. Amph-peptide ligands boost CAR-Ts in vivo for enhancedsolid tumor immunotherapy in mice. (A) Structure of amph-pepvIIIand surface expression of EGFRvIII CAR. (B) Representative histogramshowing EGFRvIII–CAR-Tproliferation in LNs 48 hours after amph-pepvIIIvaccination (n = 3 animals per group). (C and D) Expansion (C) andcytokine polyfunctionality at day 7 (D) of circulating EGFRvIII–CAR-Tsfollowing a single amph-pepvIII immunization (n = 5 animals per group).(E) Enumeration, granzyme B levels, and Ki67 levels of tumor-infiltratingEGFRvIII–CAR-Ts (n = 4 animals per group) with or without amph-pepvIIIboost. (F to J) Tumor growth [(F) and (I)], ELISPOT of enriched CD3+

splenocytes cultured with irradiated parental CT-2A tumor cells (H), and

survival [(G) and (J)] of mEGFRvIII-CT-2A tumor-bearing mice treatedwith EGFRvIII–CAR-T with or without amph-pepvIII vaccination foranimals that were lympho-depleted [(F) and (G) n = 5 animals per group;(H) n = 4 animals per group)], or lympho-replete [(I) and (J) n = 7animals per group)] prior to adoptive transfer. The black arrow indicatestime of CT-2A-EGFRvIII tumor rechallenge. The red arrow indicatestime of parental CT-2A tumor rechallenge. P values were determinedby unpaired Student’s t test [(D), (E), and (H)], by an RM two-wayANOVA with Tukey’s multiple-comparisons test [(C), (F), and (I)], orby log-rank test [(G) and (J)]. Error bars represent 95% CI. ***P < 0.001;**P < 0.01; *P < 0.05; n.s., not significant.

RESEARCH | REPORTon A

ugust 15, 2020

http://science.sciencemag.org/

Dow

nloaded from

Page 6: IMMUNOTHERAPY Enhanced CAR T cell activityagainst solid … · Ma et al., Science 365, 162–168 (2019) 12 July 2019 3of7 Day: -2 -1 0 7 LD CAR-T Vax Vax Day 7 nucleus mergedamph-FITC

CAR using the established FMC63 antibodyagainst CD19 (22) and expressed this CAR inhuman T cells (Fig. 4F). Human FITC/hCD19–CAR-Tswere stimulated by both CD19+ Raji cellsaswell as amph-FITC–coated target cells (Fig. 4G).Altogether, we present here a new vaccine ap-proach to boosting CAR-T numbers and func-tionality in vivo with low toxicity, enablingenhanced efficacy in syngeneic solid tumormodels. Although not directly evaluated here,this approach might be further enhanced bynascent strategies to improve CAR function,such as insertion of the CAR into the TRAClocus (10). The bispecific vaccinable CAR designwith amph-FITC vaccine offers a simple anduniversal solution to boosting CAR-Ts with anyantigen specificity.

REFERENCES AND NOTES

1. A. D. Fesnak, C. H. June, B. L. Levine, Nat. Rev. Cancer 16,566–581 (2016).

2. D. N. Khalil, E. L. Smith, R. J. Brentjens, J. D. Wolchok,Nat. Rev. Clin. Oncol. 13, 273–290 (2016).

3. S. Guedan et al., JCI Insight 3, e96976 (2018).4. K. Newick, S. O’Brien, E. Moon, S. M. Albelda, Annu. Rev. Med.

68, 139–152 (2017).5. C. H. June, R. S. O’Connor, O. U. Kawalekar, S. Ghassemi,

M. C. Milone, Science 359, 1361–1365 (2018).6. S. H. van der Burg, R. Arens, F. Ossendorp, T. van Hall,

C. J. Melief, Nat. Rev. Cancer 16, 219–233 (2016).7. M. Tanaka et al., Clin. Cancer Res. 23, 3499–3509 (2017).8. X. Wang et al., Clin. Cancer Res. 21, 2993–3002 (2015).9. C. Y. Slaney et al., Clin. Cancer Res. 23, 2478–2490 (2017).10. J. Eyquem et al., Nature 543, 113–117 (2017).11. H. Liu et al., Nature 507, 519–522 (2014).12. H. Liu, B. Kwong, D. J. Irvine, Angew. Chem. Int. Ed. Engl. 50,

7052–7055 (2011).13. J. S. Ma et al., Proc. Natl. Acad. Sci. U.S.A. 113, E450–E458

(2016).

14. E. T. Boder, K. S. Midelfort, K. D. Wittrup, Proc. Natl. Acad. Sci.U.S.A. 97, 10701–10705 (2000).

15. M. Sixt et al., Immunity 22, 19–29 (2005).16. J. E. Gretz, C. C. Norbury, A. O. Anderson, A. E. Proudfoot,

S. Shaw, J. Exp. Med. 192, 1425–1440 (2000).17. J. H. Sampson et al., Clin. Cancer Res. 20, 972–984 (2014).18. S. N. De Oliveira et al., J. Transl. Med. 11, 23 (2013).19. E. Zah, M. Y. Lin, A. Silva-Benedict, M. C. Jensen, Y. Y. Chen,

Cancer Immunol. Res. 4, 498–508 (2016).20. E. Sotillo et al., Cancer Discov. 5, 1282–1295 (2015).21. C. A. Del Vecchio et al., Cancer Res. 72, 2657–2671 (2012).22. J. N. Kochenderfer et al., Blood 116, 4099–4102 (2010).

ACKNOWLEDGMENTSWe thank the Koch Institute Swanson Biotechnology Center fortechnical support, specifically, the whole-animal imaging corefacility, histology core facility, and flow cytometry core facility. Wethank T. Seyfried for providing the CT-2A cell line. Funding: Thiswork was supported by the NIH (award EB022433), the MarbleCenter for Nanomedicine, and Johnson & Johnson. D.J.I. is aninvestigator of the Howard Hughes Medical Institute. The project

Ma et al., Science 365, 162–168 (2019) 12 July 2019 6 of 7

4m5.3 scFv

CD28

CD3ζ

tumor antigenspecific scFV

FITC

A

B

100

80

60

40

20

0

Nor

mal

ized

to m

ode

CAR expression

CUntransducedFITC/TRP1-CAR

Day: 4 5 6 13 20

LDCAR-T

Vax Vax Vax

D

E

100

80

60

40

20

0

Nor

mal

ized

to m

ode

CAR expression

UntransducedFITC/hCD19-CAR

F G

CAR-T

APCs Tumor cells 0

tumor cells5x105 10x106

0 10 20 30 40 500

50

100

Days post tumor inoculation

Per

cent

sur

viva

l

n.s

*****

Untra

nsdu

ced T

cells

hCD19

-CAR-T

FITC/h

CD19-C

AR-T0

100

200

300

400

IFN

sec

retio

n(pg

/ml)

Rajiamph-FITC K562

***

******

***

Contro

l T

TRP1-CAR-T

FITC/T

RP1-CAR-T

0

20

40

60

80

100

Cel

l dea

th %

******

0 5 10 15 20 250

50

100

150

200

250

Days post tumor inoculation

Tum

or a

rea

(mm

2)

Control T

FITC/TRP1-CAR-T

FITC/TRP1-CAR-T + Vax

n.s

*****

Control T

FITC/TRP1-CAR-T

FITC/TRP1-CAR-T + Vax

Fig. 4. Amph-FITC ligands boost the antitumor activity ofbispecific CAR-Ts. (A) Schematic of bispecific CAR design:the FITC-binding scFv 4m5.3 is fused through a short linker to atumor antigen–specific CAR, enabling the T cell to be triggeredby binding to either FITC-decorated cells or tumor cells.(B) Representative T cell surface expression of FITC/TRP1-CAR. (C) Killing of TRP1-expressing B16F10 cells in vitro after

6-hour coculture with FITC/TRP1–CAR-T, monospecific TRP1–CAR-T,or control untransduced T cells at an E:T of 10:1. (D and E) Tumor growth(D) and survival (E) of B16F10 tumor-bearing mice (n = 7 animals pergroup) treated with 10 × 106 CAR–Ts alone or CAR-Ts plus amph-FITCvaccination. P values were determined by an RM two-way ANOVA withTukey’s multiple-comparisons test (D) or by log-rank test (E). (F) Surface

expression of FITC/hCD19-bispecific CAR on human T cells. (G) FITC/TRP1-bispecific CAR-Ts responding to either hCD19+ Raji cells or amph-FITC–coated K562 cells as monitored by IFN-g secretion. Shown in (C) and(G) are representative experiments with technical triplicates. P values weredetermined by an unpaired Student’s t test [(C) and (G)]. Error barsrepresent 95% CI. ***P < 0.0001; **P < 0.01; *P < 0.05; n.s., not significant.

RESEARCH | REPORTon A

ugust 15, 2020

http://science.sciencemag.org/

Dow

nloaded from

Page 7: IMMUNOTHERAPY Enhanced CAR T cell activityagainst solid … · Ma et al., Science 365, 162–168 (2019) 12 July 2019 3of7 Day: -2 -1 0 7 LD CAR-T Vax Vax Day 7 nucleus mergedamph-FITC

was also supported by award no. T32GM007753 from the NationalInstitute of General Medical Sciences. M.F. was supported byDeutsche Forschungsgemeinschaft grant FI 2249/1-1:1. Thecontent is solely the responsibility of the authors and does notnecessarily represent the official views of the National Instituteof General Medical Sciences or the National Institutes of Health.Author contributions: L.M., D.J.I., and K.D.W. designed thestudies. L.M. and D.J.I. analyzed and interpreted the data andwrote the manuscript. L.M, T.D, D.G., A.Q.Z., J.Y.H.C., S.K., B.C,

C.W., S.L., M.S., M.F., N.K.M., W.A., N.T., and N.L. performedexperiments. Competing interests: D.J.I. and L.M. are inventorson international patent application PCT/US2018/051764submitted by Massachusetts Institute of Technology, which coversthe use of amphiphile-vaccine technology as a vaccine for CAR-Ts.D.J.I. is a consultant for Elicio Therapeutics that has licensedIP related to this technology. Data and materials availability:Materials are available under a material transfer agreement(contact person D.J.I.).

SUPPLEMENTARY MATERIALS

science.sciencemag.org/content/365/6449/162/suppl/DC1Materials and MethodsFigs. S1 to S9References (23–28)

28 October 2018; accepted 10 June 201910.1126/science.aav8692

Ma et al., Science 365, 162–168 (2019) 12 July 2019 7 of 7

RESEARCH | REPORTon A

ugust 15, 2020

http://science.sciencemag.org/

Dow

nloaded from

Page 8: IMMUNOTHERAPY Enhanced CAR T cell activityagainst solid … · Ma et al., Science 365, 162–168 (2019) 12 July 2019 3of7 Day: -2 -1 0 7 LD CAR-T Vax Vax Day 7 nucleus mergedamph-FITC

receptorT cell activity against solid tumors by vaccine boosting through the chimeric−Enhanced CAR

Wittrup and Darrell J. IrvineChensu Wang, Simon Liang, Murillo Silva, Sudha Kumari, Naveen K. Mehta, Wuhbet Abraham, Nikki Thai, Na Li, K. Dane Leyuan Ma, Tanmay Dichwalkar, Jason Y. H. Chang, Benjamin Cossette, Daniel Garafola, Angela Q. Zhang, Michael Fichter,

DOI: 10.1126/science.aav8692 (6449), 162-168.365Science 

, this issue p. 162; see also p. 119ScienceT cell.−and increased tumor killing. The system could potentially be applied to boost any CAR

T cell activation, expansion,−Injected ''amph-ligand'' vaccines promoted synthetic antigen presentation and led to CAR restimulating the CAR directly within the native lymph node microenvironment (see the Perspective by Singh and June).

T cells by− designed a vaccine strategy to improve the efficacy of CARet al.engineered T cells to the tumor site. Ma cancers. Yet this approach has been a challenge for solid tumors, in part because it is difficult to target functional

T cell immunotherapy has been highly successful for treating certain blood−Chimeric antigen receptor (CAR)T cells−A boost for CAR

ARTICLE TOOLS http://science.sciencemag.org/content/365/6449/162

MATERIALSSUPPLEMENTARY http://science.sciencemag.org/content/suppl/2019/07/10/365.6449.162.DC1

CONTENTRELATED

http://stm.sciencemag.org/content/scitransmed/6/261/261ra151.fullhttp://stm.sciencemag.org/content/scitransmed/10/430/eaao2731.fullhttp://stm.sciencemag.org/content/scitransmed/9/399/eaaa0984.fullhttp://stm.sciencemag.org/content/scitransmed/11/495/eaaw2293.fullhttp://science.sciencemag.org/content/sci/365/6449/119.full

REFERENCES

http://science.sciencemag.org/content/365/6449/162#BIBLThis article cites 28 articles, 13 of which you can access for free

PERMISSIONS http://www.sciencemag.org/help/reprints-and-permissions

Terms of ServiceUse of this article is subject to the

is a registered trademark of AAAS.ScienceScience, 1200 New York Avenue NW, Washington, DC 20005. The title (print ISSN 0036-8075; online ISSN 1095-9203) is published by the American Association for the Advancement ofScience

Science. No claim to original U.S. Government WorksCopyright © 2019 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of

on August 15, 2020

http://science.sciencem

ag.org/D

ownloaded from


Recommended