6
Tumor-specific IL-9producing CD8 + Tc9 cells are superior effector than type-I cytotoxic Tc1 cells for adoptive immunotherapy of cancers Yong Lu a , Bangxing Hong a , Haiyan Li a , Yuhuan Zheng a , Mingjun Zhang a , Siqing Wang b , Jianfei Qian a , and Qing Yi a,1 a Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195; and b Institute of Translational Medicine, The First Hospital, Jilin University, Changchun 130061, China Edited by James P. Allison, MD Anderson Cancer Center, University of Texas, Houston, TX, and approved January 9, 2014 (received for review September 17, 2013) Because cytokine-priming signals direct CD8 + T cells to acquire unique profiles that affect their ability to mediate specific immune responses, here we generated IL-9skewed CD8 + T (Tc9) cells by priming with Th9-polarized condition. Compared with type-I CD8 + cytotoxic T (Tc1) cells, Tc9 secreted different cytokines and were less cytolytic in vitro but surprisingly elicited greater antitumor responses against advanced tumors in OT-I/B16-OVA and Pmel-1/ B16 melanoma models. After adoptive transfer, Tc9 cells persisted longer and differentiated into IFN-γand granzyme-B (GrzB)pro- ducing cytolytic Tc1-like effector cells. Phenotypic analysis revealed that adoptively transferred Tc9 cells secreted IL-2 and were KLRG-1 low and IL-7Rα high , suggesting that they acquired a signature of youngerphenotype or became long-term lived cells with ca- pacity of self-renewal. Our results also revealed that Tc9-mediated therapeutic effect critically depended on IL-9 production in vivo. These findings have clinical implications for the improvement of CD8 + T-cell-based adoptive immunotherapy of cancers. adoptive cell therapy | less-exhausted T cells | T-cell lineage plasticity A doptive cell therapy (ACT) using ex vivo differentiated type-I CD8 + cytotoxic T (Tc1) cells has shown significant clinical promises for the treatment of established cancers (1). Recent clinical trials using ACT combined with lymphodepletion have resulted in objective responses in 5070% of patients with advanced melanoma (2). However, complete responses remain infrequent with most patients, and improvements to this approach are needed (2). Current understanding of determinants of suc- cessful CD8 + T-cell adoptive therapy includes, but is not limited to, persistence of transferred T cells (3), differentiation status of transferred T cells (4), telomere length (5), and lymphodepleting condition (6). In particular, administration of naïve or early ef- fector T cells in combination with active immunization and IL-2 can result in eradication of large established tumors (4). Cytokine priming signals direct CD8 + T cells to acquire unique profiles that affect their ability to mediate specific im- mune responses (7, 8). CD8 + T cells can acquire cytokine se- creting phenotypes that require transcription factors similar to those of T helper (Th) cells (9). Tc1 cells secrete IFN-γ and kill tumor targets by releasing cytotoxic molecules such as GrzB and Perforin (9). The contribution of adoptively transferred Tc1 cells in antitumor responses has been clearly established, and Tc1 has stronger therapeutic effect than Tc2 and regulatory CD8 + T cells (10). In addition, naïve CD8 + T cells can be differentiated into Tc17 cells in Th17 polarizing conditions (8). The effect of Tc17- mediated antitumor responses remains controversial with one group describing enhanced antitumor immunity displayed by Tc17 (8) and two more recent studies demonstrating that Tc1 cells are superior to Tc17 cells in mediating tumor regression after transfer (11, 12). However, IL-17 and IL-17producing T cells are tumor-promoting factors and can mediate IL-6-induced Stat3 activation to generate protumorigenic environment (13), which may limit the application of Tc17 for adoptive therapy. Nevertheless, identification of CD8 + T-cell subsets with optimal therapeutic potential remains a critical challenge for the advances of cancer immunotherapy. IL-9 has attracted renewed interest recently owing to the identification of the most consistent IL-9producing Th9 cells. IL-9 is a pleiotropic cytokine that has direct and indirect effects on multiple cell types (14). More recently, we and others reported that Th9 cell-derived IL-9 not only inhibited tumor progression, but also promoted greater tumor clearance than Th1 cells that have traditionally been considered as the most efficient CD4 + T-cell subset to generate antitumor immunity (1517). It would be interesting to test under Th9-polarizing conditions whether naïve CD8 + T cells could also differentiate into IL-9producing Tc9 cells, a potential CD8 + T-cell subset for ACT. In this study, we first analyzed the effect of Th9-polarizing medium on the priming of Tc9 cells. We found that differentiation of CD8 + T cells under Th9-polarizing conditions induced very low amounts of typical cytotoxic T lymphocyte (CTL)-expressing GrzB, Eomes, T-bet (encoded by Tbx21), and IFN-γ, while instead, promoted the development of IL-9positive Tc9 cells. Surprisingly, adoptive transfer of tumor-reactive Tc9 cells elicited greater an- titumor responses against large established tumors by differenti- ating into long-lasting IFN-γproducing Tc1-like cells in tumor- bearing mice. These results demonstrated the potential advantages of differentiation of tumor-reactive CD8 + T cells into Tc9 cells that may exert superior antitumor efficacy upon adoptive transfer. Significance Our laboratory has identified a critical role of IL-9 in promoting endogenous tumor-specific cytotoxic T lymphocyte response. In this study, we found that differentiation of CD8 + T cells under T helper 9-polarizing conditions induces the development of an IL-9producing less cytolytic IL-9skewed CD8 + T (Tc9) cell subset. Noticeably, adoptive transfer of tumor-reactive Tc9 cells elicited greater antitumor responses against large established tumors than classic type-I CD8 + cytotoxic T cells that are used in clinical protocols. Importantly, Tc9 cells have substantially enhanced persistence potential and possess the capacity to acquire/maintain effector function after transfer. Our results also revealed that Tc9-mediated therapeutic effect critically depended on IL-9 pro- duction in vivo. The ability of Tc9 cells to confer sustained anti- tumor responses might open an avenue for the advances of cancer immunotherapy. Author contributions: Y.L. and Q.Y. designed research; Y.L., H.L., and M.Z. performed research; J.Q. contributed new reagents/analytic tools; Y.L., B.H., Y.Z., and S.W. analyzed data; and Y.L. and Q.Y. wrote the paper. The authors declare no conflict of interest. This article is a PNAS Direct Submission. 1 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.1317431111/-/DCSupplemental. www.pnas.org/cgi/doi/10.1073/pnas.1317431111 PNAS | February 11, 2014 | vol. 111 | no. 6 | 22652270 IMMUNOLOGY Downloaded by guest on August 31, 2020

Tumor-specific IL-9 producing CD8 superior effector than ... · Tumor-specific IL-9–producing CD8+ Tc9 cells are superior effector than type-I cytotoxic Tc1 cells for adoptive immunotherapy

  • Upload
    others

  • View
    0

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Tumor-specific IL-9 producing CD8 superior effector than ... · Tumor-specific IL-9–producing CD8+ Tc9 cells are superior effector than type-I cytotoxic Tc1 cells for adoptive immunotherapy

Tumor-specific IL-9–producing CD8+ Tc9 cells aresuperior effector than type-I cytotoxic Tc1 cellsfor adoptive immunotherapy of cancersYong Lua, Bangxing Honga, Haiyan Lia, Yuhuan Zhenga, Mingjun Zhanga, Siqing Wangb, Jianfei Qiana, and Qing Yia,1

aDepartment of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195; and bInstitute of Translational Medicine, The First Hospital,Jilin University, Changchun 130061, China

Edited by James P. Allison, MD Anderson Cancer Center, University of Texas, Houston, TX, and approved January 9, 2014 (received for reviewSeptember 17, 2013)

Because cytokine-priming signals direct CD8+ T cells to acquireunique profiles that affect their ability to mediate specific immuneresponses, here we generated IL-9–skewed CD8+ T (Tc9) cells bypriming with Th9-polarized condition. Compared with type-I CD8+

cytotoxic T (Tc1) cells, Tc9 secreted different cytokines and wereless cytolytic in vitro but surprisingly elicited greater antitumorresponses against advanced tumors in OT-I/B16-OVA and Pmel-1/B16 melanoma models. After adoptive transfer, Tc9 cells persistedlonger and differentiated into IFN-γ– and granzyme-B (GrzB)–pro-ducing cytolytic Tc1-like effector cells. Phenotypic analysis revealedthat adoptively transferred Tc9 cells secreted IL-2 and wereKLRG-1low and IL-7Rαhigh, suggesting that they acquired a signatureof “younger” phenotype or became long-term lived cells with ca-pacity of self-renewal. Our results also revealed that Tc9-mediatedtherapeutic effect critically depended on IL-9 production in vivo.These findings have clinical implications for the improvement ofCD8+ T-cell-based adoptive immunotherapy of cancers.

adoptive cell therapy | less-exhausted T cells | T-cell lineage plasticity

Adoptive cell therapy (ACT) using ex vivo differentiatedtype-I CD8+ cytotoxic T (Tc1) cells has shown significant

clinical promises for the treatment of established cancers (1).Recent clinical trials using ACT combined with lymphodepletionhave resulted in objective responses in 50–70% of patients withadvanced melanoma (2). However, complete responses remaininfrequent with most patients, and improvements to this approachare needed (2). Current understanding of determinants of suc-cessful CD8+ T-cell adoptive therapy includes, but is not limitedto, persistence of transferred T cells (3), differentiation status oftransferred T cells (4), telomere length (5), and lymphodepletingcondition (6). In particular, administration of naïve or early ef-fector T cells in combination with active immunization and IL-2can result in eradication of large established tumors (4).Cytokine priming signals direct CD8+ T cells to acquire

unique profiles that affect their ability to mediate specific im-mune responses (7, 8). CD8+ T cells can acquire cytokine se-creting phenotypes that require transcription factors similar tothose of T helper (Th) cells (9). Tc1 cells secrete IFN-γ and killtumor targets by releasing cytotoxic molecules such as GrzB andPerforin (9). The contribution of adoptively transferred Tc1 cellsin antitumor responses has been clearly established, and Tc1 hasstronger therapeutic effect than Tc2 and regulatory CD8+ T cells(10). In addition, naïve CD8+ T cells can be differentiated intoTc17 cells in Th17 polarizing conditions (8). The effect of Tc17-mediated antitumor responses remains controversial with onegroup describing enhanced antitumor immunity displayed byTc17 (8) and two more recent studies demonstrating that Tc1cells are superior to Tc17 cells in mediating tumor regressionafter transfer (11, 12). However, IL-17 and IL-17–producing Tcells are tumor-promoting factors and can mediate IL-6-inducedStat3 activation to generate protumorigenic environment (13),which may limit the application of Tc17 for adoptive therapy.

Nevertheless, identification of CD8+ T-cell subsets with optimaltherapeutic potential remains a critical challenge for the advancesof cancer immunotherapy.IL-9 has attracted renewed interest recently owing to the

identification of the most consistent IL-9–producing Th9 cells.IL-9 is a pleiotropic cytokine that has direct and indirect effectson multiple cell types (14). More recently, we and othersreported that Th9 cell-derived IL-9 not only inhibited tumorprogression, but also promoted greater tumor clearance thanTh1 cells that have traditionally been considered as the mostefficient CD4+ T-cell subset to generate antitumor immunity(15–17). It would be interesting to test under Th9-polarizingconditions whether naïve CD8+ T cells could also differentiateinto IL-9–producing Tc9 cells, a potential CD8+ T-cell subset forACT. In this study, we first analyzed the effect of Th9-polarizingmedium on the priming of Tc9 cells. We found that differentiationof CD8+ T cells under Th9-polarizing conditions induced very lowamounts of typical cytotoxic T lymphocyte (CTL)-expressing GrzB,Eomes, T-bet (encoded by Tbx21), and IFN-γ, while instead,promoted the development of IL-9–positive Tc9 cells. Surprisingly,adoptive transfer of tumor-reactive Tc9 cells elicited greater an-titumor responses against large established tumors by differenti-ating into long-lasting IFN-γ–producing Tc1-like cells in tumor-bearing mice. These results demonstrated the potential advantagesof differentiation of tumor-reactive CD8+ T cells into Tc9 cellsthat may exert superior antitumor efficacy upon adoptive transfer.

Significance

Our laboratory has identified a critical role of IL-9 in promotingendogenous tumor-specific cytotoxic T lymphocyte response. Inthis study, we found that differentiation of CD8+ T cells underT helper 9-polarizing conditions induces the development of anIL-9–producing less cytolytic IL-9–skewed CD8+ T (Tc9) cell subset.Noticeably, adoptive transfer of tumor-reactive Tc9 cells elicitedgreater antitumor responses against large established tumorsthan classic type-I CD8+ cytotoxic T cells that are used in clinicalprotocols. Importantly, Tc9 cells have substantially enhancedpersistence potential and possess the capacity to acquire/maintaineffector function after transfer. Our results also revealed thatTc9-mediated therapeutic effect critically depended on IL-9 pro-duction in vivo. The ability of Tc9 cells to confer sustained anti-tumor responses might open an avenue for the advances ofcancer immunotherapy.

Author contributions: Y.L. and Q.Y. designed research; Y.L., H.L., and M.Z. performedresearch; J.Q. contributed new reagents/analytic tools; Y.L., B.H., Y.Z., and S.W. analyzeddata; and Y.L. and Q.Y. wrote the paper.

The authors declare no conflict of interest.

This article is a PNAS Direct Submission.1To 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.1317431111/-/DCSupplemental.

www.pnas.org/cgi/doi/10.1073/pnas.1317431111 PNAS | February 11, 2014 | vol. 111 | no. 6 | 2265–2270

IMMUNOLO

GY

Dow

nloa

ded

by g

uest

on

Aug

ust 3

1, 2

020

Page 2: Tumor-specific IL-9 producing CD8 superior effector than ... · Tumor-specific IL-9–producing CD8+ Tc9 cells are superior effector than type-I cytotoxic Tc1 cells for adoptive immunotherapy

ResultsIL-9–Producing Tc9 Cells Are Skewed Away from IFN-γ Production andCytolytic Phenotype. We first investigated the effect of Th9-po-larization conditions on the antigen-driven acquisition of an IL-9–producing Tc9-cell phenotype. Similar to the cytokine profileof Th9 cells, IL-9–skewed CD8+ T cells demonstrated di-minished mRNA expression of Ifng but enhanced expression ofIl9 and Il2, which were associated with diminished expression ofcytolytic molecules, such as GrzB and Perforin (Fig. 1A). Theexpression of Eomes and Tbx21, the transcriptional master reg-ulators that confer cytolytic lymphocyte lineage characteristics(18), were substantially suppressed in IL-9–skewed CD8+ T cells(Fig. 1B). However, the expression of Irf4 and Pu.1, two tran-scription factors governing Th9-cell lineage development (19),was significantly up-regulated in Tc9 cells (Fig. 1B). The sup-pressed expression of CTL hallmark molecules, such as GrzB,Perforin, Eomes, and Tbx21, suggested the impaired CTL de-velopment of Tc9 cells and possibly diminished cytotoxicity. In-deed, OT-I Tc1 cells showed strong cytotoxicity against tumortarget cells, whereas OT-I Tc9 cells exhibited minimal specificcytolytic activity (Fig. 1C and SI Appendix, Fig. S1). Therefore,compared with IL-2–primed IFN-γ–producing cytolytic Tc1cells, IL-9–skewed Tc9 cells were diverted from the classiccytolytic phenotype.

Characterization of Tc9 Cytokine Expression Profiles. To determinethe specific cytokine expression patterns of IL-9–skewed Tc9cells, we measured by ELISA the cytokine production afterrestimulation. As expected, OT-I Tc1 cells produced a largeamount of IFN-γ, whereas Tc9 cells released a minimal amountof IFN-γ but secreted a large amount of IL-9 (Fig. 2A). In ad-dition, these Tc9 cells also produced some IL-4, IL-10, and IL-17after in vitro restimulation (Fig. 2A and SI Appendix, Fig. S2A).In contrast, production of IL-9, IL-4, IL-10, or IL-17 from Tc1cells was undetectable, suggesting distinct expression patternsbetween Tc1 and Tc9 cells.To further test the cytokine expression profile of Tc9 cells, we

transferred CD45.2+ OT-I Tc1 or Tc9 cells into CD45.1 trans-genic mice followed by OVA peptide-pulsed dentritic cell (DC)vaccination and four daily doses of rhIL-2 to boost the antitumorresponses of ACT in vivo (20). CD45.2+CD8+ T cells were sortedfrom splenocytes 7 and 14 d after transfer, and cytokine pro-duction was measured after in vitro restimulation. Compared

with cells before transfer, Tc1 cells continued to produce similaramounts of IFN-γ without other cytokines examined. Noticeably,IL-9 production from Tc9 cells was decreased over time aftertransfer, and these cells were converted to IFN-γ–producing Tc1-like cells especially at day 14 after transfer (Fig. 2B). In addition,Tc9 cells kept IL-10 production in vivo, whereas IL-4 and IL-17secretion was reduced over time (Fig. 2B and SI Appendix, Fig.S2B). These results revealed that Tc9 cells might be unstable andflexible in cytokine production in vivo, and suggested that Tc9cells could gain the ability to produce IFN-γ and might furtherdifferentiate into fully cytolytic effector cells after transfer.

OVA-Specific Tc9 Cells Eradicate Large Established B16-OVAMelanoma. To assess whether the phenotypic differences be-tween Tc1 and Tc9 cells would translate into different efficacyafter transfer in vivo, we treated B16-OVA–bearing CD45.1-transgenic mice with equal numbers of adoptively transferredOT-I Tc1 or Tc9 cells, followed by DC vaccination and four dailydoses of rhIL-2. To mimic a clinically relevant scenario, tumorswere allowed to grow for 10 d before the treatment (SI Appendix,Fig. S3A). Surprisingly, only Tc9 cells mediated a significanttumor regression resulting in a complete cure and long-termsurvival, whereas Tc1 cell-treated mice relapsed 4 wk after T-cellinfusion despite the initial tumor shrinkage within the first 3 wk(Fig. 3A). To better understand the differences between Tc1 andTc9 cells in treatment outcomes, we analyzed spleens after T-cellinfusion for the presence of adoptively transferred effector cells.Flow cytometry analysis revealed that the percentages and ab-solute numbers of CD45.2+CD8+ splenocytes from Tc9 cell-treated mice were consistently higher than Tc1 cell-treatedmice over time (Fig. 3 B and C). Further evaluation by car-boxyfluorescein succinimidyl ester (CFSE) dilution assaydemonstrated that on the fourth day, CFSE intensity of Tc1 andTc9 cells in spleens was similar, suggesting that both cells pro-liferated well in the mice (Fig. 3D). In contrast, Tc1 cells re-covered on the fourth day had significantly higher percentagesof Annexin V+ apoptotic cells in spleens compared with Tc9counterparts (Fig. 3 E and F). These results suggested that the

Fig. 1. Tc9 cells produced IL-9 and were diverted from cytolytic differenti-ation. (A and B) Real-time PCR analysis of relative mRNA expression ofcytokines and cytolytic-related molecules (A) or transcription factors (B) inOT-I Tc1 and Tc9 cells. Expression relative to Gapdh is displayed. (C) T cellswere added at the indicated ratios to CFSEhi B16-OVA target cells or CFSElo

B16 nontarget cells in duplicate. Percent of specific lysis was determinedafter 8 h. Representative results from one of two performed experimentsare shown.

Fig. 2. Cytokine expression profile of Tc9 cells. (A) OT-I Tc1 or Tc9 cells wereprimed in polarized conditions and expanded with IL-2. The cells were thenrestimulated with splenocytes pulsed with OVA257–264 at indicated concen-trations for 24 h. Production of indicated cytokines was determined byELISA. (B) OT-I Tc1 or Tc9 cells (2 × 106) were adoptively transferred intoCD45.1-transgenic mice, followed by i.v. injection of 5 × 105 OVA257–264-pulsed DCs and i.p. injection of four doses of exogenous IL-2. CD45.2+

transferred cells were sorted from splenocytes at days 7 and 14. Day 0 rep-resents T cells before transfer. The cells were then restimulated with sple-nocytes pulsed with 0.01 μg/mL OVA257–264 in triplicate for 24 h. Productionof indicated cytokines was determined by ELISA. Representative results fromone of two repeated experiments are shown.

2266 | www.pnas.org/cgi/doi/10.1073/pnas.1317431111 Lu et al.

Dow

nloa

ded

by g

uest

on

Aug

ust 3

1, 2

020

Page 3: Tumor-specific IL-9 producing CD8 superior effector than ... · Tumor-specific IL-9–producing CD8+ Tc9 cells are superior effector than type-I cytotoxic Tc1 cells for adoptive immunotherapy

persistence of Tc9 cells, which might be one of the key reasons forthe rejection of established tumor after transfer, may be the resultof a survival advantage or resistance to apoptosis rather thanincreased proliferation of the cells.

Tc9 Cells Are Less Exhausted and Developed into Full Effector Cells inVivo. Driven by elevated levels of T-bet, Tc1 cells become ter-minally differentiated short-lived effector cells with KLRG-1high

and IL-7Rαlow phenotype (21). However, compared with Tc1counterparts, the tumor-specific Tc9 cells expressed significantlyhigher levels of IL-7Rα, a prosurvival cytokine receptor, suggesting

a possible mechanism of the increased persistence of these cells(Fig. 4A). Furthermore, Tc9 cells had significantly down-regu-lated expression of the exhaustion markers, such as KLRG-1,PD-1, LAG3, and 2B4 (Fig. 4B), demonstrating that Tc9 cellswere less exhausted T cells even upon repeated activation invivo. Reciprocally, Tc1 cells acquired a signature of terminaldifferentiation with high expression of exhaustion-phenotypicmarkers, leading to the failure of homeostatic proliferation,dysfunction, and apoptosis of these cells (Fig. 4B). As lessexhausted T cells, the long-term survival of Tc9 cells also allowedthem to convert to IFN-γ–positive Tc1-like cells especially 14 d

Fig. 3. OT-I Tc9 cells mediated enhanced antitumor response and displayed greater persistence. (A–C) Tc1 or Tc9 cells (2 × 106) were adoptively transferredinto CD45.1-transgenic mice bearing 10-d large established B16-OVA melanoma. DC vaccination and IL-2 were administered to some group of mice as in-dicated. (A) Tumor responses (n = 5) to adoptive transfer of Tc1 or Tc9 were shown. (B and C) Persistence of transferred Tc1 or Tc9 cells in the spleens oftreated tumor-bearing mice was analyzed by FACS. Numbers in histograms (B) represent the percentage of CD45.2+CD8+ OT-I T cells in splenocytes. (C) Totalnumber of CD45.2+CD8+ OT-I T cells was calculated from B. The spleens of three mice per condition were examined at each time point. (D) CFSE-labeled Tc1 orTc9 cells were transferred into tumor-bearing mice. Shown is CFSE dilution of gated CD45.2+CD8+ splenocytes 4 d after transfer. (E and F) Annexin V ex-pression was measured in Tc1 and Tc9 cells 4 d after transfer. (E) Numbers in histograms represent the percentage of Annexin V+ apoptotic Tc1 or Tc9 cells insplenocytes. Summarized (n = 3) percentages of apoptotic transferred cells were shown in F. Representative results from one of two repeated experiments areshown. **P < 0.01.

Fig. 4. Tc9 cells display less exhausted phenotypeand switch to Tc1-like cells in tumor-bearing mice.Tumor-bearing mice (n = 3) were transferred withOT-I Tc1 or Tc9 cells and treated the same as de-scribed in Fig. 1. Splenocytes were harvested andanalyzed. (A) Expression of IL-7Rα by transferredcells 7 d after transfer. (B) Expression of indicatedexhaustion markers by transferred cells 7 d aftertransfer. (C) FACS determination of intracellularcytokine production by Tc1 or Tc9 cells before andafter transfer. (D) Total number of IFN-γ–producingTc1 or Tc9 cells after transfer was calculated fromFACS analysis. (E) FACS determination of GrzB-pro-ducing Tc1 or Tc9 cells after transfer. (F) Totalnumber of GrzB-producing Tc1 or Tc9 cells aftertransfer was calculated from FACS analysis. Repre-sentative results from one of two performedexperiments are shown. *P < 0.05; **P < 0.01.

Lu et al. PNAS | February 11, 2014 | vol. 111 | no. 6 | 2267

IMMUNOLO

GY

Dow

nloa

ded

by g

uest

on

Aug

ust 3

1, 2

020

Page 4: Tumor-specific IL-9 producing CD8 superior effector than ... · Tumor-specific IL-9–producing CD8+ Tc9 cells are superior effector than type-I cytotoxic Tc1 cells for adoptive immunotherapy

after transfer, which was accompanied by a decrease of IL-9–producing CD8+ T cells (Fig. 4C). By calculating the totalnumber of IFN-γ–positive Tc1-like transferred cells, we foundthat Tc9 cell-transferred mice had already developed more thantwofold Tc1-like cells compared with those in Tc1 cell-trans-ferred mice 7 d after the transfer and this ratio kept increasingover time (Fig. 4D). Because production of IFN-γ is a quintes-sential characterization of cytolytic CD8+ T cells, we analyzedand calculated GrzB-producing CD8+ T cells from tumor-bear-ing mice. We enumerated significantly more GrzB-positive Tc9-derived cells than those in Tc1-cell transferred mice (Fig. 4 Eand F). These results collectively suggested that Tc9 cellscould evolve in vivo into distinct Tc1-like effector cells, whichmight be responsible for the Tc9 cell-mediated tumor destruction.

Cyclophosphamide Synergizes with Pmel-1–Derived Tc9 Cells ToMediate Enhanced Antitumor Immunity. Because OT-I cells targetartificial antigen, we next used the Pmel-1 model of adoptiveimmunotherapy, which reproduces the clinical challenge of tar-geting gp100 tumor/self-antigen in the poorly immunogenic B16melanoma (22). One day before T-cell adoptive transfer, micewere given one dose of cyclophosphamide (CTX; 250 mg/kg),which can induce lymphopenia, sensitize tumor cells to immunedestruction, and promote homeostatic proliferation of trans-ferred T cells (23, 24). Tc1 or Tc9 cells were transferred intomice bearing large established B16 melanoma in conjunctionwith DC vaccination and four daily doses of rhIL-2 (SI Appendix,Fig. S3B). Noticeably, Tc9-cell transfer mediated sustained an-titumor responses throughout the experiment, whereas Tc1 cellsonly induced temporary tumor regression, which was followed byrelapse of aggressive tumor growth (Fig. 5A). In addition, thedevelopment of autoimmune vitiligo was apparent 4 wk aftertransfer of Tc9 cells but was not observed in any of Tc1 cell-treated mice (Fig. 5B).This Pmel-1 Tc9 cell-mediated sustained antitumor response

was also associated with superbly improved in vivo expansion andpersistence of the transferred cells examined in the spleen of thetumor-bearing mice (Fig. 5C). Intracellular staining revealed

that, similar to OT-I Tc9 cells, transfer of Pmel-1 Tc9 cells alsodeveloped into large numbers of IFN-γ–positive Tc1-like cellsand GrzB-positive cytolytic effector cells (Fig. 5 D and E). Wefurther measured the cytokine production by transferred cellsisolated from the spleens and tumor tissues of tumor-bearingmice. ELISA results indicated that Tc1 cells maintained IFN-γand TNF-α production in the spleens, but the production ofthese cytokines was significantly decreased in tumor tissues (Fig.5F). In contrast, Tc9 cells gained the ability to produce IFN-γand TNF-α in vivo, and tumor-infiltrating Tc9 cells maintainedthe production of these cytokines compared with Tc9 cells in thespleens. Notably, only Tc9 cells produced significant amounts ofIL-9 and IL-2, which indicated a less differentiated phenotype ofTc9 cells (25). By comparing the cytotoxicity of these sorted cells,we found that tumor-infiltrating Tc1 and Tc9 cells were similar intheir ability to lyse target tumor cells, although splenic Tc1 cellshad slightly higher cytotoxicity than Tc9 cells (Fig. 5G). Collec-tively, Pmel-1 Tc9-cell transfer could confer effective antitumorresponse against large B16 melanoma, and the failure of Tc1cells to control the disease might be due to the inability of thesecells to expand and persist despite the higher cytotoxicity andability to secrete IFN-γ in vitro and in vivo.

Therapeutic Effect of Tc9 Cells Critically Depends on IL-9. BecauseTc9 cells acquired the ability to secrete IFN-γ in tumor-bearingmice, we next determined the importance of Tc9-derived IFN-γand IL-9 in mediating tumor rejection in MC38-gp100 tumormodel. In this model, transfer of Pmel-1 Tc9 cells could mediatesignificantly enhanced antitumor response than that of Pmel-1Tc1 or naïve CD8+ T cells, which was associated with a superiorpersistence of Tc9 cells in recipient mouse spleens (SI Appendix,Fig. S4). We treated MC38-gp100 tumor-bearing mice with iso-type controls, IL-9–neutralizing antibodies or IFN-γ–neutralizingantibodies and subsequently transferred them with Pmel-1 Tc9cells. Unexpectedly, tumor rejection was abrogated only by anti–IL-9 treatment, whereas neutralizing IFN-γ did not reach sta-tistical significance compared with isotype control (Fig. 6A). Bycalculating the absolute numbers of splenic Thy1.1+CD8+ cells in

Fig. 5. CTX synergizes with Pmel-1 Tc9 cells to controllarge established B16 melanoma in vivo. Pmel-1 Tc1 orTc9 cells were primed in polarized conditions and ex-panded with IL-2. Tc1 or Tc9 cells (2 × 106) were adop-tively transferred into C57BL/6 mice bearing 10-d largeestablished B16melanoma. One dose of CTXwas given1 d before T-cell transfer. DC vaccination and IL-2 wereadministered to some group of mice after T-cell trans-fer. (A) Tumor responses (n = 5) to adoptive transfer ofTc1 or Tc9were shown. (B) Representative autoimmunevitiligo of tumor-bearingmice 25 d after T-cell transfer.Arrows indicated thepresenceof vitiligo. (C) Persistenceof transferred Tc1 or Tc9 cells in the spleens of treatedtumor-bearing mice was analyzed by FACS. (D and E)Total numbers of IFN-γ–producing (D) or GrzB-pro-ducing (E) Thy1.1+CD8+ cells after transfer were calcu-lated from FACS analysis. (F) Transferred Tc1 or Tc9 cellswere sorted from the spleens or tumor tissues at day 14after transfer. The cells were then restimulated withsplenocytespulsedwith0.01μg/mLhgp10025–33peptidein triplicate for 24 h. Production of indicated cytokineswas determined by ELISA. BT represents cells beforetransfer. (G). Transferred Tc1 or Tc9 cells were sortedfrom the spleens or tumor tissues at day 14 aftertransfer. Cytolytic function of T cells was tested by invitro cytotoxicity assayat 10 to1effector to target ratioswith CFSEhi B16 target cells and CFSElo MC38 nontargetcells in duplicate. Percentage of specific lysis was de-termined overnight. Representative results from one oftwo performed experiments are shown. **P < 0.01.

2268 | www.pnas.org/cgi/doi/10.1073/pnas.1317431111 Lu et al.

Dow

nloa

ded

by g

uest

on

Aug

ust 3

1, 2

020

Page 5: Tumor-specific IL-9 producing CD8 superior effector than ... · Tumor-specific IL-9–producing CD8+ Tc9 cells are superior effector than type-I cytotoxic Tc1 cells for adoptive immunotherapy

treated mice, we found that IL-9 neutralization did not impairthe persistence of Tc9 cells or their ability to produce IFN-γ andGrzB (SI Appendix, Fig. S5). However, in IL-9–neutralized mice,the number of tumor-infiltrating Thy1.1+CD8+ T cells weresubstantially reduced compared with those in mice receivingisotype control or anti–IFN-γ antibodies. This impaired Thy1.1+

CD8+ T-cell infiltration could also be demonstrated by thesharply decreased numbers of IFN-γ−producing and GrzB-pro-ducing Thy1.1+CD8+ cells recovered from tumor sites (Fig. 6 Band C). On the other side, anti–IFN-γ treatment only ablated theproduction of IFN-γ from transferred Tc9 cells, whereas thehoming of cytolytic Tc9 cells to tumor tissues was not affected(Fig. 6 B and C). In addition, by examining the leukocyte subsetsin tumor microenvironment, we also observed significantly in-creased IL-9–dependent tumor-infiltrating host Thy1.1–CD8+ Tcells in Tc9 cell-transferred mice (SI Appendix, Fig. S6). BecauseTc9-cell transfer mediated a sustained antitumor response inC57BL/6 Rag-1−/− mice similar to that in wild-type mice, hostCTL responses may have contributed, but were not required, forthe antitumor efficacy of Tc9 cells in vivo (SI Appendix, Fig. S7).Taken all together, these results thus far suggested that Tc9 cellsmay kill tumor cells independent of their secreted IFN-γ, pos-sibly by using the cytolytic enzymes, and IL-9 provided criticalhelp to their migration into tumor sites to exert effector function,which is an effect of IL-9 that has been extensively elucidated inour previous studies (15, 17).

DiscussionCD8+ CTLs are thought to play a crucial role in tumor rejection,and extensive focus has been devoted to the study of CD8+ Tcells in adoptive transfer protocols. Nevertheless, complete anddurable tumor regression or cure rates remains to be archived(2). In the current study, we identified unique IL-9–skewedCD8+ T cells, termed Tc9 cells, by priming with Th9-polarizedcondition. Apart from the differences in cytokine secretion, Tc9cells differed from Tc1 cells in that they were less cytotoxic invitro. In line with our observation, a recent publication alsoconfirmed the existence of a Tc9 cell subset in both culturedsystem and in vivo (26). However, the role of this CD8+ T-cellsubset has not been tested in cancer immunotherapy settings. Inthis study, we evaluated the efficacy of Tc9 cell transfer in bothOT-I/B16-OVA and Pmel-1/B16 mouse models in comparisonwith the classic Tc1 cells. Our study demonstrated that transfer

of Tc9 cells displayed superior efficacy to mediate regression oflarge established tumors by converting to IFN-γ–producing cy-tolytic effector cells in vivo. These findings are highly relevantto the improvement of CD8+ T-cell–based adoptive cancerimmunotherapy.IL-2 can augment antigen-drive acquisition of CD8+ effector

T-cell phenotype with cytolytic function, and therefore, thegeneration of these tumor-specific Tc1 cells ex vivo involvesusing high levels of IL-2 in current clinic ACT protocols (27).However, ACT has met with only modest success in humans,possibly due to the fact that IL-2–induced T-bethigh Tc1 cellsdisplay end-effector features and short lifespan (21). It is in-creasingly evident that ACT can be improved by the transfer ofless-differentiated CD8+ T cells that possess greater persistencepotential (28). We found that tumor-specific Tc9 cells couldmediate superior antitumor responses that were associated withsuperb persistence of the transferred cells. Compared with Tc1cells, Tc9 cells did not display full-mature phenotype as de-termined by reduced expression of IFN-γ and cytotoxic effectormolecules, but instead acquired a signature of “younger” phe-notypes by IL-2–producing, KLRG-1low and IL-7Rαhigh, whichare characteristic for long-term lived cells with capacity ofself-renewal.Our results also revealed that the robust persistence of Tc9

cells resulted from a survival advantage of resistance to apoptosisrather than increased proliferation. It is possible that TGF-β1from Th9-conditioned medium contributed to the arrest of ef-fector differentiation of Tc9 cells and the conversion of apo-ptotic stimuli into the signal for IL-9 production (29). Incontrast, IL-2–primed Tc1 cells have increased susceptibility toapoptosis under repeated activation in vivo (30). In addition topersistence potential, several studies have indicated that effec-tive cells for ACT must possess the capacity to acquire/maintaineffector function after transfer (25). Although TGF-β1 fromTh9-conditioned medium may play a crucial role of rapid ex-pansion of CD8+ T cells (31), it is insufficient because CD8+ Tcells primed with TGF-β1 alone display immunosuppressiveregulatory CD8+ T-cell phenotype. Noticeably, Tc9 cells couldefficiently switch to Tc1-like cells with the production of IFN-γ,which is well established as a key factor linked to tumor rejectionand is crucial for cytolytic function of CD8+ T cells (32). In fact,we enumerated more than twofold Tc1-like cells in Tc9 cell-treated mice as early as 7 d after transfer, and 25∼30-fold of

Fig. 6. IL-9 contributes to Tc9 cell-mediated tumor rejection. Pmel-1 Tc9 cells (2 × 106) were adoptively transferred into C57BL/6 mice bearing 10-d largeestablished MC38-gp100 tumor. One dose of CTX was given 1 d before T-cell transfer. DC vaccination and IL-2 were administered to the mice that receivedT-cell transfer. mAbs neutralizing IL-9 or IFN-γ or control IgG were i.p. injected to mice as indicated. (A) Tumor responses (n = 5) to adoptive transfer of Tc9 andantibody treatment were shown. (B) FACS determination of the percentage of tumor-infiltrating, adoptively transferred Thy1.1+CD8+ T cells, IFN-γ–producingor GrzB-producing tumor-infiltrating, adoptively transferred Thy1.1+CD8+ T cells in the leukocyte fraction. Tumor tissues were harvested 3 wk after transfer.(C) Total number of tumor-infiltrating, IFN-γ–producing or GrzB-producing Thy1.1+CD8+ T cells 3 wk after transfer was calculated from FACS analysis. Cellnumber was normalized to 500-mg tumor tissues. Representative results from one of two performed experiments are shown. *P < 0.05; **P < 0.01.

Lu et al. PNAS | February 11, 2014 | vol. 111 | no. 6 | 2269

IMMUNOLO

GY

Dow

nloa

ded

by g

uest

on

Aug

ust 3

1, 2

020

Page 6: Tumor-specific IL-9 producing CD8 superior effector than ... · Tumor-specific IL-9–producing CD8+ Tc9 cells are superior effector than type-I cytotoxic Tc1 cells for adoptive immunotherapy

these cells after 3 wk compared with Tc1-cell transferred mice.Further evaluation revealed that anti–IFN-γ treatment did notsignificantly influence the protective effect against tumor, whichwas associated with little impact on the number and function oftransferred Tc9 cells in both spleens and tumor tissues. In-terestingly, when mice were treated with IL-9–neutralizingantibodies, Tc9 cells largely failed to migrate into tumor tissuesto exert long-lasting antitumor therapeutic effect. Given theimportant role of IL-9 in provoking inflammation in tumor sitesand recruitment of lymphocytes (15, 17), our results suggestedthat IL-9 secreted by Tc9 cells played an indispensible role forthe homing of these cytolytic Tc1-like cells to tumor tissues andkilling of tumor cells. These findings thus indicated that IL-9rather than IFN-γ might be the critical cytokine contributed tothe tumor destruction mediated by Tc9 cells.Furthermore, it is well known that tumor-derived factors could

provide the necessary signals/conditions for effector T cells tobecome functionally exhausted in tumor microenvironment (33).Upon adoptive transfer, splenic Tc1 cells displayed the charac-teristic features of exhausted CD8+ T cells by up-regulated ex-pression of KLRG-1, PD-1, LAG3, and 2B4 (34), whereas Tc9cells were less exhausted. Compared with splenic Tc1 cells, tu-mor-infiltrating Tc1 cells demonstrated significantly reducedcytolytic function as well as reduced ability to produce IFN-γ andTNF-α. Interestingly, tumor-infiltrating Tc9 cells maintainedtheir capacity to produce IL-2, IFN-γ, and TNF-α and killedefficiently tumor cells compared with their counterparts in thespleens. This promising aspect of Tc9 cells is possibly, in part,

owing to the extremely low surface expression of PD-1, a keyT-cell exhaustion/inhibition pathway (35), thus rescuing T-celleffector functions of Tc9 cells in vivo.Overall, our study highlights the clinical potential of tumor-

reactive Tc9 cells for adoptive cancer immunotherapy. With theability to confer tumor specificity by genetic engineering, selec-tion of optimal T-cell subsets with enhanced antitumor potencyis of great importance. As less-exhausted younger cells, the suf-ficient lineage plasticity of Tc9 cells allows these cells sub-sequently to differentiate into long-lasting IFN-γ–producing Tc1-like effector cells upon transfer. The ability of tumor-reactiveTc9 cells to confer sustained antitumor responses against largeestablished tumors might open an avenue to overcome thechallenges encountered by current ACT protocols.

Materials and MethodsAll mice were maintained in specific pathogen-free conditions, and animalstudy was approved by the Institutional Animal Care and Use Committee ofCleveland Clinic Foundation. The procedures of Tc1 and Tc9 cell differenti-ation, tumor models and adoptive transfer are described in SI Appendix,Materials and Methods. The detailed quantitative real-time PCR, flowcytometry, CFSE labeling, and cytotoxicity assay are also explained inSI Appendix, Materials and Methods.

ACKNOWLEDGMENTS. This work was supported by a startup fund fromCleveland Clinic, National Cancer Institute Grants R01 CA138402, R01CA138398, R01 CA163881, and P50 CA142509, the Leukemia and LymphomaSociety, Multiple Myeloma Research Foundation, and Commonwealth Founda-tion for Cancer Research.

1. Restifo NP, Dudley ME, Rosenberg SA (2012) Adoptive immunotherapy for cancer:Harnessing the T cell response. Nat Rev Immunol 12(4):269–281.

2. Rosenberg SA, Dudley ME (2009) Adoptive cell therapy for the treatment of patientswith metastatic melanoma. Curr Opin Immunol 21(2):233–240.

3. Rosenberg SA, et al. (2011) Durable complete responses in heavily pretreated patientswith metastatic melanoma using T-cell transfer immunotherapy. Clin Cancer Res17(13):4550–4557.

4. Hinrichs CS, et al. (2009) Adoptively transferred effector cells derived from naiverather than central memory CD8+ T cells mediate superior antitumor immunity. ProcNatl Acad Sci USA 106(41):17469–17474.

5. Hinrichs CS, et al. (2011) Human effector CD8+ T cells derived from naive rather thanmemory subsets possess superior traits for adoptive immunotherapy. Blood 117(3):808–814.

6. Gattinoni L, et al. (2005) Removal of homeostatic cytokine sinks by lymphodepletionenhances the efficacy of adoptively transferred tumor-specific CD8+ T cells. J Exp Med202(7):907–912.

7. Hinrichs CS, et al. (2008) IL-2 and IL-21 confer opposing differentiation programs toCD8+ T cells for adoptive immunotherapy. Blood 111(11):5326–5333.

8. Hinrichs CS, et al. (2009) Type 17 CD8+ T cells display enhanced antitumor immunity.Blood 114(3):596–599.

9. Shrikant PA, et al. (2010) Regulating functional cell fates in CD8 T cells. Immunol Res46(1-3):12–22.

10. Ye Z, et al. (2007) Type 1 CD8+ T cells are superior to type 2 CD8+ T cells in tumorimmunotherapy due to their efficient cytotoxicity, prolonged survival and type 1immune modulation. Cell Mol Immunol 4(4):277–285.

11. Yu Y, et al. (2013) Adoptive transfer of Tc1 or Tc17 cells elicits antitumor immunityagainst established melanoma through distinct mechanisms. J Immunol 190(4):1873–1881.

12. Garcia-Hernandez MdeL, et al. (2010) Adoptive transfer of tumor-specific Tc17 ef-fector T cells controls the growth of B16 melanoma in mice. J Immunol 184(8):4215–4227.

13. Wang L, et al. (2009) IL-17 can promote tumor growth through an IL-6-Stat3 signalingpathway. J Exp Med 206(7):1457–1464.

14. Goswami R, Kaplan MH (2011) A brief history of IL-9. J Immunol 186(6):3283–3288.15. Lu Y, et al. (2012) Th9 cells promote antitumor immune responses in vivo. J Clin Invest

122(11):4160–4171.16. Purwar R, et al. (2012) Robust tumor immunity to melanoma mediated by interleukin-

9-producing T cells. Nat Med 18(8):1248–1253.17. Lu Y, Yi Q (2013) Utilizing TH9 cells as a novel therapeutic strategy for malignancies.

OncoImmunology 2(3):e23084.18. Intlekofer AM, et al. (2005) Effector and memory CD8+ T cell fate coupled by T-bet

and eomesodermin. Nat Immunol 6(12):1236–1244.

19. Kaplan MH (2013) Th9 cells: Differentiation and disease. Immunol Rev 252(1):

104–115.20. Lou Y, et al. (2004) Dendritic cells strongly boost the antitumor activity of adoptively

transferred T cells in vivo. Cancer Res 64(18):6783–6790.21. Joshi NS, et al. (2007) Inflammation directs memory precursor and short-lived effector

CD8(+) T cell fates via the graded expression of T-bet transcription factor. Immunity

27(2):281–295.22. Overwijk WW, et al. (2003) Tumor regression and autoimmunity after reversal of

a functionally tolerant state of self-reactive CD8+ T cells. J Exp Med 198(4):569–580.23. North RJ (1982) Cyclophosphamide-facilitated adoptive immunotherapy of an es-

tablished tumor depends on elimination of tumor-induced suppressor T cells. J Exp

Med 155(4):1063–1074.24. van der Most RG, et al. (2009) Cyclophosphamide chemotherapy sensitizes tumor cells

to TRAIL-dependent CD8 T cell-mediated immune attack resulting in suppression of

tumor growth. PLoS ONE 4(9):e6982.25. Hinrichs CS, Gattinoni L, Restifo NP (2006) Programming CD8+ T cells for effective

immunotherapy. Curr Opin Immunol 18(3):363–370.26. Visekruna A, et al. (2013) Tc9 cells, a new subset of CD8(+) T cells, support Th2-

mediated airway inflammation. Eur J Immunol 43(3):606–618.27. June CH (2007) Adoptive T cell therapy for cancer in the clinic. J Clin Invest 117(6):

1466–1476.28. Gattinoni L, et al. (2005) Acquisition of full effector function in vitro paradoxically

impairs the in vivo antitumor efficacy of adoptively transferred CD8+ T cells. J Clin

Invest 115(6):1616–1626.29. Takami M, Love RB, Iwashima M (2012) TGF-β converts apoptotic stimuli into the

signal for Th9 differentiation. J Immunol 188(9):4369–4375.30. Waldmann TA (2006) The biology of interleukin-2 and interleukin-15: Implications for

cancer therapy and vaccine design. Nat Rev Immunol 6(8):595–601.31. Liu S, et al. (2010) TGF-beta1 induces preferential rapid expansion and persistence of

tumor antigen-specific CD8+ T cells for adoptive immunotherapy. J Immunother

33(4):371–381.32. Böhm W, et al. (1998) T cell-mediated, IFN-gamma-facilitated rejection of murine B16

melanomas. J Immunol 161(2):897–908.33. Baitsch L, et al. (2011) Exhaustion of tumor-specific CD8+ T cells in metastates from

melanoma patients. J Clin Invest 121(6):2350–2360.34. Crespo J, Sun H, Welling TH, Tian Z, Zou W (2013) T cell anergy, exhaustion, senes-

cence, and stemness in the tumor microenvironment. Curr Opin Immunol 25(2):

214–221.35. Topalian SL, et al. (2012) Safety, activity, and immune correlates of anti-PD-1 antibody

in cancer. N Engl J Med 366(26):2443–2454.

2270 | www.pnas.org/cgi/doi/10.1073/pnas.1317431111 Lu et al.

Dow

nloa

ded

by g

uest

on

Aug

ust 3

1, 2

020