Author Manuscript Published OnlineFirst on August 1, 2019; DOI: 10.1158/1078-0432.CCR-19-1259 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

1

Anti-OX40 antibody directly enhances the function of tumor-reactive CD8+ T cells

and synergizes with PI3Kβ inhibition in PTEN loss melanoma

Weiyi Peng1,5*, Leila J. Williams1, Chunyu Xu1,5, Brenda Melendez1, Jodi A.

McKenzie1,6, Yuan Chen1, Heather Jackson2, Kui S. Voo3, Rina M. Mbofung1,7,, Sara E.

Leahey1, Jian Wang4, Greg Lizee1, Hussein A. Tawbi1, Michael A. Davies1, Axel Hoos2,

James Smothers2, Roopa Srinivasan2, Elaine Paul2, Niranjan Yanamandra2* and Patrick

Hwu1*

1Department of Melanoma Medical Oncology, The University of Texas MD Anderson

Cancer Center, Houston, TX.

2Oncology R&D, Immuno-Oncology and Combinations RU, GlaxoSmithKline, 1250 S.

Collegeville Rd, Collegeville, PA 19426, United States

3Oncology Research for Biologics and Immunotherapy Translation Platform, The

University of Texas MD Anderson Cancer Center, Houston, TX.

4Department of Biostatistics, The University of Texas MD Anderson Cancer Center,

Houston, TX.

5Present address: Department of Biology and Biochemistry, University of Houston,

Houston, TX.

6Present address: Eisai Inc., Woodcliff Lake, NJ.

7Present address: Merck Research Laboratories, Palo Alto, CA.

Running Title: OX40 agonist-based cancer immunotherapy

Keywords: OX40, PI3K, cancer immunotherapy

Downloaded from clincancerres.aacrjournals.org on September 25, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on August 1, 2019; DOI: 10.1158/1078-0432.CCR-19-1259 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

2

*Corresponding Authors: Patrick Hwu, The University of Texas MD Anderson Cancer

Center, 1515 Holcombe Boulevard, Houston, TX 77030. Phone: 713-563-1728; Fax:

713-745-1046; Email: [email protected]

Niranjan Yanamandra, Immuno-Oncology and Combinations RU, GlaxoSmithKline,

1250 s. Collegeville Rd, Collegeville PA, 19426, Phone: 610-917-5123; Email:

[email protected]

Weiyi Peng, The University of Houston, 3517 Cullen Blvd, Houston, TX, 77204. Phone:

713-743-6941; Fax: 713-743-3415; Email: [email protected]

Conflict of Interest:

The authors of this publication have research support from GlaxoSmithKline (GSK). The

terms of this agreement have been reviewed and approved by the University of Texas

MD Anderson Cancer Center (MDACC) in accordance with its policy on objectivity in

research. W. Peng received honoraria and travel support from Bristol-Myers Squibb

(BMS). H. Jackson, A. Hoos, J. Smothers, R. Srinivasan, E. Paul and N. Yanamandra are

full-time employees of GSK. P. Hwu is a consultant/an advisory board member for

Immatics, Dragonfly, Sanofi, and GSK. M.A. Davies is an advisory board member for

BMS, GSK, Novartis, Roche/Genentech, Array, Sanofi, and Vaccinex. M.A. Davies is

also the PI of research funding to MDACC from GSK, AstraZeneca, Roche/Genentech,

Myriad, Oncothyreon, and Sanofi-Aventis. No potential conflicts of interest were

disclosed by other authors.

Grant Support

Downloaded from clincancerres.aacrjournals.org on September 25, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on August 1, 2019; DOI: 10.1158/1078-0432.CCR-19-1259 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

3

This work was supported in part by the following National Cancer Institute grants:

R01CA187076 (PH&MD), P50CA093459 (UT M.D. Anderson Cancer Center SPORE in

Melanoma), T32CA009666-21(MD) and P30CA016672 (UT MDACC CCSG for the

Flow Cytometry & Cellular Imaging facility), by philanthropic contributions to MDACC

Melanoma Moon Shots Program; Melanoma Research Alliance Young Investigator

Award (WP, 558998); Dr. Miriam and Sheldon G. Adelson Medical Research

Foundation; Aim at Melanoma Foundation, Miriam and Jim Mulva Research Fund; and

by Cancer Prevention and Research Institute of Texas (PH, RP170401; JAM, RP140106

and RP170067).

Downloaded from clincancerres.aacrjournals.org on September 25, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on August 1, 2019; DOI: 10.1158/1078-0432.CCR-19-1259 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

4

ABSTRACT

Purpose: OX40 agonist-based combinations are emerging as a novel avenue to improve

the effectiveness of cancer immunotherapy. To better guide its clinical development, we

characterized the role of the OX40 pathway in tumor-reactive immune cells. We also

evaluated combining OX40 agonists with targeted therapy to combat resistance to cancer

immunotherapy.

Experimental Design: We utilized patient-derived tumor infiltrating (TILs)

and multiple preclinical models to determine the direct effect of anti-OX40 agonistic

antibodies on tumor-reactive CD8+ T cells. We also evaluated the antitumor activity of an

anti-OX40 antibody plus PI3K inhibition in a transgenic murine melanoma model

(Braf-mutant, PTEN null), which spontaneously develops immunotherapy-resistant

melanomas.

Results: We observed elevated expression of OX40 in tumor reactive CD8+ TILs upon

encountering tumors; activation of OX40 signaling enhanced their cytotoxic function.

OX40 agonist antibody improved the antitumor activity of CD8+ T cells and the

generation of tumor-specific T cell memory in vivo. Furthermore, combining anti-OX40

with GSK2636771, a PI3K selective inhibitor, delayed tumor growth and extended the

survival of mice with PTEN-null melanomas. This combination treatment did not

increase the number of TILs, but it instead significantly enhanced proliferation of CD8+

TILs and elevated the serum levels of CCL4, CXCL10, and IFN-γ, which are mainly

produced by memory and/or effector T cells.

Downloaded from clincancerres.aacrjournals.org on September 25, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on August 1, 2019; DOI: 10.1158/1078-0432.CCR-19-1259 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

5

Conclusion: These results highlight a critical role of OX40 activation in potentiating the

effector function of tumor-reactive CD8+ T cells and suggest further evaluation of OX40

agonist-based combinations in patients with immune-resistant tumors.

Translational Relevance:

Cancer immunotherapy is revolutionizing cancer treatment. However, most patients still

fail to respond to currently available immunomodulatory agents. Thus, there remains a

critical need to identify novel immunoregulatory targets and rational combinatorial

strategies to induce robust and durable antitumor immune responses. Here, we used

patient samples and clinically relevant animal models to evaluate the immunological and

antitumor effects of OX40 agonist-based immunotherapy. Our results add to the growing

body of evidence that OX40 agonists can boost antitumor immune responses by

modulating T cell effector function and tumor-specific memory. Our results also identify

a novel therapeutic strategy of combining OX40 agonist antibodies with targeted therapy

in cancer patients, particularly those with tumors with loss of the tumor suppressor

PTEN.

Downloaded from clincancerres.aacrjournals.org on September 25, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on August 1, 2019; DOI: 10.1158/1078-0432.CCR-19-1259 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

6

INTRODUCTION

Several immunomodulatory agents that target T cell co-inhibitory receptors, such

as PD-1 and CTLA-4, have been developed to boost T cell-mediated antitumor immune

responses in cancer patients. These immunotherapies have demonstrated durable clinical

benefit in many types of cancer, and immune checkpoint blockade has become a standard

front-line treatment in multiple solid cancers, including melanoma, cancer, bladder

cancer, and kidney cancer (1,2). This new clinical paradigm has shifted research efforts in

tumor immunology to prioritize the identification of additional immunoregulatory targets

and rational combinatorial treatments to further increase the rate of potent and durable

antitumor immune responses.

T cell activation is tightly regulated by two sets of signals via T cell receptors

(TCR) and T cell co-signaling receptors. Positive (co-stimulatory) and negative (co-

inhibitory) signals from T cell co-signaling receptors direct T cell function in response to

TCR stimulation. Several studies have demonstrated that activating T cell co-stimulatory

receptors, such as OX40 and 4-1BB, can facilitate T cell-mediated antitumor immunity

(3,4). Moreover, disrupting T cell co-inhibitory signaling pathways, such as PD-1 and

CTLA-4, has been reported to reinvigorate tumor-reactive T cells and stem tumor

development in patients with a variety of tumors (5). However, a durable and effective

antitumor immune response only can be achieved in a small percentage of cancer patients

treated with immune checkpoint blockade (ICB) (6). One mechanism of primary

resistance to ICB is insufficient tumor-reactive T cells in patients with non-immunogenic

tumors (7). Under the notion that activation of T cell co-stimulatory signaling pathways

can augment the generation of effector and memory T cells (8), more studies are focused

Downloaded from clincancerres.aacrjournals.org on September 25, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on August 1, 2019; DOI: 10.1158/1078-0432.CCR-19-1259 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

7

on targeting T cell co-stimulatory receptors to overcome primary resistance to ICB

therapy in cancer patients. One such prominent T cell co-stimulatory molecule is OX40.

Indeed, early phase clinical trials evaluating agonist antibodies targeting the OX40

pathway alone or in combination with ICB in cancer patients are ongoing, such as

NCT02221960 (formerly of MedImmune), NCT02528357 (GlaxoSmithKline) and

NCT02554812 (Pfizer). While these trials have begun, an improved understanding of the

impact of OX40 activation on immune effector cells may help to optimize the clinical

evaluation of OX40-based immunotherapy and develop novel combinatorial approaches

to treat cancer patients with primary resistance to ICB.

Here, by utilizing melanoma patient-derived cell lines and multiple preclinical

models, we sought to determine the role of the OX40 pathway in regulating the effector

function of tumor-reactive T cells and evaluate the therapeutic potential of combining

OX40 agonist antibody with cancer targeted therapy. Our results describe the value of an

OX40 agonist antibody to augment T cell-mediated antitumor response by directly

enhancing proliferation and cytotoxicity of CD8+ tumor-reactive T cells. This study also

provides critical rationale for the clinical evaluation of the combination of an OX40

agonist antibody and a selective PI3K inhibitor in patients with immunoresistant PTEN

loss tumors.

Material and Methods

Cell lines and Mice

Human Mel2400, Mel2549, and their autologous TILs were established from

metastatic melanoma patients enrolled in the adoptive T cell therapy (ACT) trial at MD

Anderson Cancer Center (MDACC) as previously described (9). The murine

Downloaded from clincancerres.aacrjournals.org on September 25, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on August 1, 2019; DOI: 10.1158/1078-0432.CCR-19-1259 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

8

MC38/gp100 cell line was generated in our previous study (10). All tumor cell lines were

maintained in RPMI 1640 complete medium supplemented with 10% heat-inactivated

fetal bovine serum (Atlanta Biologicals) and normocin (InvivoGen). TIL cell lines were

maintained in RPMI 1640 with 10% human type AB serum (GEMINI), 3000 U/ml IL-2

(Prometheus Laboratories), and normocin. Cells were routinely monitored for

mycoplasma contamination by using the MycoAlert kit (Lonza). Short tandem repeat

(STR) profiling was used to confirm the identity of patient-derived cell lines. The

maximum length of time of in vitro cell culture between thawing and use in the described

experiments was two weeks.

C57BL/6 mice and C57BL/6 albino mice were purchased from Charles River

Frederick Research Model Facility. Tyr:CreER; PTENlox/lox; BRAF V600E/+ (BP) mice bred

onto a C57BL/6 background were kindly provided by Dr. Bosenberg (Yale University

School of Medicine). Pmel-1 TCR/Thy1.1 mice were from in-house breeding colonies.

All mice were maintained in a specific pathogen-free barrier facility at MDACC. All

studies were conducted in accordance with the MDACC and GlaxoSmithKline (GSK)

Policy on the Care, Welfare and Treatment of Laboratory Animals. All animal

experiment protocols were reviewed by the Institutional Animal Care and Use Committee

either at GSK or at MDACC, the institution where the work was performed.

Caspase-3 based T cell killing assay

Patient-derived tumor cells were labelled with DDAO dye (Thermo Fisher)

according to the manufacturer’s instructions. Peripheral blood mononuclear cells

(PBMCs) from healthy donors were isolated from buffy coats (Gulf Coast Regional

Blood Center) and irradiated with 5000 rad of gamma-radiation. Irradiated PBMCs were

Downloaded from clincancerres.aacrjournals.org on September 25, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on August 1, 2019; DOI: 10.1158/1078-0432.CCR-19-1259 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

9

washed with PBS and then incubated with 10 g/ml full length or Fc-fragment deleted

anti-human OX40 (GSK3174998, GlaxoSmithKline) at 37°C for 1 hour. Antibody-pulsed

PBMCs were mixed with DDAO-labelled tumor cells and autologous TILs at 37°C for an

additional 3 hours. The ratio of T cells to PBMCs used in this assay was 1:1. To evaluate

the effect of the activation of the OX40 pathway in murine CD8+ T cells, we cross-linked

anti-mouse OX40 antibody by pretreating bone marrow derived dendritic cells (DCs)

from C57BL/6 mice with 10 µg/ml anti-murine OX40 antibody at 37°C for 1 hour. After

washing with PBS, antibody-pulsed DCs were mixed with gp100-specific CD8+ Pmel-1

T cells and MC38/gp100 tumor cells for an additional 3 hours. The ratio of DCs to T cells

was 1:1. The cell mixtures were then permeabilized with Fix/Perm solution (BD

Biosciences) for 20 min at room temperature and stained with a PE-conjugated anti-

cleaved caspase-3 monoclonal antibody (BD Biosciences) as previously described (11).

Samples were analyzed using a FACSCanto II (BD Biosciences). The percentage of

cleaved caspase-3+ tumor cells was calculated and used to determine the extent of T-cell

induced tumor apoptosis.

Retroviral transduction of pmel-1 T cells

Full-length human OX40 was amplified and cloned into a retroviral vector,

pMXs-IG, which was kindly provided by Dr. Kitamura (University of Tokyo, Japan)(12).

The retroviral vector expressing an enhanced firefly luciferase was generated in our

previous study (13). Retroviral vectors and the packaging vectors were transiently co-

transfected into the packaging cell line, Plate-E, using Lipofectamine 2000 (Invitrogen).

Supernatants containing viral particles were used to infect pre-activated splenocytes from

pmel-1 mice as previously described (10). Three days after transduction, transduced

Downloaded from clincancerres.aacrjournals.org on September 25, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on August 1, 2019; DOI: 10.1158/1078-0432.CCR-19-1259 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

10

pmel-1 T cells were sorted using a FACSAria (BD Biosciences) based on the expression

of appropriate reporter genes embedded in the expression vectors.

Tumor and vaccination models

To determine the in vivo effect of targeting OX40 on the function of tumor-

reactive CD8+ T cells, luciferase-expressing pmel-1 T cells were transferred into

C57BL/6 albino mice bearing MC38/gp100 tumor as previously described (10). One

hundred micrograms of anti-mouse OX40 (OX-86, BioXcell) or mouse anti-human

OX40 (Kindly provided Dr. Voo) (14) was intraperitoneally administered to tumor-

bearing mice (twice per week). Tumor size was monitored every two days, and in vivo

bioluminescence imaging analyses were performed by using an IVIS 200 system

(Xenogen) on day 6 after T cell transfer.

To evaluate the antitumor activity of anti-OX40 alone and in combination with

PI3K inhibition, Tyr:CreER; PTENlox/lox; BRAF V600E/+ mice (BP mice, 6-8 weeks of

age) were treated with 4-hydroxytamoxifen to induce tumor formation. Tumor-bearing

mice were randomized into four groups to receive anti-OX40 and/or GSK2636771.

GSK2636771 (GlaxoSmithKline) was suspended in 1% (w/v) methylcellulose and

administered to mice daily by oral gavage at a dose of 30 mg/kg. Anti-OX40 (OX-86,

BioXcell) was administered at a dose of 50 g/per mouse. The relevant solvent and

control rat IgG antibody (Sigma) were administered to animals in the control group.

Because our previous study showed that CD40 agonistic antibody can promote in

vivo proliferation and activation of T cells (15), when we evaluated the in vivo effect of

anti-OX40 monotherapy and in combination with selective PI3K inhibition on antigen-

specific T cells, we re-designed our vaccine model to eliminate the possible confounding

Downloaded from clincancerres.aacrjournals.org on September 25, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on August 1, 2019; DOI: 10.1158/1078-0432.CCR-19-1259 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

11

effects of a CD40 antibody-containing immunoadjuvant on the antitumor activity of an

OX40 agonist (11,16). Briefly, C57BL/6 mice received 1,000 naive pmel-1 T cells

intravenously (i.v.) and were vaccinated with two distinct subcutaneous (s.c.) injections

in each flank with 100 µl of saline containing 100 µg of human gp10025–33. In addition,

vaccinated mice received 100,000 IU rhIL-2 protein intraperitoneally (i.p.) once on the

day of vaccination and twice daily on the next 2 days and were topically treated with 50

mg of 5% imiquimod cream (Aldara, Fougera) on the vaccination site once after each

vaccination.

Luminex assay and profiling of tumor infiltrating immune cells

Serum, spleen, and tumor tissue samples were collected from BP mice on day 6

after treatment. Twenty-five microliters of serum from each mouse was assayed using the

MILLIPLEX mouse / panels I, II, and III according to the

manufacturer's protocol (EMD Millipore). The concentration of each cytokine/chemokine

present in the serum samples was measured using a Luminex 200 system (Luminex

Corporation). Fresh tumor tissues were incubated with RPMI medium containing 1

mg/ml collagenase, 100 μg/ml hyaluronidase (Sigma-Aldrich) at 37°C for 60 min, and

manually dissociated to generate single cell suspensions. Single cell suspensions from

tumor or spleen tissues were then washed twice with staining buffer and incubated with a

cocktail of antibodies targeting surface markers at 4°C for 30 min. Cells were then fixed

and permeabilized using the Foxp3 fix and permeabilization kit according to

manufacturer’s protocol (eBioscience), and then incubated with a cocktail of antibodies

against intracellular markers. Stained samples were analyzed with a FACSCanto II or a

Downloaded from clincancerres.aacrjournals.org on September 25, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on August 1, 2019; DOI: 10.1158/1078-0432.CCR-19-1259 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

12

Helios mass cytometer (Fluidigm). Antibody details for the flow cytometry and mass

cytometry staining panels used in this study are provided in Supplementary Table 1.

Statistical analyses

Summary statistics (e.g., mean, SEM) of the data are reported. Assessments of

differences in continuous measurements between two groups were made using two-

sample t-test posterior to data transformation (typically logarithmic, if necessary), or

Wilcoxon rank-sum test. Differences in tumor size and T cell numbers among several

treatments were evaluated using analysis of variance (ANOVA) models. The Kaplan-

Meier method and log-rank test were used to compare survival between groups. P<0.05

was considered statistically significant. Graph generation and statistical analyses were

performed using GraphPad Prism (version 7) and R software programming language

(version 3.1.0).

RESULTS

The OX40 pathway plays a critical role in regulating the antitumor function of

tumor-infiltrating T cells (TILs) in melanoma patients

To determine the importance of the OX40 pathway in regulating the effector

function of TILs in melanoma patients, we first assessed the expression of OX40 on TILs

before and after re-stimulation with autologous tumors. We used two tumor-reactive TIL

cell lines previously generated from advanced melanoma patients (11,17). Cryopreserved

TILs were thawed and cultured in fresh culture medium in the presence of IL-2 for 3-4

days. Revived TILs were incubated with autologous tumors at the varying ratios of T

cells to tumor cells (E:T ratio), and the expression level of OX40 on the surface of TILs

over time was determined by flow cytometry analysis (Fig. 1A). The majority of resting

Downloaded from clincancerres.aacrjournals.org on September 25, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on August 1, 2019; DOI: 10.1158/1078-0432.CCR-19-1259 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

13

CD4+ TILs, but less than 10% of CD8+ TILs, expressed OX40 before re-stimulation. The

percentage of OX40+ CD4+ and CD8+ TILs peaked at 6 hours after TCR stimulation with

autologous tumors. Seventy-two hours after stimulation, the percentage of OX40+ TILs

returned to baseline levels. To validate that the OX40 expression on re-stimulated TILs is

tumor dependent, we assessed the percentage of OX40+ TILs after a 12-hour co-

incubation with tumors at E:T ratios ranging from 0.1:1 to 3:1. The results showed that

increasing the E:T ratio can enhance the percentage of both OX40+ CD4+ and CD8+ TILs

(Supplementary Fig 1A). These results demonstrate that OX40 expression on patient-

derived TILs is inducible and under the control of TCR signaling. Notably, OX40 was

expressed on a significant portion of CD8+ TILs (≈50%) at 6 hours after encountering

autologous tumors (Fig 1A), suggesting that activation of the OX40 pathway had the

potential to directly modulate the function of CD8+ T cells at tumor sites. To test this

hypothesis, we used a cytotoxicity assay based on the expression of cleaved caspase-3 in

tumor cells to evaluate whether activation of the OX40 signaling can alter cytotoxicity of

patient-derived TILs against autologous tumors. Given that immobilization of anti-OX40

antibody is required to activate the OX40 pathway in T cells (14), gamma-irradiated

PBMCs were pulsed with anti-human OX40 (hOX40) antibody (GSK3174998) for one

hour and used to stimulate TILs in the presence of autologous tumors. When compared

with Fc-fragment deleted anti-hOX40 antibody, full-length anti-hOX40-pulsed PBMCs

significantly increased TIL-induced apoptosis of tumors (Fig. 1B). In contrast, treatment

with anti-hOX40-pulsed PBMCs alone had no impact on tumor apoptosis. Additionally,

anti-mouse OX40-pulsed dendritic cells enhanced murine tumor apoptosis induced by

tumor-reactive CD8+ T cells (Supplementary Fig 1B). Although OX40 was not highly

Downloaded from clincancerres.aacrjournals.org on September 25, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on August 1, 2019; DOI: 10.1158/1078-0432.CCR-19-1259 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

14

expressed on resting cytotoxic CD8+ TILs, our results suggest that tumor exposure

induced the expression of OX40 on CD8+ TILs and that activation of OX40 signaling

enhances the cytotoxic function of CD8+ TILs.

Activation of the OX40 pathway improves antitumor activity of CD8+ T cells and

facilitates the generation of tumor-specific T cell memory

To characterize the in vivo effects of OX40 agonist antibody on antitumor activity

of CD8+ T cells, we adoptively transferred luciferase-expressing tumor-reactive CD8+ T

cells (pmel-1) into tumor-bearing mice one day after sublethal irradiation, which is

required for expansion of transferred T cells. These mice were then treated with either a

control antibody or an anti-mouse OX40 (mOX40) antibody as shown in Figure 2A. The

gp100-expressing MC38 tumors in mice treated with pmel-1 T cells grew significantly

slower than those not treated with T cells (P<0.0001, Fig. 2B). Importantly, anti-mOX40

treatment significantly delayed tumor growth in all T cell-treated mice (P<0.001, Fig.

2B). We also used bioluminescence imaging analysis to determine the change in tumor

trafficking of transferred tumor-reactive T cells in response to anti-mOX40 treatment.

Although the average bioluminescence intensity at the tumor site in mice treated with

anti-mOX40 was higher than the control group on day 6 after T-cell transfer, the

difference between these two group was not statistically significant (P=0.053, Fig 2C).

Given that an OX40 agonist antibody has been reported to enhance the

proliferation of CD4+ T helper (Th) cells and suppress the function of T regulatory (Treg)

cells (14), the OX40 agonist-enhanced antitumor activity of transferred CD8+ T cells

observed in this model may have been achieved by indirect regulation via CD4+ T cells

(18). To test whether anti-OX40 treatment can directly promote CD8+ T cell function, we

Downloaded from clincancerres.aacrjournals.org on September 25, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on August 1, 2019; DOI: 10.1158/1078-0432.CCR-19-1259 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

15

modified our murine ACT model to ensure that the OX40 agonist antibody only targeted

OX40-expressing transferred CD8+ T cells. We first transduced full-length human OX40

(hOX40) cDNA into murine pmel-1 T cells, and adoptively transferred hOX40-

expressing CD8+ pmel-1 T cells into tumor-bearing mice. Instead of using the mOX40

agonist, we treated all experimental mice with either isotype control antibody or anti-

human OX40 antibody. Reduced tumor size was observed in mice from the anti-hOX40

group as early as two days after the first dose of antibody treatment (Fig 2D).

Bioluminescence imaging analysis of mice on day 6 after T cell transfer revealed that the

number of transferred CD8+ T cells in the tumors was comparable between the control

and anti-hOX40 groups (Fig 2E). Although OX40 agonist antibody treatment had limited

impact on directly regulating tumor trafficking of CD8+ T cells, data from both models

supports that activation of OX40 signaling promotes the effector function of tumor

reactive CD8+ T cells.

We then examined the impact of anti-OX40 on the generation of antigen-specific

CD8+ memory T cells using a murine vaccine model (Fig. 3A) (11,16). The composition

of adjuvants in our previously described murine vaccination model was simplified to

avoid confounding effects of the anti-CD40 antibody on anti-OX40 antibody activity.

Briefly, fresh splenocytes from pmel-1 mice were adoptively transferred into C57BL/6

mice. Experimental mice were then vaccinated with the gp100 peptide on day 0 and day

28, and treated with anti-mOX40 on days 0, 4, 7, 12, 28, 31, 35, and 40. One week after

the last anti-OX40 treatment, experimental mice were challenged with gp100-expressing

MC38 tumor cells. By monitoring the percentage of transferred pmel-1 T cells in

PBMCs, we found that anti-OX40 enhanced the proliferation of pmel-1 T cells after

Downloaded from clincancerres.aacrjournals.org on September 25, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on August 1, 2019; DOI: 10.1158/1078-0432.CCR-19-1259 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

16

initial antigen stimulation (on day 5), and this positive effect of anti-OX40 was dose-

dependent (Fig 3B). Five days after the booster vaccine (on day 33), the percentage of

antigen-specific T cells in peripheral blood CD8+ T cells in mice is also significantly

higher than the rest of vaccinated mice (Fig 3B). Moreover, administration of 200 µg of

anti-OX40 to gp100-vaccinated mice before tumor inoculation successfully suppressed

the development of gp100-expressing tumors, indicating that OX40 agonists can induce

the generation of tumor-reactive memory T cells (Fig 3C).

OX40 agonist antibody synergizes with GSK2636771 in controlling the development

of PTEN-null melanoma

Previously, we demonstrated that oncogenic activation of the PI3K pathway by

PTEN loss promotes tumor-associated immunosuppressive mechanisms and is associated

with poor clinical outcomes in melanoma patients treated with anti-PD-1(11). We also

found that Braf-mutant, PTEN-null melanomas developed in Tyr:CreER; BRAFV600E/+;

PTENlox/lox mice (BP mice) are resistant to immune checkpoint inhibitory antibodies.

However, improved tumor growth inhibition was observed with the combination of anti-

PD-1 antibodies with GSK2636771, a PI3Kselective inhibitor, which was selected

based on data supporting a role for this PI3K isoform selective inhibition in cells with

loss of PTEN (11,19,20). The combination of pembrolizumab and GSK2636771 is now

being evaluated in a phase I/II clinical trial (NCT03131908).

To evaluate the effectiveness of combining T cell co-stimulatory receptor-based

immunotherapy and targeted therapy, we combined anti-OX40 with GSK2636771 in the

BP model (Fig. 4A). BP mice bearing measurable melanoma lesions were randomized

and treated with isotype control antibody, GSK2636771, anti-OX40 antibody, or a

Downloaded from clincancerres.aacrjournals.org on September 25, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on August 1, 2019; DOI: 10.1158/1078-0432.CCR-19-1259 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

17

combination of the two agents. Single agent anti-OX40 and single-agent GSK2636771

both failed to significantly inhibit tumor growth, but the combination was highly

effective and markedly extended the survival of BP tumor-bearing mice (the median

survival times of control, GSK2636771, anti-OX40 and combination groups are 14.5

days, 18 days, 14 days and 30 days respectively; p= 0.0021) (Fig. 4B and 4C). A linear

mixed model (21) determined that the anti-tumor effect of GSK2636771 with anti-OX40

was synergistic (P=0.0004, Supplementary Fig 2).

Importantly, no overt adverse effects or toxicities were observed with the

combination treatment. We further tested whether GSK2636771, anti-OX40, or the

combination affected the proliferation of antigen-specific T cells upon in vivo antigen

stimulation. These experiments showed that the combination treatment did not

significantly reduce whole blood cell counts or inhibit the proliferation of gp100-specific

T cells upon gp100 peptide immunization, (Supplementary Fig 3). Taken together, these

data suggest that combining anti-OX40 with GSK2636771 is another potentially effective

strategy to overcome immune resistance in melanomas with PTEN loss.

Combining anti-OX40 with a PI3Kβ inhibitor enhances T cell-mediated antitumor

immune activity

We then explored the underlying mechanisms by which PI3K inhibition

synergizes with anti-OX40 to control Braf-mutant, PTEN-null melanomas. Additional

tumor-bearing BP mice were treated with anti-OX40 and/or GSK2636771 as described

above. On day 6, serum, spleen, and tumor tissue samples were collected for immune

profiling. We measured the serum concentrations of a broad set of /

in each experimental mouse using Luminex assays. Among 43 tested

Downloaded from clincancerres.aacrjournals.org on September 25, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on August 1, 2019; DOI: 10.1158/1078-0432.CCR-19-1259 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

18

chemokines/cytokines, the combination treatment significantly increased the serum

concentrations of CCL2, CCL4, CCL15, CXCL10, and G-CSF in comparison to the

monotherapy or control treatments (Fig 5A and Supplementary Fig 4). Two of these

factors, CCL4 and CXCL10, are mainly produced by memory and/or effector T cells. In

addition, the serum levels of IFN-, another important antitumor cytokine produced by T

cells, were significantly higher in mice who received the combination treatment than in

mice treated with the control antibody or PI3K inhibitor alone (Fig 5A). Next, we

characterized the function and phenotype of immune cells in spleens from mice in the

different treatment groups using a 24-channel mass cytometry (CyTOF) panel

(Supplementary table 1). High-dimensional analysis using SPADE was performed to

examine the changes in immune cells in the treatment groups. Anti-OX40 monotherapy

reduced the percentage of M2 , which are immunosuppressive immune cells

found in peripheral lymphoid organs. In comparison to anti-OX40 monotherapy,

combination treatment further reduced the percentage of M2 macrophages and

significantly increased the expression of Ki-67 in T cells, dendritic cells, and M1

macrophages, suggesting enhanced proliferation of antitumor immune cells (Fig 5B). Due

to poor tumor infiltration of immune cells in this tumor model, the number of immune

cells at the tumor site was insufficient to perform CyTOF analysis. Therefore, we

evaluated the changes of T cell compartments at the tumor sites by a 5-channel flow

cytometry panel (Supplementary table 1). Although GSK2636771 significantly increased

the number of CD8+ T cells within tumors, there was no significant difference in the

number of tumor infiltrating CD8+ T cells in the GSK2636771 monotherapy group versus

the combination treatment group (Fig 6A). In addition, neither monotherapy treatment

Downloaded from clincancerres.aacrjournals.org on September 25, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on August 1, 2019; DOI: 10.1158/1078-0432.CCR-19-1259 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

19

nor the combination significantly altered the total number of tumor infiltrating CD4+ T

cells (Fig 6A). In addition, the number of Tregs in the tumors from the combination

treatment group was comparable to those of the monotherapy-treated tumors (Fig 6B). By

using the expression of Ki-67 to determine T cell function at tumor sites, we observed

that a significant increase in the percentage of Ki-67+ CD8+ T cells, but not in the

percentage of Ki-67+ CD4+ T cells, was detected with the combination versus each of the

other treatment groups (Fig 6C). These results suggest that combining GSK2636771 with

anti-OX40 promotes T cell-mediated antitumor immune responses by inducing robust

proliferation of CD8+ tumor infiltrating T cells.

DISCUSSION

In this article, we examined the expression of OX40 on tumor infiltrating

lymphocytes (TILs) derived from melanoma patients and tested whether stimulating

OX40 signaling can promote cytotoxicity of TILs against autologous tumor cells. Data

from preclinical tumor models confirmed that OX40 agonist antibody can improve T cell-

mediated antitumor immune responses by OX40 receptor engagement on CD8+ T cells

and inducing a protective tumor-specific T cell memory. To develop effective therapeutic

approaches in cancer patients who fail to respond to immune checkpoint blockade, we

used a transgenic Braf-mutant and PTEN loss murine model, which can spontaneously

develop immune-resistant melanomas, to evaluate the efficacy of combining OX40

agonist antibody with targeted therapy. The combination of an OX40 agonist antibody

and a selective PI3K inhibitor successfully potentiated the proliferation of antitumor

Downloaded from clincancerres.aacrjournals.org on September 25, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on August 1, 2019; DOI: 10.1158/1078-0432.CCR-19-1259 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

20

immune cells and suppressed tumor development in mice bearing Braf-mutant and PTEN

loss melanoma.

OX40, also known as TNFRSF4 or CD134, belongs to the

receptor superfamily (TNFRSF) (22). The engagement of three molecules of OX40 and

trimeric OX40 ligand (OX40L) initiates the signaling cascade through TNF receptor-

associated factors (TRAFs) and eventually drives NF-B activation (8). Although OX40

expression can be induced by TCR activation in both CD4+ and CD8+ T cells, the

expression of OX40 in CD4+ TILs is significantly higher than in CD8+ TILs (23,24). In

addition, in vitro and in vivo studies using viral infection and autoimmune disease models

have demonstrated that the effect of OX40 activation on CD8+ T cells is largely indirect

and is mediated by OX40 regulation of CD4+ T helper cell function (25-27). The

activation of the OX40 pathway in regulatory CD4+ T (Treg) cells has also been reported

to blunt the immunosuppressive function of Treg cells (14). Therefore, the current

working model of OX40 agonist antibody function in tumors mainly focuses on its effect

on CD4+ T cells. In our studies, we evaluated the OX40 expression levels in established

TIL lines from patients with advanced melanoma under different culture conditions.

Consistent with the results from other types of cancer, OX40 is predominantly expressed

on resting CD4+ TILs. However, upon encountering autologous tumor cells, the OX40

expression on CD8+ TILs is significantly upregulated and is restored to the baseline 72

hours after TCR stimulation. These results prompted us to test whether OX40 agonist

antibody can directly potentiate the function of tumor-reactive CD8+ TILs.

This hypothesis was first supported by data demonstrating that in vitro, anti-OX40

antibody can promote the proliferation of naïve CD8+ T cells after anti-CD3 stimulation

Downloaded from clincancerres.aacrjournals.org on September 25, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on August 1, 2019; DOI: 10.1158/1078-0432.CCR-19-1259 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

21

(14). In our study, by using patient-derived TILs and paired autologous tumor cells, we

found that cross-linked OX40 agonist antibody facilitated tumor apoptosis induced by

autologous TILs in vitro. Given that the tumor-specific cytotoxicity of TILs used in this

study has been previously shown to be largely dependent on the expression of MHC class

I molecules (11), our results suggest that OX40 signaling can directly regulate

cytotoxicity of tumor-reactive CD8+ TILs. Additionally, we examined the changes in

CD8+ T cells in response to anti-OX40 treatment using multiple murine tumor models.

Our in vivo data further demonstrated that anti-OX40 treatment enhances antitumor

activity of tumor-reactive CD8+ T cells. Furthermore, we used a vaccination model

established in our previous studies to evaluate T cell memory formation (16) and found

that anti-OX40 treatment promoted CD8+ T cell-mediated antitumor memory induced by

antigen vaccination. In particular, when treating tumor-bearing mice with anti-human

OX40 antibody without cross-reactivity to mouse OX40, we consistently observed

improved tumor suppression by human OX40-expressing CD8+ T cells. These results

imply that the direct role of OX40 signaling in tumor-reactive CD8+ T cells should not be

overlooked.

In clinic, the potential of OX40 as a target for cancer immunotherapy was initially

tested by using a murine anti-human OX40 IgG1 monoclonal antibody, 9B12 (28).

Although no patients achieved a clinical response based on the Response Evaluation

Criteria in Solid Tumors (RECIST), 12 of 30 treated patients had at least one regressed

metastatic nodule. Multiple fully human or humanized OX40 agonist antibodies have

been generated in the last two decades. At least five different antibodies have entered

clinical development, including GSK3174998 (GlaxoSmithKline), INCAGN01949

Downloaded from clincancerres.aacrjournals.org on September 25, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on August 1, 2019; DOI: 10.1158/1078-0432.CCR-19-1259 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

22

(Agenus), MEDI0562 (formerly of MedImmune), MOXR0916 (Genentech), and PF-

04518600 (Pfizer) (29-33). Similar to the preclinical results from murine tumor models

(34), the early data from two phase I clinical trials showed anti-OX40 monotherapy was

well tolerated in cancer patients, with only one serious treatment-related grade 3 adverse

event (pneumonitis responsive to corticosteroids) out of 71 patients reported (29,30).

These results suggest that OX40 agonist antibody treatment in cancer patients is generally

well-tolerated. In addition, up to 200 µg of anti-OX40 per dose for two weeks had no

adverse effect on the health and well-being of experimental mice in this study.

The antitumor effect of OX40 agonist antibody in cancer patients has not been

fully elucidated. However, primary and acquired resistance to anti-OX40 monotherapy

are expected in cancer patients due to a wide variety of tumor associated

immunosuppressive factors. Therefore, combining anti-OX40 therapy with other

treatments targeting these tumor-associated immunosuppressive factors may result in

better response rates and improved overall survival in cancer patients. Additionally,

eradication of well-established tumors has been reported in mice treated with anti-OX40

in combination with other immune reagents, such as TLR9 agonist in a spontaneous

breast cancer murine model (35). However, when evaluating the combination of anti-

OX40 and anti-PD-1, two research groups independently demonstrated that concurrent

treatment using these two agents induced T cell apoptosis and produced antagonistic

antitumor responses. Enhanced antitumor effect was only observed in mice sequentially

treated with anti-OX40 and anti-PD-1 (36,37). A similar potentially antagonistic

antitumor effect has been observed with the combination of immunotherapy and targeted

therapy. In that, although both CpG-based tumor vaccine and BRAF inhibitor have

Downloaded from clincancerres.aacrjournals.org on September 25, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on August 1, 2019; DOI: 10.1158/1078-0432.CCR-19-1259 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

23

therapeutic benefit as monotherapies in cancer patients, combining CpG with BRAF

inhibitor negates the antitumor effect of BRAF inhibitor in Braf-mutant tumors in a B-

cell dependent manner (38). Therefore, to develop potent therapeutic strategies for

OX40-based cancer treatment, we need to not only rationally choose combination

partners with complementing effects, but also optimize treatment schedules to maximize

the antitumor effect of anti-OX40.

Melanomas that spontaneously develop in transgenic mice bearing the Braf V600E

mutation and PTEN loss in melanocytes, display primary resistance to cancer

immunotherapy, due to lack of tumor-associated antigens and upregulated

immunosuppressive factors induced by oncogenic activation of the PI3K pathway

(11,39,40). In this study, we used this immune-resistant tumor model to evaluate the

therapeutic efficacy of anti-OX40 in combination with PI3K inhibition. Although anti-

OX40 monotherapy did not effectively control tumor development, concurrent treatment

of anti-OX40 and PI3K selective inhibition significantly delayed tumor growth and

extended the survival of mice bearing Braf-mutant and PTEN loss melanomas. Unlike the

combination of PI3K inhibition and ICB, this combinatorial approach did not

significantly increase the number of CD4+ and CD8+ T cells at the tumor sites but

promoted the proliferation of CD8+ T cells at the tumor sites. Anti-OX40 plus PI3K

inhibitor treatment also systemically enhanced the proliferation of antitumor immune

cells but reduced the number of immunosuppressive M2 macrophages. We also found

elevated serum levels of cytokine/chemokines, which were predominantly produced by

effector T cells in mice treated with the combination therapy. In addition, our studies

showed that this combination did not increase the susceptibility of T cells to activation-

Downloaded from clincancerres.aacrjournals.org on September 25, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on August 1, 2019; DOI: 10.1158/1078-0432.CCR-19-1259 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

24

induced apoptosis. Overall, our results offer the first preclinical evidence demonstrating

that combining anti-OX40 with PI3K inhibitor could be an effective treatment for

patients with PTEN loss tumors. These results also provide the rationale to clinically test

this combination in patients with immunoresistant PTEN loss tumors.

Taken together, our studies suggest that OX40 agonist-based combination

treatment can induce a robust and durable antitumor immune response by promoting

effector T cell function and the generation of memory T cells.

Downloaded from clincancerres.aacrjournals.org on September 25, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on August 1, 2019; DOI: 10.1158/1078-0432.CCR-19-1259 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

25

Authors’ Contributions

Conception and Design W. Peng, N. Yanamandra, P. Hwu, E. Paul.

Acquisition of data (provided required animals, cells, patient samples, reagents

clinical information, etc.): W. Peng, L. Williams, C. Xu, J. McKenzie, Y. Chen, R.

Mbofung, K. Voo, S. Leahey, A. Hoos, J Smothers.

Analysis and interpretation of data (statistical analysis and bioinformatic analysis):

W. Peng, B. Melendez, H. Jackson, J. Wang, G. Lizee, H. Tawbi, M. Davies, N.

Yanamandra, P. Hwu.

Writing and/or revision of the manuscript: W. Peng, J. McKenzie, H. Tawbi, M.

Davies, N. Yanamandra, P. Hwu.

Manuscript Review: E. Paul.

Study supervision: W. Peng, N. Yanamandra, P. Hwu. R. Srinivasan

Downloaded from clincancerres.aacrjournals.org on September 25, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on August 1, 2019; DOI: 10.1158/1078-0432.CCR-19-1259 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

26

FIGURE LEGENDS:

Figure 1. Kinetics of OX40 expression and in vitro effect of OX40 agonist antibody

on tumor infiltrating T cells (TILs) from melanoma patients. (A) Increased OX40

expression on both CD4+ and CD8+ TILs upon re-stimulation by autologous tumor cells.

Two patient-derived TIL lines, TIL2400 and TIL2559, were cultured in T cell growth

medium in the presence of IL-2 for at least 3 days. Revived TILs were then co-cultured

with autologous tumor cells at different ratios of effector to target cells (E:T). The

percentage of TILs expressing OX40 was determined by flow cytometry analysis at

indicated the time points. (B) Cross-linked anti-OX40 antibody enhanced the cytotoxicity

of TILs against autologous tumors. Irradiated PBMCs from healthy donors were pulsed

with 10 µg/ml of full length or Fc-fragment-deleted anti-human OX40 (GSK3174998) at

37°C for 1 hour. After washing with PBS, antibody-pulsed PBMCs were mixed with

DDAO-labelled tumor cells (Mel2400) and autologous TILs (TIL2400) at 37°C for an

additional three hours. The cell mixtures were stained intracellularly with an anti-cleaved

caspase-3 antibody. The cytotoxicity of TILs against tumors was evaluated by flow

cytometry analysis based on the percentage of cleaved caspase-3+ DDAO-labeled tumor

cells. One-way ANOVA demonstrated statistical significance (*P<0.05). Representative

data from three independent experiments are shown.

Figure 2. In vivo effect of OX40 activation on antitumor activity and memory

generation of tumor-reactive T cells. (A) Experimental setup of a murine ACT protocol

to evaluate in vivo effect of the activation of OX40 signaling on tumor-reactive T cells.

(B) Increased antitumor activity of tumor-reactive T cells in the presence of anti-mouse

OX40 antibody. Pmel-1 T cells that express a TCR specifically recognizing a melanoma

Downloaded from clincancerres.aacrjournals.org on September 25, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on August 1, 2019; DOI: 10.1158/1078-0432.CCR-19-1259 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

27

tumor antigen (gp100) were modified to express luciferase for in vivo monitoring of

tumor trafficking. Luciferase-expressing pmel-1 T cells were transferred into mice

bearing gp100-expressing MC38 tumors (N=4 per group). All experimental mice were

then treated with DC vaccine and IL-2 as described previously (41). One hundred

microgram per dose of anti-OX40 or control antibody was used to treat mice twice

weekly for two weeks. Tumor size was monitored every two days. (C) Luciferase

signaling intensity at tumor sites in mice with ACT. Tumor tracking of transferred pmel-1

T cells was evaluated on day 6 after T-cell transfer. Quantitative imaging analysis

revealed that anti-mOX40 did not significantly alter tumor tracking of transferred T cells.

(D) Anti-human OX40 antibody facilitated human OX40-expressing pmel-1 T cells

control of the growth of MC38/gp100 tumors (N= 5 per group). (E) In vivo tumor

tracking of human OX40-expressing T cells in response to anti-human OX40 antibody

treatment. Quantitative imaging analysis revealed that anti-hOX40 did not significantly

alter tumor trafficking of transferred T cells. The pairwise multiple comparisons after

two-way ANOVA test and the t-test were used to evaluate the statistical significance of

the difference in tumor growth and tumor trafficking, respectively. *** P<0.001 and

****P<0.0001. Representative data from two independent experiments are shown.

Figure 3. In vivo OX40 agonist antibody treatment enhanced the proliferation of

tumor-reactive T cells upon TCR stimulation and induced the generation of tumor-

specific T cell memory. (A) Schematic representation of a murine vaccine model used to

evaluate the in vivo effect of the activation of OX40 signaling on tumor-reactive T cells.

C57BL/6 mice were transferred with the splenocytes from pmel-1 mice and vaccinated

with gp100 peptide. Vaccinated mice received either control antibody or anti-mouse

Downloaded from clincancerres.aacrjournals.org on September 25, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on August 1, 2019; DOI: 10.1158/1078-0432.CCR-19-1259 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

28

OX40 antibody. After 4 weeks, mice received a second gp100 peptide vaccine (booster).

Gp100-expressing MC38 tumors were subcutaneously injected into vaccinated mice on

day 47. (B) The percentage of gp100-specific T cells in CD8+ T cells in the peripheral

blood of mice treated with OX40 agonist antibody. Thy1.1, a congenic marker for

transferred pmel-1 T cells, was used to determine the number of gp100-specific T cells in

peripheral blood after antigen stimulation. The pairwise multiple comparisons after two-

way ANOVA test demonstrated statistical significance (*P<0.05): control/ -OX40 50

g versus-OX40 100 g/-OX40 200 g on day 5; control/-OX40 50 g versus-

OX40 200 g on day 33. (C) The growth curves of MC38/gp100 tumors in vaccinated

mice treated with anti-OX40 (N=8 per group). Representative data from two independent

experiments are shown.

Figure 4. OX40 agonist antibody synergized with PI3K selective inhibition to

control the growth of PTEN loss tumors. (A) The treatment schedule of antibody and

the PI3K inhibitor (GSK2636771) is shown. Melanoma was induced in a group of

Tyr:CreER; PTENlox/lox; BRAFV600E/+ mice. Mice with measureable tumors were

randomized and treated with control, GSK2636771 (30 mg/kg/d), anti-mouse OX40 (50

µg/dose), and the combination of both reagents. (B) Tumor size was monitored in each of

the treatment groups every two days. The pairwise multiple comparisons after two-way

ANOVA test were used to determine statistical significance. *P<0.05. (D) Kaplan-Meier

survival curves of mice treated with GSK2636771 and/or anti-mouse OX40. Log-rank

test demonstrated statistical significance (P<0.05): GSK2636771+anti-OX40 vs control,

GSK2636771, anti-OX40 (N=4–7). Data presented are a summary of two independent

experiments.

Downloaded from clincancerres.aacrjournals.org on September 25, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on August 1, 2019; DOI: 10.1158/1078-0432.CCR-19-1259 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

29

Figure 5. OX40 agonist antibody in combination with PI3K selective inhibition

altered the immune cell profile and the serum concentration of cytokine/chemokines

produced by T cells. Tyr:CreER; PTENlox/lox; BRAFV600E/+ mice with measureable

tumors were treated with control, GSK2636771, anti-mouse OX40 , and the combination

of both reagents. Mice were euthanized on day 6 after treatment and used to characterize

the changes in the immune profile of mice in the different treatment groups. (A) The

serum levels of T cell-associated cytokines/chemokines. Serum from each experimental

mouse was collected and used to measure the concentration of 43 cytokines/chemokines

using the MILLIPLEX MAP mouse cytokine/chemokine panels. The average of the

serum cytokine/chemokine concentration in each group is shown. *P < 0.05, ** P<0.01,

****P<0.0001 (N=3). (B) The results of CyTOF analysis revealed the systemic effects of

anti-OX40 alone or in combination with PI3Ki. Spleens were collected from mice in the

different treatment groups and processed into single cell suspensions at the concentration

of 20 million cells/ml. Equal amounts of single cell suspensions from experimental mice

in each group were pooled (N=3). Pooled samples of control, anti-OX40, and the

combination groups were analyzed by CyTOF to determine the percentages of different

immune cell subsets and their proliferation (measured by Ki-67 expression). High-

dimensional visualization of changes in Ki-67 expression in response to treatment was

generated using SPADE. The ratio of Ki-67 (OX40 alone or combination group): Ki-67

(control group) is represented by the color scale, with blue indicating a reduced level of

Ki-67 after treatment. The number and size of nodules in each immune cell subset

represents the percentage of the indicated immune cell subset in spleens. Data presented

are a summary of two independent experiments.

Downloaded from clincancerres.aacrjournals.org on September 25, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on August 1, 2019; DOI: 10.1158/1078-0432.CCR-19-1259 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

30

Figure 6. OX40 agonist antibody in combination with PI3K selective inhibition

promoted the proliferation of tumor infiltrating CD8+ T cells in mice with PTEN-

loss tumor. Tyr:CreER; PTENlox/lox; BRAFV600E/+ mice with measureable tumors were

treated with control, GSK2636771, anti-mouse OX40, and the combination of both

reagents (N=3). On day 6 after treatment, tumor tissues were harvested, weighted, and

stained with antibodies for flow cytometry analysis. (A) The total number of CD4+ and

CD8+ T cells in tumors from mice treated with anti-OX40 and/or GSK2636771. (B) The

percentage of Treg cells (CD25+FOXp3+) in CD4+ T cells in tumors from mice treated

with anti-OX40 and/or GSK2636771. (C) The percentage of Ki-67+ CD4+ and CD8+ T

cells in tumors from mice treated with anti-OX40 and/or GSK2636771. One-Way

ANOVA test demonstrated statistical significance (P<0.05): *P < 0.05, ** P<0.01, and

N.S (no statistical significance). Data presented are a summary of two independent

experiments.

Downloaded from clincancerres.aacrjournals.org on September 25, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on August 1, 2019; DOI: 10.1158/1078-0432.CCR-19-1259 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

31

Reference

1. Sharma P, Allison JP. The future of immune checkpoint therapy. Science

2015;348(6230):56-61 doi 10.1126/science.aaa8172.

2. Park YJ, Kuen DS, Chung Y. Future prospects of immune checkpoint blockade in

cancer: from response prediction to overcoming resistance. Exp Mol Med

2018;50(8):109 doi 10.1038/s12276-018-0130-1.

3. Jensen SM, Maston LD, Gough MJ, Ruby CE, Redmond WL, Crittenden M, et al.

Signaling through OX40 enhances antitumor immunity. Semin Oncol

2010;37(5):524-32 doi 10.1053/j.seminoncol.2010.09.013.

4. Chester C, Sanmamed MF, Wang J, Melero I. Immunotherapy targeting 4-1BB:

mechanistic rationale, clinical results, and future strategies. Blood

2018;131(1):49-57 doi 10.1182/blood-2017-06-741041.

5. Callahan MK, Postow MA, Wolchok JD. CTLA-4 and PD-1 Pathway Blockade:

Combinations in the Clinic. Frontiers in oncology 2014;4:385 doi

10.3389/fonc.2014.00385.

6. Darvin P, Toor SM, Sasidharan Nair V, Elkord E. Immune checkpoint inhibitors:

recent progress and potential biomarkers. Exp Mol Med 2018;50(12):165 doi

10.1038/s12276-018-0191-1.

7. Jenkins RW, Barbie DA, Flaherty KT. Mechanisms of resistance to immune

checkpoint inhibitors. Br J Cancer 2018;118(1):9-16 doi 10.1038/bjc.2017.434.

8. Croft M, So T, Duan W, Soroosh P. The significance of OX40 and OX40L to T-

cell biology and immune disease. Immunological reviews 2009;229(1):173-91 doi

10.1111/j.1600-065X.2009.00766.x.

Downloaded from clincancerres.aacrjournals.org on September 25, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on August 1, 2019; DOI: 10.1158/1078-0432.CCR-19-1259 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

32

9. Chacon JA, Wu RC, Sukhumalchandra P, Molldrem JJ, Sarnaik A, Pilon-Thomas

S, et al. Co-stimulation through 4-1BB/CD137 improves the expansion and

function of CD8(+) melanoma tumor-infiltrating lymphocytes for adoptive T-cell

therapy. PLoS One 2013;8(4):e60031 doi 10.1371/journal.pone.0060031

PONE-D-12-34281 [pii].

10. Peng W, Ye Y, Rabinovich BA, Liu C, Lou Y, Zhang M, et al. Transduction of

tumor-specific T cells with CXCR2 improves migration to

tumor and antitumor immune responses. Clin Cancer Res 2010;16(22):5458-68

doi 10.1158/1078-0432.CCR-10-07121078-0432.CCR-10-0712 [pii].

11. Peng W, Chen JQ, Liu C, Malu S, Creasy C, Tetzlaff MT, et al. Loss of PTEN

Promotes Resistance to T Cell-Mediated Immunotherapy. Cancer Discov

2016;6(2):202-16 doi 10.1158/2159-8290.CD-15-0283.

12. Kitamura T, Koshino Y, Shibata F, Oki T, Nakajima H, Nosaka T, et al.

Retrovirus-mediated gene transfer and expression cloning: powerful tools in

functional genomics. Exp Hematol 2003;31(11):1007-14.

13. Rabinovich BA, Ye Y, Etto T, Chen JQ, Levitsky HI, Overwijk WW, et al.

Visualizing fewer than 10 mouse T cells with an enhanced firefly luciferase in

immunocompetent mouse models of cancer. Proc Natl Acad Sci U S A

2008;105(38):14342-6 doi 10.1073/pnas.08041051050804105105 [pii].

14. Voo KS, Bover L, Harline ML, Vien LT, Facchinetti V, Arima K, et al.

Antibodies targeting human OX40 expand effector T cells and block inducible

and natural regulatory T cell function. J Immunol 2013;191(7):3641-50 doi

10.4049/jimmunol.1202752.

Downloaded from clincancerres.aacrjournals.org on September 25, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on August 1, 2019; DOI: 10.1158/1078-0432.CCR-19-1259 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

33

15. Liu C, Lewis CM, Lou Y, Xu C, Peng W, Yang Y, et al. Agonistic antibody to

CD40 boosts the antitumor activity of adoptively transferred T cells in vivo. J

Immunother 2012;35(3):276-82 doi 10.1097/CJI.0b013e31824e7f4300002371-

201204000-00008 [pii].

16. Hailemichael Y, Dai Z, Jaffarzad N, Ye Y, Medina MA, Huang XF, et al.

Persistent antigen at vaccination sites induces tumor-specific CD8(+) T cell

sequestration, dysfunction and deletion. Nat Med 2013;19(4):465-72 doi

10.1038/nm.3105nm.3105 [pii].

17. Cascone T, McKenzie JA, Mbofung RM, Punt S, Wang Z, Xu C, et al. Increased

Tumor Glycolysis Characterizes Immune Resistance to Adoptive T Cell Therapy.

Cell Metab 2018;27(5):977-87 e4 doi 10.1016/j.cmet.2018.02.024.

18. Gough MJ, Ruby CE, Redmond WL, Dhungel B, Brown A, Weinberg AD. OX40

agonist therapy enhances CD8 infiltration and decreases immune suppression in

the tumor. Cancer Res 2008;68(13):5206-15 doi 10.1158/0008-5472.CAN-07-

6484.

19. Mateo J, Ganji G, Lemech C, Burris HA, Han SW, Swales K, et al. A First-Time-

in-Human Study of GSK2636771, a Phosphoinositide 3 Kinase Beta-Selective

Inhibitor, in Patients with Advanced Solid Tumors. Clin Cancer Res

2017;23(19):5981-92 doi 10.1158/1078-0432.CCR-17-0725.

20. Schmit F, Utermark T, Zhang S, Wang Q, Von T, Roberts TM, et al. PI3K

isoform dependence of PTEN-deficient tumors can be altered by the genetic

context. Proc Natl Acad Sci U S A 2014;111(17):6395-400 doi

10.1073/pnas.1323004111.

Downloaded from clincancerres.aacrjournals.org on September 25, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on August 1, 2019; DOI: 10.1158/1078-0432.CCR-19-1259 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

34

21. Slinker BK. The statistics of synergism. J Mol Cell Cardiol 1998;30(4):723-31

doi 10.1006/jmcc.1998.0655.

22. Buchan SL, Rogel A, Al-Shamkhani A. The immunobiology of CD27 and OX40

and their potential as targets for cancer immunotherapy. Blood 2018;131(1):39-48

doi 10.1182/blood-2017-07-741025.

23. Lai C, August S, Albibas A, Behar R, Cho SY, Polak ME, et al. OX40+

Regulatory T Cells in Cutaneous Squamous Cell Carcinoma Suppress Effector T-

Cell Responses and Associate with Metastatic Potential. Clin Cancer Res

2016;22(16):4236-48 doi 10.1158/1078-0432.CCR-15-2614.

24. Montler R, Bell RB, Thalhofer C, Leidner R, Feng Z, Fox BA, et al. OX40, PD-1

and CTLA-4 are selectively expressed on tumor-infiltrating T cells in head and

neck cancer. Clin Transl Immunology 2016;5(4):e70 doi 10.1038/cti.2016.16.

25. Serghides L, Bukczynski J, Wen T, Wang C, Routy JP, Boulassel MR, et al.

Evaluation of OX40 ligand as a costimulator of human antiviral memory CD8 T

cell responses: comparison with B7.1 and 4-1BBL. J Immunol

2005;175(10):6368-77.

26. Sitrin J, Suto E, Wuster A, Eastham-Anderson J, Kim JM, Austin CD, et al. The

Ox40/Ox40 Ligand Pathway Promotes Pathogenic Th Cell Responses,

Plasmablast Accumulation, and Lupus Nephritis in NZB/W F1 Mice. J Immunol

2017;199(4):1238-49 doi 10.4049/jimmunol.1700608.

27. Kopf M, Ruedl C, Schmitz N, Gallimore A, Lefrang K, Ecabert B, et al. OX40-

deficient mice are defective in Th cell proliferation but are competent in

Downloaded from clincancerres.aacrjournals.org on September 25, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on August 1, 2019; DOI: 10.1158/1078-0432.CCR-19-1259 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

35

generating B cell and CTL Responses after virus infection. Immunity

1999;11(6):699-708.

28. Curti BD, Kovacsovics-Bankowski M, Morris N, Walker E, Chisholm L, Floyd

K, et al. OX40 is a potent immune-stimulating target in late-stage cancer patients.

Cancer Res 2013;73(24):7189-98 doi 10.1158/0008-5472.CAN-12-4174.

29. Infante JR, Ahlers CM, Hodi FS, Postel-Vinay S, Schellens JHM, Heymach J, et

al. ENGAGE-1: A first in human study of the OX40 agonist GSK3174998 alone

and in combination with pembrolizumab in patients with advanced solid tumors.

Journal of Clinical Oncology 2016;34(15_suppl):TPS3107-TPS doi

10.1200/JCO.2016.34.15_suppl.TPS3107.

30. Glisson BS, Leidner R, Ferris RL, Powderly J, Rizvi N, Norton JD, et al. Phase 1

study of MEDI0562, a humanized OX40 agonist monoclonal antibody (mAb), in

adult patients (pts) with advanced solid tumors. Annals of Oncology

2016;27(suppl_6):1052PD-PD doi 10.1093/annonc/mdw378.07.

31. Gonzalez AM, Manrique ML, Breous E, Savitsky D, Waight J, Gombos R, et al.

Abstract 3204: INCAGN01949: an anti-OX40 agonist antibody with the potential

to enhance tumor-specific T-cell responsiveness, while selectively depleting

intratumoral regulatory T cells. Cancer Research 2016;76(14 Supplement):3204-

doi 10.1158/1538-7445.Am2016-3204.

32. Infante JR, Hansen AR, Pishvaian MJ, Chow LQM, McArthur GA, Bauer TM, et

al. A phase Ib dose escalation study of the OX40 agonist MOXR0916 and the

PD-L1 inhibitor atezolizumab in patients with advanced solid tumors. Journal of

Downloaded from clincancerres.aacrjournals.org on September 25, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on August 1, 2019; DOI: 10.1158/1078-0432.CCR-19-1259 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

36

Clinical Oncology 2016;34(15_suppl):101- doi

10.1200/JCO.2016.34.15_suppl.101.

33. Diab A, El-Khoueiry A, Eskens FA, Ros W, Thompson JA, Konto C, et al. A

first-in-human (FIH) study of PF-04518600 (PF-8600) OX40 agonist in adult

patients (pts) with select advanced malignancies. Annals of Oncology

2016;27(suppl_6):1053PD-PD doi 10.1093/annonc/mdw378.08.

34. Aspeslagh S, Postel-Vinay S, Rusakiewicz S, Soria JC, Zitvogel L, Marabelle A.

Rationale for anti-OX40 cancer immunotherapy. Eur J Cancer 2016;52:50-66 doi

10.1016/j.ejca.2015.08.021.

35. Sagiv-Barfi I, Czerwinski DK, Levy S, Alam IS, Mayer AT, Gambhir SS, et al.

Eradication of spontaneous malignancy by local immunotherapy. Sci Transl Med

2018;10(426) doi 10.1126/scitranslmed.aan4488.

36. Shrimali RK, Ahmad S, Verma V, Zeng P, Ananth S, Gaur P, et al. Concurrent

PD-1 Blockade Negates the Effects of OX40 Agonist Antibody in Combination

Immunotherapy through Inducing T-cell Apoptosis. Cancer Immunol Res

2017;5(9):755-66 doi 10.1158/2326-6066.CIR-17-0292.

37. Messenheimer DJ, Jensen SM, Afentoulis ME, Wegmann KW, Feng Z, Friedman

DJ, et al. Timing of PD-1 Blockade Is Critical to Effective Combination

Immunotherapy with Anti-OX40. Clin Cancer Res 2017;23(20):6165-77 doi

10.1158/1078-0432.CCR-16-2677.

38. Huang L, Wang Z, Liu C, Xu C, Mbofung RM, McKenzie JA, et al. CpG-based

immunotherapy impairs antitumor activity of BRAF inhibitors in a B-cell-

dependent manner. Oncogene 2017;36(28):4081-6 doi 10.1038/onc.2017.35.

Downloaded from clincancerres.aacrjournals.org on September 25, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on August 1, 2019; DOI: 10.1158/1078-0432.CCR-19-1259 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

37

39. Hooijkaas A, Gadiot J, Morrow M, Stewart R, Schumacher T, Blank CU.

Selective BRAF inhibition decreases tumor-resident frequencies in a

mouse model of human melanoma. Oncoimmunology 2012;1(5):609-17 doi

10.4161/onci.20226.

40. Cooper ZA, Juneja VR, Sage PT, Frederick DT, Piris A, Mitra D, et al. Response

to BRAF inhibition in melanoma is enhanced when combined with immune

checkpoint blockade. Cancer Immunol Res 2014;2(7):643-54 doi 10.1158/2326-

6066.CIR-13-0215.

41. Peng W, Liu C, Xu C, Lou Y, Chen J, Yang Y, et al. PD-1 blockade enhances T-

cell migration to tumors by elevating IFN-gamma inducible chemokines. Cancer

Res 2012;72(20):5209-18 doi 10.1158/0008-5472.CAN-12-11870008-5472.CAN-

12-1187 [pii].

Downloaded from clincancerres.aacrjournals.org on September 25, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on August 1, 2019; DOI: 10.1158/1078-0432.CCR-19-1259 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Downloaded from clincancerres.aacrjournals.org on September 25, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on August 1, 2019; DOI: 10.1158/1078-0432.CCR-19-1259 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Downloaded from clincancerres.aacrjournals.org on September 25, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on August 1, 2019; DOI: 10.1158/1078-0432.CCR-19-1259 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Downloaded from clincancerres.aacrjournals.org on September 25, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on August 1, 2019; DOI: 10.1158/1078-0432.CCR-19-1259 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Downloaded from clincancerres.aacrjournals.org on September 25, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on August 1, 2019; DOI: 10.1158/1078-0432.CCR-19-1259 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Downloaded from clincancerres.aacrjournals.org on September 25, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on August 1, 2019; DOI: 10.1158/1078-0432.CCR-19-1259 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Downloaded from clincancerres.aacrjournals.org on September 25, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on August 1, 2019; DOI: 10.1158/1078-0432.CCR-19-1259 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Anti-OX40 antibody directly enhances the function of tumor-reactive CD8 + T cells and synergizes with PI3Kβ inhibition in PTEN loss melanoma

Weiyi Peng, Leila J. Williams, Chunyu Xu, et al.

Clin Cancer Res Published OnlineFirst August 1, 2019.

Updated version Access the most recent version of this article at: doi:10.1158/1078-0432.CCR-19-1259

Supplementary Access the most recent supplemental material at: Material http://clincancerres.aacrjournals.org/content/suppl/2019/08/01/1078-0432.CCR-19-1259.DC1

Author Author manuscripts have been peer reviewed and accepted for publication but have not yet been Manuscript edited.

E-mail alerts Sign up to receive free email-alerts related to this article or journal.

Reprints and To order reprints of this article or to subscribe to the journal, contact the AACR Publications Subscriptions Department at [email protected].

Permissions To request permission to re-use all or part of this article, use this link http://clincancerres.aacrjournals.org/content/early/2019/08/01/1078-0432.CCR-19-1259. Click on "Request Permissions" which will take you to the Copyright Clearance Center's (CCC) Rightslink site.

Downloaded from clincancerres.aacrjournals.org on September 25, 2021. © 2019 American Association for Cancer Research.