Top Banner
Bioengineering T cells to target carbohydrate to treat opportunistic fungal infection Pappanaicken R. Kumaresan a,1 , Pallavi R. Manuri a,1 , Nathaniel D. Albert b , Sourindra Maiti a , Harjeet Singh a , Tiejuan Mi a , Jason Roszik a , Brian Rabinovich a , Simon Olivares a , Janani Krishnamurthy a , Ling Zhang a , Amer M. Najjar c , M. Helen Huls a , Dean A. Lee a,d , Richard E. Champlin e , Dimitrios P. Kontoyiannis b , and Laurence J. N. Cooper a,d,2 a Division of Pediatrics, b Department of Infectious Diseases, c Division of Diagnostic Imaging, Department of Cancer Systems Imaging, d The University of Texas Graduate School of Biomedical Sciences, and e Stem Cell Transplantation and Cell Therapy, University of Texas MD Anderson Cancer Center, Houston, TX, 77030 Edited by Philippa Marrack, Howard Hughes Medical Institute, National Jewish Health, Denver, CO, and approved June 10, 2014 (received for review July 8, 2013) Clinical-grade T cells are genetically modified ex vivo to express chimeric antigen receptors (CARs) to redirect their specificity to target tumor-associated antigens in vivo. We now have developed this molecular strategy to render cytotoxic T cells specific for fungi. We adapted the pattern-recognition receptor Dectin-1 to activate T cells via chimeric CD28 and CD3-ζ (designated D-CAR) upon binding with carbohydrate in the cell wall of Aspergillus germ- lings. T cells genetically modified with the Sleeping Beauty system to express D-CAR stably were propagated selectively on artificial activating and propagating cells using an approach similar to that approved by the Food and Drug Administration for manufacturing CD19-specific CAR + T cells for clinical trials. The D-CAR + T cells exhibited specificity for β-glucan which led to damage and inhibi- tion of hyphal growth of Aspergillus in vitro and in vivo. Treat- ment of D-CAR + T cells with steroids did not compromise antifungal activity significantly. These data support the targeting of carbohydrate antigens by CAR + T cells and provide a clinically appealing strategy to enhance immunity for opportunistic fungal infections using T-cell gene therapy. T-cell therapy | β-1,3-glucan | fungus | adoptive immunotherapy O pportunistic invasive fungal infections (IFI) by Aspergillus spp. cause morbidity and mortality in immunocompromised patients. Mortality rates associated with invasive Aspergillus (IA) are 22% in patients receiving solid-organ transplants and 6085% in patients receiving hematopoietic stem cell transplants (HSCT) (1, 2). Antifungal agents such as polyenes, triazoles, and echinocandins can be rendered ineffective by suboptimal host immunity, emergence of drug-resistant strains, and attendant toxicities in the recipients (3, 4). Thus, new approaches for treating IAs are needed. Because the adoptive transfer of ge- netically modified T cells expressing CD19-specific chimeric antigen receptors (CARs) has resulted in successful treatment of patients with B-cell malignancies (59), we sought to determine if a CAR could be developed to redirect T-cell specificity to Aspergillus. Among immunocompetent individuals, intact innate immunity can prevent and eradicate IFI. Endogenous alveolar macro- phages recognize, phagocytize, and kill fungal spores (10), and neutrophils can recognize and eliminate germinating hyphae (11). Immunotherapeutic strategies such as adoptive transfer of preselected CD4 + T-cell clones can help control Aspergillus in- fection indirectly by producing interferon-gamma (IFN-γ) (12). An immunotherapeutic strategy deploying cytotoxic T cells appears to be clinically advantageous, because such cells have an endogenous ability to kill, be propagated to large numbers ex vivo, be genetically modified to recognize desired target anti- gens, and contribute to immunologic memory (8). We previously have redirected the specificity of genetically modified T cells via the stable introduction of a CD19-specific second-generation CAR (8, 9). Recognition of CD19 was ach- ieved by a CAR exodomain composed of a CD19-specific single- chain variable fragment attached to the T-cell surface via a modified IgG 4 hinge and fragment-crystallized (Fc) region (13). CAR-dependent and MHC-independent T-cell activation can be achieved through the CAR endodomain composed of CD3-ζ and CD28 (14, 15). We modified this prototypical CAR design to achieve recognition of carbohydrate by accommodating the pattern-recognition properties of Dectin-1 (16, 17). This dectin is a type II transmembrane protein expressed on macro- phages, neutrophils, and dendritic cells (18) and is specific for β-glucans, which are glucose polymers consisting of β-1,3-glucan and β-1,6-glucan expressed on the cell wall of fungi (19). Because Dectin-1 mediates recognition of Aspergillus fumigatus (20), we hypothesized that the extracellular portion of Dectin-1 could be adapted as the specificity domain for a CAR (designated D-CAR) on T cells to redirect their specificity for this fungus. We report that T cells can be genetically modified by using the Sleeping Beauty (SB) transposon/transposase system to express D-CAR stably and can be propagated selectively on artificial activating and propagating cells (aAPCs). We demonstrate that the D-CAR + T cells (i ) bind specifically to laminarin (which is rich in β-1,3-glucan), (ii ) exhibit a central memory phenotype, (iii ) inhibit growth of Aspergillus hyphae even in the presence of Significance Patients with compromised T-cell function are at risk for op- portunistic fungal infections. We have developed a novel ap- proach to restore immunity by using a fungal pattern-recognition receptor Dectin-1 to redirect T-cell specificity to carbohydrate antigen in the fungal cell wall. We did so by genetically modifying T cells using the nonviral Sleeping Beauty gene-transfer system to enforce expression of a chimeric antigen receptor (CAR) that recapitulates the specificity of Dectin-1 (D-CAR). The D-CAR + T cells can be electroporated and propagated on artificial activating and propagating cells in a manner suitable for human application, enabling this immunology to be translated into immunotherapy. This approach has implications for genetically modifying T cells to express CARs with specificity for carbohydrate and thus broad- ening their application in the investigational treatment of patho- gens and malignancies. Author contributions: P.R.K., P.R.M., D.P.K., and L.J.N.C. designed research; P.R.K., P.R.M., N.D.A., S.M., T.M., J.R., S.O., J.K., and L.Z. performed research; P.R.K. and P.R.M. contrib- uted new reagents/analytic tools; P.R.K., P.R.M., H.S., B.R., J.K., A.M.N., M.H.H., D.A.L., and R.E.C. analyzed data; and P.R.K., P.R.M., D.P.K., and L.J.N.C. wrote the paper. Conflict of interest statement: L.J.N.C. founded and owns InCellerate, Inc. He has patents with Sangamo BioSciences with artificial nucleases. He consults with Targazyme, Inc. (formerly American Stem Cells, Inc.), GE Healthcare, Ferring Pharmaceuticals, Inc., and Bristol-Myers Squibb. He receives honoraria from Miltenyi Biotec. This article is a PNAS Direct Submission. 1 P.R.K. and P.R.M. contributed equally to this work. 2 To whom correspondence should be addressed. E-mail: [email protected]. This article contains supporting information online at www.pnas.org/lookup/suppl/doi:10. 1073/pnas.1312789111/-/DCSupplemental. 1066010665 | PNAS | July 22, 2014 | vol. 111 | no. 29 www.pnas.org/cgi/doi/10.1073/pnas.1312789111 Downloaded by guest on November 17, 2020
6

Bioengineering T cells to target carbohydrate to treat ... · Bioengineering T cells to target carbohydrate to treat opportunistic fungal infection Pappanaicken R. Kumaresana,1, Pallavi

Aug 14, 2020

Download

Documents

dariahiddleston
Welcome message from author
This document is posted to help you gain knowledge. Please leave a comment to let me know what you think about it! Share it to your friends and learn new things together.
Transcript
Page 1: Bioengineering T cells to target carbohydrate to treat ... · Bioengineering T cells to target carbohydrate to treat opportunistic fungal infection Pappanaicken R. Kumaresana,1, Pallavi

Bioengineering T cells to target carbohydrate to treatopportunistic fungal infectionPappanaicken R. Kumaresana,1, Pallavi R. Manuria,1, Nathaniel D. Albertb, Sourindra Maitia, Harjeet Singha, Tiejuan Mia,Jason Roszika, Brian Rabinovicha, Simon Olivaresa, Janani Krishnamurthya, Ling Zhanga, Amer M. Najjarc,M. Helen Hulsa, Dean A. Leea,d, Richard E. Champline, Dimitrios P. Kontoyiannisb, and Laurence J. N. Coopera,d,2

aDivision of Pediatrics, bDepartment of Infectious Diseases, cDivision of Diagnostic Imaging, Department of Cancer Systems Imaging, dThe University of TexasGraduate School of Biomedical Sciences, and eStem Cell Transplantation and Cell Therapy, University of Texas MD Anderson Cancer Center, Houston,TX, 77030

Edited by Philippa Marrack, Howard Hughes Medical Institute, National Jewish Health, Denver, CO, and approved June 10, 2014 (received for review July8, 2013)

Clinical-grade T cells are genetically modified ex vivo to expresschimeric antigen receptors (CARs) to redirect their specificity totarget tumor-associated antigens in vivo. We now have developedthis molecular strategy to render cytotoxic T cells specific for fungi.We adapted the pattern-recognition receptor Dectin-1 to activateT cells via chimeric CD28 and CD3-ζ (designated “D-CAR”) uponbinding with carbohydrate in the cell wall of Aspergillus germ-lings. T cells genetically modified with the Sleeping Beauty systemto express D-CAR stably were propagated selectively on artificialactivating and propagating cells using an approach similar to thatapproved by the Food and Drug Administration for manufacturingCD19-specific CAR+ T cells for clinical trials. The D-CAR+ T cellsexhibited specificity for β-glucan which led to damage and inhibi-tion of hyphal growth of Aspergillus in vitro and in vivo. Treat-ment of D-CAR+ T cells with steroids did not compromiseantifungal activity significantly. These data support the targetingof carbohydrate antigens by CAR+ T cells and provide a clinicallyappealing strategy to enhance immunity for opportunistic fungalinfections using T-cell gene therapy.

T-cell therapy | β-1,3-glucan | fungus | adoptive immunotherapy

Opportunistic invasive fungal infections (IFI) by Aspergillusspp. cause morbidity and mortality in immunocompromised

patients. Mortality rates associated with invasive Aspergillus (IA)are 22% in patients receiving solid-organ transplants and60–85% in patients receiving hematopoietic stem cell transplants(HSCT) (1, 2). Antifungal agents such as polyenes, triazoles, andechinocandins can be rendered ineffective by suboptimal hostimmunity, emergence of drug-resistant strains, and attendanttoxicities in the recipients (3, 4). Thus, new approaches fortreating IAs are needed. Because the adoptive transfer of ge-netically modified T cells expressing CD19-specific chimericantigen receptors (CARs) has resulted in successful treatment ofpatients with B-cell malignancies (5–9), we sought to determineif a CAR could be developed to redirect T-cell specificity toAspergillus.Among immunocompetent individuals, intact innate immunity

can prevent and eradicate IFI. Endogenous alveolar macro-phages recognize, phagocytize, and kill fungal spores (10), andneutrophils can recognize and eliminate germinating hyphae(11). Immunotherapeutic strategies such as adoptive transfer ofpreselected CD4+ T-cell clones can help control Aspergillus in-fection indirectly by producing interferon-gamma (IFN-γ) (12).An immunotherapeutic strategy deploying cytotoxic T cellsappears to be clinically advantageous, because such cells have anendogenous ability to kill, be propagated to large numbers exvivo, be genetically modified to recognize desired target anti-gens, and contribute to immunologic memory (8).We previously have redirected the specificity of genetically

modified T cells via the stable introduction of a CD19-specificsecond-generation CAR (8, 9). Recognition of CD19 was ach-ieved by a CAR exodomain composed of a CD19-specific single-

chain variable fragment attached to the T-cell surface via amodified IgG4 hinge and fragment-crystallized (Fc) region(13). CAR-dependent and MHC-independent T-cell activationcan be achieved through the CAR endodomain composed ofCD3-ζ and CD28 (14, 15). We modified this prototypical CARdesign to achieve recognition of carbohydrate by accommodatingthe pattern-recognition properties of Dectin-1 (16, 17). Thisdectin is a type II transmembrane protein expressed on macro-phages, neutrophils, and dendritic cells (18) and is specific forβ-glucans, which are glucose polymers consisting of β-1,3-glucanand β-1,6-glucan expressed on the cell wall of fungi (19). BecauseDectin-1 mediates recognition of Aspergillus fumigatus (20), wehypothesized that the extracellular portion of Dectin-1 couldbe adapted as the specificity domain for a CAR (designated“D-CAR”) on T cells to redirect their specificity for this fungus.We report that T cells can be genetically modified by using the

Sleeping Beauty (SB) transposon/transposase system to expressD-CAR stably and can be propagated selectively on artificialactivating and propagating cells (aAPCs). We demonstrate thatthe D-CAR+ T cells (i) bind specifically to laminarin (which isrich in β-1,3-glucan), (ii) exhibit a central memory phenotype,(iii) inhibit growth of Aspergillus hyphae even in the presence of

Significance

Patients with compromised T-cell function are at risk for op-portunistic fungal infections. We have developed a novel ap-proach to restore immunity by using a fungal pattern-recognitionreceptor Dectin-1 to redirect T-cell specificity to carbohydrateantigen in the fungal cell wall. We did so by genetically modifyingT cells using the nonviral Sleeping Beauty gene-transfer systemto enforce expression of a chimeric antigen receptor (CAR) thatrecapitulates the specificity of Dectin-1 (D-CAR). The D-CAR+ Tcells can be electroporated and propagated on artificial activatingand propagating cells in a manner suitable for human application,enabling this immunology to be translated into immunotherapy.This approach has implications for genetically modifying T cells toexpress CARs with specificity for carbohydrate and thus broad-ening their application in the investigational treatment of patho-gens and malignancies.

Author contributions: P.R.K., P.R.M., D.P.K., and L.J.N.C. designed research; P.R.K., P.R.M.,N.D.A., S.M., T.M., J.R., S.O., J.K., and L.Z. performed research; P.R.K. and P.R.M. contrib-uted new reagents/analytic tools; P.R.K., P.R.M., H.S., B.R., J.K., A.M.N., M.H.H., D.A.L., andR.E.C. analyzed data; and P.R.K., P.R.M., D.P.K., and L.J.N.C. wrote the paper.

Conflict of interest statement: L.J.N.C. founded and owns InCellerate, Inc. He has patentswith Sangamo BioSciences with artificial nucleases. He consults with Targazyme, Inc.(formerly American Stem Cells, Inc.), GE Healthcare, Ferring Pharmaceuticals, Inc., andBristol-Myers Squibb. He receives honoraria from Miltenyi Biotec.

This article is a PNAS Direct Submission.1P.R.K. and P.R.M. contributed equally to this work.2To whom correspondence should be addressed. E-mail: [email protected].

This article contains supporting information online at www.pnas.org/lookup/suppl/doi:10.1073/pnas.1312789111/-/DCSupplemental.

10660–10665 | PNAS | July 22, 2014 | vol. 111 | no. 29 www.pnas.org/cgi/doi/10.1073/pnas.1312789111

Dow

nloa

ded

by g

uest

on

Nov

embe

r 17

, 202

0

Page 2: Bioengineering T cells to target carbohydrate to treat ... · Bioengineering T cells to target carbohydrate to treat opportunistic fungal infection Pappanaicken R. Kumaresana,1, Pallavi

dexamethasone, and (iv) target Aspergillus infection in the skinand lung of immunocompromised mice. Thus, using a genetransfer and propagation approach adapted for human applica-tion (21, 22), we have demonstrated that an innate immune re-sponse to fungi can be harnessed by bioengineering cytotoxicT cells. This demonstration provides the foundation for testingwhether D-CAR+ T cells can be infused to improve the survivalof immunocompromised patients who develop opportunistic IAinfection.

ResultsGenerating D-CAR+ Human T Cells. As expected, neither circulatingnor propagated human αβ T cells express Dectin-1 or Dectin-2and thus do not directly recognize Aspergillus using these pattern-recognition receptors. To generate a CAR with the specificity ofDectin-1 capable of activating T cells from peripheral blood, wefused the extracellular domain of human Dectin-1 (23) to ourstandard CAR cassette (15) encoding a modified IgG4 hinge/Fc(13), human CD28 transmembrane and cytoplasmic domains,and the CD3-ζ signaling motif. The resulting D-CAR constructwas subcloned as an SB transposon (Fig. 1A) and electro-trans-ferred with SB11 transposase into peripheral blood-derived hu-man primary T cells (Fig. S1). These data demonstrate that atype II transmembrane receptor can be fashioned into a CAR(containing HA and FLAG epitope tags to validate assembly)and stably expressed on T cells following transposition (Fig.S1B). The propagation of D-CAR+ T cells was not significantly

different from CD19-specific CAR+ T cells cocultured withaAPC (clone #4) (24) that activates/propagates T cells viatransgenic human CD19. The addition of γ-irradiated aAPC(Fig. S2) every 7 d was calculated to support a 6-log numericexpansion (from 106 to 1012) for both D-CAR+ and CD19-spe-cific CAR+ T cells (Fig. 1B). There was no outgrowth of CD3−

cells [e.g., natural killer (NK) cells]. Thirty-five days after elec-troporation, the propagated T cells were stained with mAbspecific for Dectin-1 and analyzed by flow cytometry. Almost all(average 97.6% ± 1.6; n = 3) of the T cells coexpressed CD3 andD-CAR; 90–95% of the propagated D-CAR+ T cells coex-pressed CD8 (Fig. 1C); and, as anticipated, there was no ex-pression of D-CAR on CD19-specific CAR+CD8+ or CAR+

CD4+ T cells. As such, the CD19-specific CAR+ T cells serve asa negative control (hereafter termed “control T cells”). Thesedata demonstrate that D-CAR+ T cells can be expanded nu-merically on aAPCs using an approach similar to that used forthe manufacture of clinical-grade CD19-specific T cells in ourongoing clinical trials.

Genetic Signature and Immunophenotype of D-CAR+ T Cells.We usednCounter analysis of direct digital readouts to quantify mRNAabundance (25), coding for IFN-γ (a Th1 proinflammatorymarker), granzymes A/B (markers of cytotoxicity), DNAX-acti-vating protein (DAP-10) for PI 3-kinase signaling, NK-cell–activating receptor (NKG2D) for costimulation, and zeta-chain–associated protein kinase 70 (ZAP70) and lymphocyte-specificprotein tyrosine kinase (Lck) for T-cell activation. Comparedwith unmodified T cells in peripheral blood, the D-CAR+ T cellsexhibited an increase in levels of IFN-γ (eightfold), granzyme A(94-fold), granzyme B (12-fold), DAP10 (threefold), NKG2D(threefold), Lck (threefold), and ZAP-70 (twofold) (Fig. 1D). Inaddition, there was a 15-fold decrease in killer cell lectin-likereceptor subfamily G, member 1 (KLRG1), which is consistentwith the preservation of T cells that avoid replication senescence(26, 27), and a six- to sevenfold decrease in the nuclear receptorretinoic acid receptor–related orphan receptor α (RORα), whichcontributes to the development of CD4+ Th17 cells (28). Thetherapeutic activity of CD19-specific CAR+ T cells in vivo hasbeen shown to correlate positively with T-cell persistence and isassociated with a central memory (TCM) phenotype (29). Gatingon the CD3+ D-CAR+ subset showed that 96% of the cellscoexpress CD45RO, as is consistent with an outgrowth of mem-ory T cells. Further analysis of the CD45RO+ T cells revealedthat 84% exhibited a TCM immunophenotype on the basis ofcoexpression of CD28, CD62L, and CCR7 (Fig. 1E). These datasuggest that D-CAR+ T cells numerically expanded on aAPCsmay sustain persistence after adoptive transfer with the potentialto provide long-term protection from IA. Moreover, using ourdirect T-cell receptor (TCR) expression analysis (DTEA)method, we found no significant difference in the abundance anddiversity of TCR Vα and Vβ family members expressed by un-modified and genetically modified D-CAR+ T cells (Fig. S3).These data indicate that D-CAR+ T cells emerge from andmaintain a polyclonal population.

Redirected Specificity by D-CAR+ T Cells. Dectin-1 binds to theglucose polymer laminarin that is similar to the sugar moietyfound in fungal walls but does not bind to mannan, the mannosepolymer derived from Saccharomyces cerevisiae (30). D-CAR+ Tcells exhibited the same binding preference, because laminarin,but not mannan, abrogated binding of mAb specific for Dectin-1to D-CAR on genetically modified T cells in a dose-dependentmanner (Fig. 2A). The specificity was evaluated further bycoculturing genetically modified T cells with germinatingAspergillus spores using three in vitro assays. We used microscopyto demonstrate that fungal growth was inhibited significantly byD-CAR+ T cells as compared with CD19-specific CAR+ controlT cells (Fig. 2B). Preincubation of a rabbit polyclonal antibodyraised against the soluble extract of A. fumigatus did prevent theability of D-CAR+ T cells to damage fungal growth, suggesting

Infe

rre

d c

ell

nu

mb

er

0 7 14 21 28

10 6

10 7

10 8

10 14

10 12

35

Days of co-culture

D-CAR+ CD3+ T cells

CD19-specific CAR+ CD3+ T cells 88.0% 8.0%

0.5%3.5%

4.0% 88.0%

7.0%1.0%

0.0% 1.0%

95.0%

1.0% 1.0%

4.0%94.0% 4.0%

CD8CD4

De

ctin

-1D-C

AR

+

T c

ells

CD

19

-sp

eci

fic

CA

R+ T

ce

lls

Tran

scri

pt

nu

mb

er

T cells from peripheral blood

D-CAR+ T cells

50000

25000

10000

5000

2000

1000

0

KLRG1

RORα

EOM

ES

ZAP-70

Lck

NKG2D

DAP10

D-C

AR

IFN-γ

Gzm B

Gzm A

5.0% 94.0%

1.0%0.0%

0.0%

96.0%0.5%CD

45R

A

12%

4.0% 0.5%

5.0% 84.0%

0.0% 11.0%

CCR7

CD45RODectin-1

CD28

3.5%

83.0%

CD

3C

D62

L

IgG4 (Ec)

CD28(Cyto)

CD3ζ(cyto)

(Flag)3 (HA)

3Dectin-1 (Ec)

-COOH--NH2-

ExtracellularIntracellular

CD28(Tm) A

B C

D E

Fig. 1. Bioengineering of D-CAR+ T cells. (A) Components of D-CAR. Theextracellular (Ec) sugar-binding domain of human Dectin-1 was fused inframe to a modified human IgG4 hinge and Fc region and the trans-membrane (Tm) and cytoplasmic (Cyto) domain of human CD28, and CytoCD3-ζ. HA3 and FLAG3 epitope tags were fused on the C and N termini, re-spectively. (B) Numeric expansion of T cells cocultured with aAPC clone #4preloaded with Dectin-1–specific mAb in the presence of soluble IL-2 andIL-21. Data are plotted as mean ± SD from three different donors. Upwardarrows indicate the addition of γ-irradiated aAPC. (C) Expression of D-CARon CD4+ and CD8+ T cells after electroporation and propagation for 35 d.CD19-specific CAR+ T cells from the same donor were used as a gatingcontrol. (D) Comparative gene-expression profiles of the expanded D-CAR+ Tcells and circulating T cells from healthy donors. Abundance of mRNAtranscripts was measured by digital gene expression using the nCounteranalysis system. (E) D-CAR+ T cells propagated for 35 d were costained withanti-CD3 and anti–Dectin-1 mAbs, in addition to staining with anti-CD45RA,CD45RO, CD28, CD62L, and CCR7, and were analyzed by flow cytometry.D-CAR+CD3+CD45RA−RO+ T cells were gated for expression of CD62L, CD28,and CCR7 to define central memory T cells.

Kumaresan et al. PNAS | July 22, 2014 | vol. 111 | no. 29 | 10661

IMMUNOLO

GYAND

INFLAMMATION

Dow

nloa

ded

by g

uest

on

Nov

embe

r 17

, 202

0

Page 3: Bioengineering T cells to target carbohydrate to treat ... · Bioengineering T cells to target carbohydrate to treat opportunistic fungal infection Pappanaicken R. Kumaresana,1, Pallavi

that the redirected specificity is achieved by a specific interactionwith the fungal cell wall. Next, we investigated the capacity ofgenetically modified T cells to kill germinating conidia usinga colorimetric assay to reveal the viability of Aspergillus based onthe ability to reduce a tetrazolium dye (31). Coculture ofD-CAR+ T cells led to an ∼75–80% loss of viability in germi-nating spores compared with a 15–30% loss of viability whencocultured with CD19-specific CAR+ T cells (P < 0.01; Fig. 2C).In the third in vitro assay, we undertook video time-lapsed micros-copy (VTLM) to visualize serially the targeting of germinatingAspergillus by D-CAR+ T cells (Movie S1) and CD19-specificCAR+ T cells (Movie S2) compared with untreated controls(Movie S3). D-CAR+ T cells and background low levels ofCD19-specific T cells bound to germlings (Fig. 2 D, c and g) by3 h. However, by 7 h there was a rapid influx of D-CAR+ T cells(Fig. 2 D, d) but not of control T cells (Fig. 2 D, h). The con-sequence of this binding of D-CAR+ T cells was destruction ofhyphal growth (Fig. 2 D, d) compared with the control T cells(Fig. 2 D, h) and in the absence of T cells (Fig. 2 D, k and l). Themean hyphal length at 24 h was calculated from VTLM toquantify the ability of T cells to destroy hyphae. Compared withcontrol T cells, D-CAR+ T cells reduced hyphal length from 180μm to 70 μm (62% inhibition). In the absence of T cells, hyphaegrew on average to 200 μm over same time period (Fig. 2E).These imaging data support the ability of D-CAR+ T cells totarget the germinating hyphae specifically and as desired, andnot the dormant Aspergillus conidia.

Effect of Dexamethasone on D-CAR+ T Cells. A likely translationalapplication for infusing D-CAR+ T cells will be in recipients

of solid-organ transplants or allogeneic HSCT. Dexamethasone(DEX), a corticosteroid systemically administered for immunosup-pression, has been demonstrated to down-regulate endogenousDectin-1 expression (32) and predisposes patients to fungalinfections (33). We evaluated the effect of DEX on the expres-sion of D-CAR on T cells and found that levels of the introducedimmunoreceptor diminished partially in a dose-dependent man-ner, so that 64% (n = 2) of genetically modified T cells continuedto express D-CAR in 1 μM of DEX. Despite this down-regulationof the introduced immunoreceptor, these T cells retained theirability to damage hyphae (Fig. 3A).

Activation of D-CAR+ T Cells by Aspergillus Germlings. To corrobo-rate the hyphal killing, we evaluated up-regulation of CD107a(LAMP1) that is expressed on the T-cell surface following cy-totoxic degranulation (34). We observed a threefold up-reg-ulation of CD107a on D-CAR+ T cells after coculture withAspergillus germlings (Fig. 3B). Damage to hyphae likely wasmediated by perforin and granzymes, because D-CAR+ T cellsexpressed perforin (92%) and granzyme B (100%) (Fig. 3C).Furthermore, perforin secretion was increased (P < 0.05) inconditioned supernatant after stimulation with aAPCs (clone#4) preloaded with Dectin-1–specific mAb (Fig. 3D). Resultsobtained from our customized bar-coded probe set used tomeasure abundance of mRNA levels suggested that D-CAR+ Tcells produced higher IFN-γ levels than T cells from peripheralblood (Fig. 1D). Therefore, IFN-γ expression was measured inD-CAR+ T cells exposed to germinating Aspergillus, and we alsoobserved a six- to sevenfold increase in IFN-γ protein levels

C1 D1

BA C D

A1

E F G H

I J K L

0.03h 1.15h 3.15h 7.15h

Hyp

ha

e a

nd

D-C

AR

+ T

ce

lls

Hyp

ha

e a

nd

CD

19

-

spe

cifi

c C

AR

+ T

ce

llsH

yph

ae

97.0%3.0% 6.0% 94.0%

Dectin

Co

un

tsLaminarin Mannan

CD19RCD28 D-CAR

Donor # 1 Donor # 2 Donor # 30

2 0

4 0

6 0

8 0

1 00

% h

yph

al d

amag

eM

ea

n h

yp

ha

l le

ng

th (μ

m)

0

50

100

150

200

250

** ** **

**

B1

1G1 H1E1 F1

K1 L1I1 J1

D-CAR+ T cells

and Aspergillus- specific Ab

CD19-specific

CAR+ T cells

D-CAR+ T cells No treatment

No treatment

CD19-specific

CAR+ T cells

D-CAR+ T cells

A

C D

B

E

Fig. 2. Specificity and damage to Aspergillus hyphae by D-CAR+ T cells. (A)Binding of D-CAR+ T cells by Dectin-1–specific mAb in the absence or pres-ence (200 μg/mL, solid line; 400 μg/mL, dashed line; and 600 μg/mL, dottedline) laminarin or mannan, as evaluated by flow cytometry. Shaded andopen regions in the histograms represent mAb binding in the absence orpresence of sugar, respectively. (B) Preincubation with Aspergillus-specificpolyclonal antibody blocked binding of D-CAR+ T cells to hyphae. Fungalgrowth was inhibited more by D-CAR+ T cells than by CD19-specific CAR+ Tcells. (C) Comparison by XTT assay of the ability of D-CAR+ and CD19-specificCAR+ T cells to damage germinating A. fumigatus hyphae specifically; **P <0.01. (D) D-CAR+ effector (E) T cells were coincubated with targets (T) GFP+

AF293 swollen conidia (E:T ratio of 2:1) and imaged continuously by VTLM.Conidia germlings cocultured with CD19-specific CAR+ T cells and without Tcells served as negative control. (See Movies S1–S3.) Pictures of conidia takenat various time points under fluorescent light (a–l) and with white light (a1–l1) show inhibition of growth by germinating conidia (green fluorescence) byD-CAR+ T cells (a–d), CD19-specific CAR+ T cells (e–h), and in the absence of Tcells (i–l). (Scale bars: 10 μm.) (E) At 24 h, the ability of T cells to inhibit As-pergillus germination as measured by hyphae length obtained from VTLM.Data are shown as mean ± SD. **P < 0.01; n = 3.

Co

un

ts

Perforin

92.5%

Granzyme B

100.0%

D-CAR+ T cellsHyphal damage

0.0 0.1 1.0

Pe

rce

nt

40

60

80

100

*

DEX (μM)

1

Pe

rfo

rin

Fold

Incr

ea

se

2

3

4

0

IFN

-γ (μ

g/m

L)

No D

NA

CD19RCD

28

D-C

AR

PMA/Io

n0.0

0.5

1.0

1.58

101214

0

5

10

15

D-C

AR

+ C

D1

07

a+ T

ce

lls

Without

hyphae

With

hyphae

86% 4%

0%9 %

65% 25%

1 %9 % 5 %1 %

96% 1%

3 % 0 %

97% 1%

0 %2 %

8% 92%

0 %0 %

9%

An

ti-De

ctin-1

IFN-γ

An

ti-hu

ma

n Fc

Without hyphae With hyphae PMA/Ionomycin

D-C

AR

+ T

ce

llsC

D1

9-s

pe

cifi

c

CA

R+ T

ce

lls

*

**

**

A B C

DE

F

85%

Fig. 3. D-CAR+ T cells are activated by germinating Aspergillus. (A) D-CAR+

T cells cultured for 1 wk on aAPC preloaded with Dectin-1–specific mAb inthe presence of DEX and soluble IL-2 and IL-21 were analyzed for expressionof D-CAR by flow cytometry (open bars; D-CAR+ T cells cultured in the ab-sence of DEX served as a control) and for their ability to kill A. fumigatushyphae by XTT assay (gray bars; A. fumigatus cultured without T cells servedas control). Data are shown as mean ± SD; n = 2. (B) CD107a expression onD-CAR+ T cells in the absence or presence of A. fumigatus hyphae, analyzedby flow cytometry. Data are shown as mean ± SD; n = 3. (C) Expression ofperforin and granzyme B analyzed by flow cytometry; shaded regions areisotype controls. (D) Perforin secretion in conditioned T-cell culture super-natants after stimulation by aAPCs loaded with Dectin-1–specific mAb. Foldchanges in perforin level were calculated by normalizing against unstimu-lated T cells. (E) D-CAR+ T cells and CD19-specific CAR+ T cells (expressingCD19RCD28) were incubated with germinating A. fumigatus hyphae for 4–6 h.The percentages of D-CAR+IFN-γ+ T cells and CD19-specific CAR+IFN-γ+ T cellswere measured by flow cytometry. T cells stimulated with phorbol12-myr-istate13-acetate/ionomycin served as positive controls. Shown are repre-sentative dot plots from two independent experiments. (F) IFN-γ cytokinelevels were measured in T-cell culture conditioned supernatants after stim-ulation with 50 μg/mL Aspergillus cell lysate.*P < 0.05; **P < 0.01.

10662 | www.pnas.org/cgi/doi/10.1073/pnas.1312789111 Kumaresan et al.

Dow

nloa

ded

by g

uest

on

Nov

embe

r 17

, 202

0

Page 4: Bioengineering T cells to target carbohydrate to treat ... · Bioengineering T cells to target carbohydrate to treat opportunistic fungal infection Pappanaicken R. Kumaresana,1, Pallavi

(Fig. 3E). In contrast, no change in IFN-γ levels was observedin control T cells cocultured with germinating Aspergillus. Wealso validated the presence of soluble IFN-γ in conditionedsupernatants of D-CAR+ T cells (P < 0.01) cultured with fungalhyphae extract (Fig. 3F). We detected no secreted IL-17, as isconsistent with decreased levels of mRNA coding for RORα(28). In aggregate, these data demonstrate that electroporatedand propagated D-CAR+ T cells are cytotoxic in response to hy-phae from Aspergillus and can secrete proinflammatory cytokines.

Targeting Aspergillus Infection in Mice by D-CAR+ T Cells. To assessthe in vivo therapeutic efficacy of the D-CAR+ T cells, wetreated immunosuppressed NOD SCID-γ (NSG) mice bearingIA with D-CAR+ T cells. Mice that received no T cells (PBScontrol) or CD19-specific T cells demonstrated Aspergillus in-fection as shown by staining of lung tissue sections with FITC-conjugated Aspergillus-specific antibody. In contrast, treatmentwith D-CAR+ T cells resulted in diminished fungal infection andretention of D-CAR+ T cells (Fig. 4A). Indeed, infused T cellsidentified by phycoerythrin (PE)-conjugated CD3-specific mAbrevealed that fungal hyphae were preferentially covered withD-CAR+ T cells rather than CD19-specific CAR+ T cells. An an-tifungal effect mediated by D-CAR+ T cells was supported by theobservation that the number of visualized genetically modified Tcells, as assessed by red fluorescence, was greater in the mice thatreceived D-CAR+ T cells than in the two controls (P < 0.001;Fig. 4B). In aggregate, quantification of fluorescence showedthat mice receiving D-CAR+ T cells had a significantly lower

pulmonary Aspergillus load than mice treated with control CAR+

T cells or no T cells (P < 0.001; Fig. 4B). This result was sup-ported by measurements of fungal burden in lungs by quantita-tive PCR (qPCR), which revealed that the introduced germlingsunderwent greater damage in the mice receiving D-CAR+ T cellsthan in controls (P < 0.001; Fig. 4C). We extended our obser-vation to another clinically relevant model of IA. Previously, ithas been shown that skin lesions and fungal burden are directlyproportional, and thus the infected surface area is an objectivemeasure of the degree of fungal infection (35). We observed thatsurface area of cutaneous fungal lesions (Fig. 4D) was smaller inmice that received D-CAR+ T cells than in control mice thatreceived CD19-specific CAR+ T cells or no T cells (P < 0.01; Fig.4E). These in vivo data support our in vitro observations thatD-CAR+ T cells can target IA.

Bispecific T Cells Coexpressing D-CAR and CD19-Specific CAR. Patientsat risk for progression of B-cell malignancies, such as patientswith advanced disease undergoing HSCT, are also vulnerable forIA. Therefore, we investigated if the SB system could be adoptedto coexpress CD19-specific CAR and D-CAR in T cells. T cellsthat underwent double transposition were propagated on aAPCclone #4 (36) and were demonstrated by flow cytometry tocoexpress the two CARs (Fig. S4A). To establish the ability ofD-CAR to activate T cells expressing two CAR species, we againdemonstrated that hyphae growth could be targeted on Aspergillusgermlings (Fig. S4B) with damage proportional to the ratio of Tcells to fungal burden (Fig. S4C). Specificity for CD19 was vali-dated by chromium release assay for CD19+ vs. CD19− tumor cells(Fig. S4D). These data demonstrate that T cells can be engineeredto have specificity for both carbohydrate and protein antigens.

DiscussionWe demonstrate a previously unidentified approach for immu-notherapy of Aspergillus based on redirecting T-cell specificitythrough a CAR that recognizes carbohydrate antigen: D-CARwith specificity of Dectin-1 fused to CD28 and CD3-ζ cyto-plasmic signaling domain that delivers a fully competent T-cell–activation signal as defined by killing, cytokine production, andproliferation. We genetically modified primary circulating Tcells using our SB transposon/transposase system that weadapted for human gene transfer. Indeed, we have achievedinstitutional and federal regulatory approvals for four clinical trials(NCT00968760, NCT01497184, NCT01362452, NCT01653717)infusing autologous and allogeneic T cells genetically modifiedwith an SB encoding a CD19-specific CAR and propagationon aAPC (clone #4) (21, 22, 37). To generate clinically relevantnumbers of D-CAR+ T cells for adoptive transfer, T cells wereexpanded numerically on “designer” aAPC. These geneticallymodified T cells coexpress perforin and granzyme and exhibit anability to recognize and lyse Aspergillus germlings. The thera-peutic potential of these T cells is highlighted further by themajority of D-CAR+ T cells (84%) exhibiting a TCM phenotypethat likely can self-renew as well as differentiate into effector Tcells in vivo (38). nCounter Digital profiling was used to ex-amine the abundance of mRNA species; both the transcriptionfactors Eomesodermin and KLRG1 were down-regulated inpropagated D-CAR+ T cells, supporting the observation thatinfused D-CAR+ T cells can sustain proliferation without ter-minal differentiation and avoid replicative senescence (39, 40).Because this is the first time, to our knowledge, that a pattern-

recognition receptor has been adapted to redirect T-cell speci-ficity, we used multiple experiments (fungal cell-killing assay,cytokine production, up-regulation of CD107a, VTLM, and twomouse models) to assess the ability of D-CAR+ T cells to targetgerminating Aspergillus. Based on the results obtained from invitro and in vivo studies, D-CAR+ T cells can directly target andcontrol Aspergillus infection. These assays also demonstrated thatD-CAR is able to activate proinflammatory and cytolytic ma-chinery such as the perforin/granzyme pathway of geneticallymodified T cells. The exocytosis of cytolytic granules apparently

20x

Me

an

inte

nsi

ty c

ou

nts

/ o

bje

ct

PBS

CD3-PE Aspergillus-FITC

40x

CD19-specific

CAR+ T cellsD-CAR+ T cells

CD19RCD28

D-C

AR

No DNA

***

***

***

CD19RCD28

D-C

AR

80

60

40

20

0

No DNA

80

60

40

20

0

*abc

Su

rfa

ce a

rea

of

infe

ctio

n (

mm

2)

CD19RCD28

D-C

AR

CD19RCD28

D-C

AR

Aspergillus

Aspergillus

Aspergillus

Aspergillus

5

6

7

8

9

1 0

CD19RD28

D-C

AR

Aspergillus

Log

Y (

A. fum

igatus

CE

DN

A)

***

***

A

B C

D

E

Fig. 4. Targeting Aspergillus infection in mice by D-CAR+ T cells: (A) Pul-monary Aspergillus infection in NSG mice. Fluorescent microscopic pictures oflung sections of Aspergillus (AF293) in mice that received PBS, CD19-specificCAR+ T cells, and D-CAR+ T cells. (B) Mean fluorescence intensity for stainingwith anti-human CD3-PE and anti-Aspergillus-FITC was calculated using In-form imaging software. Values are shown as mean fluorescent intensity(counts per image objects) from three lung slides per group. ***P < 0.001. (C)Quantification of A. fumigatus burden in lungs of mice 4 d after infection, asdetermined by qPCR. Data are expressed as the number of conidial equivalent(CE) copies of DNA in aliquots of lung homogenates. Data are shown as mean± SD; ***P < 0.001 compared with control mice receiving PBS (labeled “As-pergillus”) or CD19-specific CAR+ T cells; n = 5 mice. (D) Cutaneous Aspergillusinfection in NSG mice. Mice received no therapy, CD19-specific (CD19RCD28)CAR+ T cells, D-CAR+ T cells, and T cells expressing no CAR (No DNA). Site offungal lesions are indicated by arrows. After 96 h, mice were anesthetized,and skin lesions were measured. (E) Cutaneous fungal burden of mice treatedwith D-CAR+ T cells compared with Aspergillus without treatment, CD19-specific CAR+ T-cells, or (c) no DNA electroporated T-cells. *P < 0.05, treat-ment with D-CAR+ cells vs. other three conditions; n = 5.

Kumaresan et al. PNAS | July 22, 2014 | vol. 111 | no. 29 | 10663

IMMUNOLO

GYAND

INFLAMMATION

Dow

nloa

ded

by g

uest

on

Nov

embe

r 17

, 202

0

Page 5: Bioengineering T cells to target carbohydrate to treat ... · Bioengineering T cells to target carbohydrate to treat opportunistic fungal infection Pappanaicken R. Kumaresana,1, Pallavi

can disrupt fungal germination directly, as evident from thereports that granzyme-deficient mice are susceptible to IFI (41,42). The production of IFN-γ from the D-CAR+ T cells mayfurther augment immunity to IFIs, because pharmacological dos-ing of recombinant IFN-γ (43) or of IFN-γ derived from CD4+

helper T cells or NK cells has been shown to augment anti-As-pergillus activity (44, 45). However, it remains to be determinedwhether the D-CAR–dependent production of IFN-γ can con-tribute directly to the clearance of fungal infections or indirectlythrough the activation of granulocytes. Other cytokines, such asIL-17, produced by subsets of T cells also may participate in an-tifungal immunity by activating neutrophils (46). However, we didnot observe IL-17 secretion by D-CAR+ T cells, as is is consistentwith decreased expression of RORα, which plays a critical role inaugmenting IL-17 production from T cells (28, 47).Targeting by D-CAR+ T cells may be augmented by combina-

tion therapies. For example, Aspergillus preexposed to caspofungincan unmask β-glucan residues in the cell wall of fungi and enhanceantifungal activity mediated by neutrophils (31). Engineering aT-cell response to Aspergillus hyphae also may overcome con-genital impairment of the innate immune system, as with theDectin-1 Y238X polymorphism (48). This mutation is associatedwith diminished Dectin-1 receptor activity, increased suscepti-bility to IFI among recipients of HSCT (48), and loss of TLR4-mediated signals during hyphal germination, thereby contribut-ing to the evasion of immune recognition by Aspergillus (49).To help translate our findings to clinical practice, D-CAR+ T

cells must maintain effector function in the presence of system-ically administered immunosuppressive medications. The corti-costeroid DEX can down-regulate expression of Dectin-1 ongranulocytes and thereby increase recipients’ susceptibility toIFI. Although we did observe a decrease in D-CAR expression inelectroporated/propagated T cells exposed to DEX, the level ofD-CAR expression still was sufficient to damage germinatingAspergillus hyphae. This result indicates that adoptive transfer ofD-CAR+ T cells may remain effective in patients at risk for IFIbecause of the systemic administration of DEX. In addition, wegenerated bispecific T cells using the SB system that coexpressCD19-specific CAR (CD19RCD28) and D-CAR. These T cellsexhibited specificity for both CD19 and Aspergillus, raising thepossibility that one population of T cells can be customized totarget relapse and infection, which are two common causes ofmorbidity and mortality after allogeneic HSCT.In summary, we report a bioengineering approach to redirect

the specificity of T cells by the expression of a novel CAR thatincorporates the pattern-recognition ability of Dectin-1 to rec-ognize Aspergillus germlings. The D-CAR design enabled us toadapt an immunoreceptor that operates within the landscape ofinnate immunity for expression in T cells to co-opt their en-dogenous cytolytic machinery to target Aspergillus hyphae. Thisapproach is clinically appealing, because long-lived T cells can begenetically modified (e.g., using the SB system) and propagatedto large numbers (e.g., using γ-irradiated aAPC) ex vivo incompliance with cGMP for Phase I/II trials. One proposedclinical application for the administration of donor-derivedD-CAR+ T cells is administration following allogeneic HSCT,because the introduced D-CAR is capable of recognizing β-glucanmoieties present on opportunistic fungal infections. Furthermore,the recognition of carbohydrate antigens by CAR broadens thetranslational appeal of genetically modified T cells to targetpathogens as well as tumor-associated carbohydrate antigens.

Materials and MethodsPlasmids. D-CAR design was based on our second-generation CD19-specificCAR, CD19RCD28 (15). In brief, the sequence encoding the extracellular (Ec)sugar-binding domain of human Dectin-1 (GenBank accession no. AY026769)was fused to the modified human IgG4 hinge and Fc regions (13), which inturn were combined with the transmembrane and cytoplasmic residues ofhuman CD28 molecule and then the human cytoplasmic CD3-ζ chain. Thehuman codon optimized (CoOp) D-CAR (CoOp D-CAR), synthesized by GeneArt,was fused at the (Ec) C terminus to a triplicate HA epitope (amino acid

sequence YPYDVPDYA) and to the FLAG3 epitope (amino acid sequenceDYKDDDDC) on the N terminus by PCR to obtain FLAG(CoOp)–D-CAR–HA3 (Fig.1A) with flanking 5′ SpeI and 3′ NotI restriction enzyme sites. After sequencevalidation, the FLAG3(CoOp)–D-CAR–HA3 fragment was subcloned into the SBtransposon DNA plasmid CoOpCD19RCD28/pSBSO (used to express CD19RCD28for human application), by restriction digestion at SpeI-NruI and NheI-SmaI sites,thus replacing the CD19RCD28 CAR fragment with the FLAG3(CoOp)–D-CAR–HA3

fragment to create the final plasmid CoOpD D-CAR/pSBSO (designated “pSBD-CAR”). The DNA plasmid pCMV-SB11 expresses the SB11 transposase (21).

Cells. Primary human T cells were isolated by density gradient centrifugationover Ficoll-Paque-Plus (GE Healthcare Bio-Sciences AB) from peripheral bloodobtained from the Gulf Coast Regional Blood Center (Houston, TX) afterinformed consent. All primary cells and cell lines were cultured in RPMImedium 1640 (HyClone Laboratories) supplemented with 2 mM Glutamax-1(catalog no. 35050-061; Life Technologies) and 10% heat-inactivated FBS(HyClone). D-CAR+ T cells were generated as previously described for CD19-specific CAR+ T cells (37). In brief, T cells were electroporated with DNAplasmids from the SB system coding for D-CAR and SB11 and were propa-gated on aAPCs (clone #4) preloaded with Dectin-1–specific mAb in thepresence of IL-2 and IL-21. Bispecific CD19-specific CAR+ T cells coexpressingD-CAR were generated using double transposition whereby the SB systemwas used to coexpress two CARs. Mock-electroporated (“no DNA”) CAR−

T cells were propagated on aAPCs (clone #4) preloaded with CD3-specificmAb OKT3 (catalog no. 16-0037-85; eBioscience) in the presence of IL-2 andIL-21 (50). All cell lines used in this study were authenticated by fingerprinting at the MD Anderson Cancer Center sequencing core facility. Addi-tional details are provided in SI Materials and Methods.

Fungal Isolates, Growth, and Germination. Two A. fumigatus isolates wereused: a reference wild-type strain, A. fumigatus 293 (AF293), and a strain of A.fumigatus expressing GFP (GFP-AF293, GFP+AF), a kind gift from Kieren Marr(The Johns Hopkins University School of Medicine, Baltimore, MD) (51). Bothisolates were grown on potato dextrose agar for 7 d at 37 °C before conidiawere harvested by flooding the plates with PBS containing 0.1% Tween 20(35). For killing assays, conidia were counted using a hemocytometer andresuspended at a concentration of 105 spores/mL in RPMI containing 10% FBS.One milliliter containing 105 spores in Eppendorf tubes was shaken (220 rpm,digital incubator shaker, Innova 4300, New Brunswick Scientific, Inc.) at 37 °Cfor 12–16 h for germination. Similarly, for VTLM, harvested GFP+AF conidiawere incubated and shaken for 4–5 h before analysis.

XTT Assay. The targeting of Aspergillus was assessed using a 2,3-bis(2-methoxy-4-nitro-5-sulfophenyl)-5-[(phenylamino) carbonyl]-sH-tetrazoliumhydroxide (XTT)-based colorimetric assay (31). Germinating conidia wereincubated for 2–3 h with 106 T cells (effector:target ratio, 10:1) in RPMI 1640with 10% FBS. CD19-specific CAR+ T cells were used as a negative control.After incubation, T cells were lysed with cold, sterile water (hypotonic lysis).Damage to germlings was calculated following the manufacturer’s protocol(XTT assay, catalog no. X4251; Sigma).

Animal Studies. All animal experiments were approved by the University ofTexas MD Anderson Cancer Center Institutional Animal Care and Use Com-mittee. Eight-week-old female NSG mice (Jackson Laboratory) weighing 18–20g were used and housed in a sterile facility. Cyclophosphamide (100 or 150mg/kg of body weight) was injected i.p. to achieve immunosuppression 4 dand 1 d before inoculation with Aspergillus (AF293) spores (defined as day 0).Pulmonary fungal infection was induced by infecting with AF293 conidia (1.5 ×106 per mouse) via the sino-pulmonary route (52). The mice were grouped (n =5) into four groups. The first group received no T cells; the second group re-ceived CD19-specific CAR+ T cells; the third group received D-CAR+ T cells; andthe fourth group received T cells that did not express CAR (“no DNA”). T cellswere dosed at 2 × 107 by i.v. injection every day for 3 days beginning on day 0.All surviving mice were killed on day 4 to determine fungal burden. The sameexperimental design was used to establish a cutaneous fungal infection, ex-cept that T cells were administered s.c. into the same site at a 1:10 ratio ofAF293 conidia (1.5 × 106) per mouse) to T cells (15 × 106 per mouse). Skinlesions were measured daily using digital calipers (Fisher Scientific), and thelongest and shortest diameters were recorded (35).

Statistics. Graphs were plotted using Prism v. 4.0 software (Graph PadSoftware). Statistical analysis was carried out using SAS 9.3 (SAS Inc.). A two-tailed P value of <0.05 was considered statistically significant. Skin lesionareas and fungal burden were compared among groups of mice using anunpaired Student t test.

10664 | www.pnas.org/cgi/doi/10.1073/pnas.1312789111 Kumaresan et al.

Dow

nloa

ded

by g

uest

on

Nov

embe

r 17

, 202

0

Page 6: Bioengineering T cells to target carbohydrate to treat ... · Bioengineering T cells to target carbohydrate to treat opportunistic fungal infection Pappanaicken R. Kumaresana,1, Pallavi

ACKNOWLEDGMENTS. We thank the flow cytometry and cellular imaging corefacility at the MD Anderson Cancer Center (MDACC) [supported by MDACCCancer Center Support Grant (CCSG) CA016672]; Dr. Perry Hackett (Universityof Minnesota) for assistance with the SB system; Dr. Kieren Marr (The JohnHopkins University) for helping develop the concept; Dr. Judy Moyes (MDACC)for editing; Dr. GeorgeMcNamara (MDACC) for help with image processing; andDr. Jianlang Dai (MDACC) for statistical analysis. This work was supported byCancer Center Core Grant CA16672; National Institutes of Health (NIH) GrantsR01 CA124782, CA120956, CA141303, and CA141303; NIH Grant R33 CA116127;NIH Grant P01 CA148600; the Burroughs Wellcome Fund; the Cancer Prevention

and Research Institute of Texas; the Chronic Lymphocytic Leukemia Global Re-search Foundation; the Defense Advanced Research Projects Agency (DefenseSciences Office); the Department of Defense; the estate of Noelan L. Bibler; theGillson Longenbaugh Foundation; the Harry T. Mangurian, Jr. Fund for LeukemiaImmunotherapy; the Institute of Personalized Cancer Therapy; the Leukemia andLymphoma Society; the Lymphoma Research Foundation; MDACC’s Sister Insti-tution Network Fund; the Miller Foundation; Mr. Herb Simons; Mr. and Mrs. JoeH. Scales; Mr. Thomas Scott; the National Foundation for Cancer Research; thePediatric Cancer Research Foundation; and the William Lawrence and BlancheHughes Children’s Foundation.

1. Marr KA, Carter RA, Boeckh M, Martin P, Corey L (2002) Invasive aspergillosis in al-logeneic stem cell transplant recipients: Changes in epidemiology and risk factors.Blood 100(13):4358–4366.

2. Steinbach WJ, et al. (2012) Clinical epidemiology of 960 patients with invasive as-pergillosis from the PATH Alliance registry. J Infect 65(5):453–464.

3. Kontoyiannis DP (2011) Antifungal prophylaxis in hematopoietic stem cell transplant re-cipients: The unfinished tale of imperfect success. BoneMarrow Transplant 46(2):165–173.

4. Georgiadou SP, Kontoyiannis DP (2012) The impact of azole resistance on aspergillosisguidelines. Ann N Y Acad Sci 1272:15–22.

5. Brentjens RJ, et al. (2013) CD19-targeted T cells rapidly induce molecular remissions inadults with chemotherapy-refractory acute lymphoblastic leukemia. Sci Transl Med5(177):177ra138.

6. Grupp SA, et al. (2013) Chimeric antigen receptor-modified T cells for acute lymphoidleukemia. N Engl J Med 368(16):1509–1518.

7. Kochenderfer JN, et al. (2012) B-cell depletion and remissions of malignancy alongwith cytokine-associated toxicity in a clinical trial of anti-CD19 chimeric-antigen-receptor-transduced T cells. Blood 119(12):2709–2720.

8. Jena B, Dotti G, Cooper LJ (2010) Redirecting T-cell specificity by introducing a tumor-specific chimeric antigen receptor. Blood 116(7):1035–1044.

9. Kebriaei P, et al. (2012) Infusing CD19-directed T cells to augment disease control inpatients undergoing autologous hematopoietic stem-cell transplantation for ad-vanced B-lymphoid malignancies. Hum Gene Ther 23(5):444–450.

10. Waldorf AR, Levitz SM, Diamond RD (1984) In vivo bronchoalveolar macrophage de-fense against Rhizopus oryzae and Aspergillus fumigatus. J Infect Dis 150(5):752–760.

11. Schaffner A, Douglas H, Braude A (1982) Selective protection against conidia bymononuclear and against mycelia by polymorphonuclear phagocytes in resistance toAspergillus. Observations on these two lines of defense in vivo and in vitro withhuman and mouse phagocytes. J Clin Invest 69(3):617–631.

12. Perruccio K, et al. (2005) Transferring functional immune responses to pathogensafter haploidentical hematopoietic transplantation. Blood 106(13):4397–4406.

13. Cooper LJ, et al. (2003) T-cell clones can be rendered specific for CD19: Toward theselective augmentation of the graft-versus-B-lineage leukemia effect. Blood 101(4):1637–1644.

14. Maher J, Brentjens RJ, Gunset G, Rivière I, Sadelain M (2002) Human T-lymphocytecytotoxicity and proliferation directed by a single chimeric TCRzeta /CD28 receptor.Nat Biotechnol 20(1):70–75.

15. Kowolik CM, et al. (2006) CD28 costimulation provided through a CD19-specific chi-meric antigen receptor enhances in vivo persistence and antitumor efficacy ofadoptively transferred T cells. Cancer Res 66(22):10995–11004.

16. Adachi Y, et al. (2004) Characterization of beta-glucan recognition site on C-typelectin, dectin 1. Infect Immun 72(7):4159–4171.

17. Brown GD (2006) Dectin-1: A signalling non-TLR pattern-recognition receptor. NatRev Immunol 6(1):33–43.

18. Taylor PR, et al. (2002) The beta-glucan receptor, dectin-1, is predominantly expressedon the surface of cells of the monocyte/macrophage and neutrophil lineages.J Immunol 169(7):3876–3882.

19. Bowman SM, Free SJ (2006) The structure and synthesis of the fungal cell wall. Bio-Essays 28(8):799–808.

20. Werner JL, et al. (2009) Requisite role for the dectin-1 beta-glucan receptor in pul-monary defense against Aspergillus fumigatus. J Immunol 182(8):4938–4946.

21. Huls MH, et al. (2013) Clinical application of Sleeping Beauty and artificial antigenpresenting cells to genetically modify T cells from peripheral and umbilical cordblood. J Vis Exp (72):e50070.

22. Maiti SN, et al. (2013) Sleeping beauty system to redirect T-cell specificity for humanapplications. J Immunother 36(2):112–123.

23. Willment JA, Gordon S, Brown GD (2001) Characterization of the human beta -glucanreceptor and its alternatively spliced isoforms. J Biol Chem 276(47):43818–43823.

24. Suhoski MM, et al. (2007) Engineering artificial antigen-presenting cells to expressa diverse array of co-stimulatory molecules. Mol Ther 15(5):981–988.

25. Geiss GK, et al. (2008) Direct multiplexed measurement of gene expression with color-coded probe pairs. Nat Biotechnol 26(3):317–325.

26. Henson SM, et al. (2009) KLRG1 signaling induces defective Akt (ser473) phosphory-lation and proliferative dysfunction of highly differentiated CD8+ T cells. Blood113(26):6619–6628.

27. Voehringer D, Koschella M, Pircher H (2002) Lack of proliferative capacity of humaneffector and memory T cells expressing killer cell lectinlike receptor G1 (KLRG1). Blood100(10):3698–3702.

28. Yang XO, et al. (2008) T helper 17 lineage differentiation is programmed by orphannuclear receptors ROR alpha and ROR gamma. Immunity 28(1):29–39.

29. Kalos M, et al. (2011) T cells with chimeric antigen receptors have potent antitumoreffects and can establish memory in patients with advanced leukemia. Sci Transl Med3(95):95ra73.

30. Adams EL, et al. (2008) Differential high-affinity interaction of dectin-1 with naturalor synthetic glucans is dependent upon primary structure and is influenced by poly-mer chain length and side-chain branching. J Pharmacol Exp Ther 325(1):115–123.

31. Lamaris GA, et al. (2008) Caspofungin-mediated beta-glucan unmasking and en-hancement of human polymorphonuclear neutrophil activity against Aspergillus andnon-Aspergillus hyphae. J Infect Dis 198(2):186–192.

32. Willment JA, et al. (2003) Dectin-1 expression and function are enhanced on alter-natively activated and GM-CSF-treated macrophages and are negatively regulated byIL-10, dexamethasone, and lipopolysaccharide. J Immunol 171(9):4569–4573.

33. Lionakis MS, Kontoyiannis DP (2003) Glucocorticoids and invasive fungal infections.Lancet 362(9398):1828–1838.

34. Betts MR, Koup RA (2004) Detection of T-cell degranulation: CD107a and b. MethodsCell Biol 75:497–512.

35. Ben-Ami R, Lewis RE, Leventakos K, Latgé JP, Kontoyiannis DP (2010) Cutaneousmodel of invasive aspergillosis. Antimicrob Agents Chemother 54(5):1848–1854.

36. Hurton LV, et al. (2013) Tethered IL-15 mutein on CD19-specific T cells sustains per-sistence when tumor antigen is low and can treat minimal residual disease. Mol Ther21:S237–S237.

37. Singh H, et al. (2013) Manufacture of clinical-grade CD19-specific T cells stably ex-pressing chimeric antigen receptor using Sleeping Beauty system and artificial anti-gen presenting cells. PLoS ONE 8(5):e64138.

38. Klebanoff CA, et al. (2005) Central memory self/tumor-reactive CD8+ T cells confersuperior antitumor immunity compared with effector memory T cells. Proc Natl AcadSci USA 102(27):9571–9576.

39. Pearce EL, et al. (2003) Control of effector CD8+ T cell function by the transcriptionfactor Eomesodermin. Science 302(5647):1041–1043.

40. Intlekofer AM, et al. (2005) Effector and memory CD8+ T cell fate coupled by T-betand eomesodermin. Nat Immunol 6(12):1236–1244.

41. Voskoboinik I, Dunstone MA, Baran K, Whisstock JC, Trapani JA (2010) Perforin:Structure, function, and role in human immunopathology. Immunol Rev 235(1):35–54.

42. Schmidt S, et al. (2011) Human natural killer cells exhibit direct activity against Asper-gillus fumigatus hyphae, but not against resting conidia. J Infect Dis 203(3):430–435.

43. Kelleher P, et al. (2006) Interferon-gamma therapy in two patients with progressivechronic pulmonary aspergillosis. Eur Respir J 27(6):1307–1310.

44. Beck O, et al. (2006) Generation of highly purified and functionally active human TH1cells against Aspergillus fumigatus. Blood 107(6):2562–2569.

45. Bouzani M, et al. (2011) Human NK cells display important antifungal activity againstAspergillus fumigatus, which is directly mediated by IFN-γ release. J Immunol 187(3):1369–1376.

46. Ouyang W, Kolls JK, Zheng Y (2008) The biological functions of T helper 17 cell ef-fector cytokines in inflammation. Immunity 28(4):454–467.

47. Ivanov II, Zhou L, Littman DR (2007) Transcriptional regulation of Th17 cell differ-entiation. Semin Immunol 19(6):409–417.

48. Cunha C, et al. (2010) Dectin-1 Y238X polymorphism associates with susceptibility to in-vasive aspergillosis in hematopoietic transplantation through impairment of both recipient-and donor-dependent mechanisms of antifungal immunity. Blood 116(24):5394–5402.

49. Netea MG, et al. (2003) Aspergillus fumigatus evades immune recognition duringgermination through loss of toll-like receptor-4-mediated signal transduction. J InfectDis 188(2):320–326.

50. O’Connor CM, et al. (2012) Adoptive T-cell therapy improves treatment of canine non-Hodgkin lymphoma post chemotherapy. Sci Rep 2:249.

51. Bretz C, et al. (2008) MyD88 signaling contributes to early pulmonary responses toAspergillus fumigatus. Infect Immun 76(3):952–958.

52. Lewis RE, Albert ND, Kontoyiannis DP (2008) Efficacy of single-dose liposomal am-photericin B or micafungin prophylaxis in a neutropenic murine model of invasivepulmonary aspergillosis. Antimicrob Agents Chemother 52(11):4178–4180.

Kumaresan et al. PNAS | July 22, 2014 | vol. 111 | no. 29 | 10665

IMMUNOLO

GYAND

INFLAMMATION

Dow

nloa

ded

by g

uest

on

Nov

embe

r 17

, 202

0