Purpose:

Immunotherapy is a promising approach for many oncological malignancies, including glioblastoma, however, there are currently no available tools or biomarkers to accurately assess whole-body immune responses in patients with glioblastoma treated with immunotherapy. Here, the utility of OX40, a costimulatory molecule mainly expressed on activated effector T cells known to play an important role in eliminating cancer cells, was evaluated as a PET imaging biomarker to quantify and track response to immunotherapy.

Experimental Design:

A subcutaneous vaccination approach of CpG oligodeoxynucleotide, OX40 mAb, and tumor lysate at a remote site in a murine orthotopic glioma model was developed to induce activation of T cells distantly while monitoring their distribution in stimulated lymphoid organs with respect to observed therapeutic effects. To detect OX40-positive T cells, we utilized our in-house–developed 89Zr-DFO-OX40 mAb and in vivo PET/CT imaging.

Results:

ImmunoPET with 89Zr-DFO-OX40 mAb revealed strong OX40-positive responses with high specificity, not only in the nearest lymph node from vaccinated area (mean, 20.8%ID/cc) but also in the spleen (16.7%ID/cc) and the tumor draining lymph node (11.4%ID/cc). When the tumor was small (<106 p/sec/cm2/sr in bioluminescence imaging), a high number of responders and percentage shrinkage in tumor signal was indicated after only a single cycle of vaccination.

Conclusions:

The results highlight the promise of clinically translating cancer vaccination as a potential glioma therapy, as well as the benefits of monitoring efficacy of these treatments using immunoPET imaging of T-cell activation.

Translational Relevance

Glioblastoma remains one of most challenging malignancies to treat. Immunotherapy has altered conventional strategies for treating cancer and is a promising option for glioblastoma. Despite the promise of such therapies, there is a critical need for tools that enable accurate monitoring of immune responses to evaluate the effectiveness of immunotherapy. In this study, we specifically monitored stimulated T-cell responses in a murine orthotopic glioma model using immunoPET. 89Zr-DFO-OX40 mAb immunoPET enabled detection of whole-body immune stimulation of antigen-specific T cells in lymphoid organs. OX40/CD4 T-cell ratio was significantly elevated in the vaccinated cohort, corresponding with treatment response. This novel noninvasive imaging method is predicted to support the management of immunotherapy in the clinical setting for patients with glioblastoma.

Glioblastoma (GBM) is a devastating fatal malignancy of the central nervous system and the most common type of histology representing up to 48% of all gliomas (1). It is uniformly difficult to treat, and the majority of patients experience disease relapse despite first-line intensive therapies such as surgical resections, radiotherapy, and chemotherapy with the DNA-alkylating agent temozolomide (2). Because GBM relapses can occur rapidly while also developing a resistance to therapeutic drugs, the median survival time is between 10 to 14 months (1, 3). Because of this poor prognosis and the associated diminished quality of life, more effective treatments are urgently needed.

Cancer immunotherapies are being increasingly investigated as a potential breakthrough for the treatment of various refractory malignancies (4). Among actively investigated immunocompetent cells, T cells play a major role in tumor eradication. Although immunotherapy in GBM is quite challenging due to tumor heterogeneity (5), capacities to subvert T-cell antitumor responses (6), and T-cell exhaustion (7), a number of preclinical and clinical attempts have been reported with successful results in the literature (8–11). There are two primary modes of immunotherapy being investigated with autologous T cells: ex vivo adoptive T-cell transfer (12, 13) and in vivo T-cell stimulating molecules. Adoptive T-cell transfer is a powerful personalized therapeutic approach with high specificity, but it is associated with higher cost and more resources per individual. T-cell targeting antibodies have been studied thoroughly and commercialized, as represented by anti–CTLA-4 antibodies and PD-1 antibodies (14, 15). OX40 (or CD134) is a promising activated T-cell surface marker, known to be a costimulatory transmembrane molecule of the tumor necrosis factor (TNF) superfamily, primarily expressed on activated effector T cells and regulatory T cells (Treg). Importantly, OX40 is not found on resting naïve T cells or the majority of resting memory T cells (16, 17). OX40 promotes proliferation and longer survival of effector and memory T-cell populations, while also suppressing the differentiation and activity of Tregs (18). Given these properties, an OX40 receptor monoclonal antibody (mAb) was tested as a treatment and showed preferable response in murine tumors (19), as well as glioma (20, 21). A third category of immune therapeutic methods is vaccination. Vaccinations prime the immune system against the tumor by introducing something highly immunogenic to the body, thus stimulating immunologic memory (2). Particularly, CpG-ODN, a Toll-like receptor (TLR) 9 agonist, is a promising stimulant, promoting maturation and activation of antigen-presenting cells and induction of Th1 T cells. We previously determined that direct injection of CpG and anti-OX40 antibody is a highly promising immunostimulatory regimen in subcutaneous murine lymphoma tumors (22). However, the effectiveness of local injection of CpG-ODN to GBM in humans has been previously reported as limited (23). On the other hand, subcutaneous vaccinations incorporating tumor cell lysates with dendritic cells were reported as an effective therapy for GBM in clinical trials (24, 25), possibly indicating that, in case of GBM, vaccination works more effectively when loaded at a remote site from the tumor than injected directly to the tumor. Indeed, multiple administrations of intradermal tumor lysate vaccine cocktail with CpG, followed by intraperitoneal administration of Fc-OX40L, was reported to exhibit a significant increase in the proliferation of brain-infiltrating CD4 and CD8 T cells followed by therapeutic effects in a murine glioma-bearing model (26).

While there has been much progress in developing immunotherapies, commercially available imaging tools capable of quantifying and monitoring response to these therapies remain limited in the clinical field. The well-established medical imaging systems CT and MRI can evaluate tumor morphology characteristics with adequate spatial resolution and can indirectly provide the information of cell density and vascularity. However, these approaches lack systemic molecular information. PET is a unique nuclear medicine modality, which enables real-time whole-body imaging of molecular targets and/or biochemical processes by administering a radiolabeled tracer. The most commonly used tracer is 18F-FDG, a glucose analogue radiolabeled with F-18. Because glucose metabolism is significantly elevated in malignant cells (27), the sensitivity of depicting tumor lesions using 18F-FDG-PET imaging is sufficient for most malignancies for the purpose of staging and monitoring therapeutic effect. The disadvantage, however, is that 18F-FDG exhibits low specificity, owing to accumulation in all hypermetabolic cells, including those found in the brain and active inflammatory cells including macrophages (28). Moreover, the half-life of F-18 is 109.7 minutes, and therefore, multiple tracer injections will be required for longitudinal observation, resulting in increased radiation dose. Recently, there have been several reports focused on engineering new tracers targeting CD8 T cells (29–31) or CAR T cells (32) using biologics labeled with longer half-life radioisotopes such as Cu-64 (12.7 hours) or Zr-89 (78.4 hours). Imaging lymphocyte trafficking with ex vivo labeled cells were reported (33), such as Th1 cells labeled with 64Cu-PTSM (34) and γδ T cells with 89Zr (35), but it takes more effort than in vivo labeling. Labeled antibodies are also used to track T cells in vivo. PET probes targeting checkpoint molecules such as PD-1/PD-L1/CTLA-4 have been developed as well to map the localization of checkpoint molecules expressing tumor-infiltrating lymphocytes in murine models (36) and clinical trials (37) to estimate the response for checkpoint inhibitors in various cancers. These imaging tools are aimed to predict the response to immune checkpoint inhibitors; however, emerging varieties of cancer therapy highlight the need of direct evaluation of immune cells. It is worthwhile to explore a method to depict the live T cells that act as key players to eradicate the tumor.

The specific functional expression of OX40 on T cells suggests that it could act as a biomarker of activated memory T cells working to eradicate tumors (38). We previously demonstrated activated tumor-infiltrating T cells using intratumoral vaccination to a lymphoma xenograft model using 64Cu-DOTA-OX40 mAb (39). To expand this approach to a murine orthotopic glioma model, we treated mice with a subcutaneous cocktail vaccination of CpG, tumor lysate, and OX40 mAb, near left axillar lymph node, which is remote from the tumor. In addition, we utilized radiolabeled 89Zr-DFO-OX40 mAb to achieve longer observation than 64Cu-DOTA-OX40 mAb, and investigated the kinetics of whole-body immune response induced by a remote vaccination noninvasively and longitudinally by PET/CT. This is the first study to demonstrate the feasibility of visualizing and quantifying the whole-body immune response to a remotely introduced cancer vaccination for treating GBM.

The aim of the study was to explore the potential advantages of our newly developed 89Zr-DFO-OX40 mAb PET/CT imaging approach for monitoring T-cell activation noninvasively in a glioma model using a remote immune-stimulating vaccination strategy.

Cell lines and animal models

All animal studies were conducted under a protocol approved by the Administrative Panel on Laboratory Animal Care (APLAC) of our institute. GL26 mouse glioma cells (gift in 2015 from Dr. Gerry Grant, Stanford University, Stanford, CA) labeled with Luc2-eGFP (GL26α8) were generated by isolation from the dissociated brain tumor using GFP as a selection marker to be implanted in multiple mice intracranially and monitored their growth noninvasively by bioluminescence of FL activities to select for the clones that had optimal in vivo growth properties. GL26α8 cells were cultured in DMEM with penicillin (100 U/mL), antibiotics and anti-mycotic (100 μg/mL), and FBS [10% (v/v)] and incubated at 37°C with 5% CO2. Cells were used for in vivo experiments when they reached 80% confluence. Mycoplasma testing was performed every month. Orthotopic GL26α8 brain tumors were induced in 6- to 8-week-old female albino C57/BL6 mice (The Jackson Laboratory) by intracranial injection. Mice were anesthetized with 1.5% to 2.0% of isoflurane, sterilized with ethanol, and 105 GL26α8 cells suspension in 3 μL PBS was injected in 3 mm depth from the surface, 3 mm right lateral to the sagittal suture, and 3 mm posterior to coronal suture. Postsurgical mice were monitored under a warm circumstance until they resumed normal behavior. Tumor engraftment was confirmed with bioluminescent imaging (BLI) with IVIS spectrum (Perkin Elmer) between 7 and 14 days after tumor inoculation. The BLI signal above 105 (p/sec/cm2/sq) was defined as a baseline with at least two consecutive tumor growth confirmations.

Cancer vaccination

Once tumor growth was confirmed, mice were randomly classified into vaccine group and control, and monitored every 3 to 4 days with BLI. We modified the previously reported vaccine strategy for glioma model (26). Vaccine group received a mixture of 50 μg of CpG-ODN (CpG) and 65 μg of GL26α8 tumor lysate in 100 μL PBS, and on the following day, 50 μg of anti-mouse OX40 mAb (clone: OX86, RRID: AB_1107592, Bio X Cell) in 100 μL PBS, in their left shoulders. Tumor lysate was prepared referring to the same prior study (26). The lysates were stored at −80°C freezer and thawed prior to injection. OX40 was injected 24 hours later, referring to a report that effect is well observed at that time point after in situ administration of CpG in subcutaneous lymphoma model (40). Control mice received 100 μL of PBS on both days.

89Zr-DFO-OX40 mAb generation

Briefly, 1 mg of anti-mouse OX40 antibody or isotype control (rat IgG1, clone: HRPN, RRID: AB_1107775, Bio X Cell) was diluted to 1 mg/mL with PBS, then buffer exchanged with pH 8.8–9.0 PBS solution. Subsequently, 10-fold excess of p-SCN-Bn-deferoxamine (DFO) was added to the antibody solution and incubated for 60 minutes at 37°C. Conjugated product showed 2.49 ± 0.12 molecules of DFO per mAb. Finally, the mixture was buffer exchanged with PBS (pH 7.4) using Vivaspin 2, 50K cutoff centrifugal concentrator. The concentration of DFO conjugated mAb was measured by Thermo Scientific NanoDrop One Microvolume UV-Vis Spectrophotometer. Na2CO3 (1M solution) was added to 89Zr-oxalic acid until the pH reached 7.1–7.8, and subsequently added to DFO-OX40 mAb or DFO-isotype control solution. After 60-minute incubation at 37°C, 89Zr-DFO-OX40 mAb was then purified by Zeba Spin Desalting Columns, 7K MWCO, 0.5 mL, 1,000 × g, 1 minute. Finally, radiochemical purity was assessed and confirmed >95% by iTLC (Instant Thin Layer Chromatography).

PET/CT imaging and volumetric quantitative analysis

89Zr-DFO-OX40 mAb or 89Zr-DFO-rat IgG (1.5–2 MBq) was administered intravenously to orthotopic mice via tail vein, and sequential standalone PET/CT scans were acquired three times; day 1, day 2, and day 5. Time points were selected considering the physical half-life of 89Zr and typical long circulation times of full-length antibodies (41). PET/CT studies were performed on an Inveon microPET/CT rodent model scanner, carrying a cone beam X-ray source and a flat panel detector on CT (Siemens Medical Solutions USA, Inc.). Anesthesia was induced with 2.5% isoflurane and maintained at 2%, and mice were placed in a prone position in the scanner. Ten minutes of attenuation correction of CT scan was performed just before the 15-minute static 3D PET scan. Image reconstructions were carried out on an Inveon Acquisition Workplace (Siemens Preclinical Solutions) workstation using an ordered subset expectation maximization 3D/maximum a posteriori reconstruction algorithm.

Quantitative region-of-interest (ROI) analysis of the PET images was performed on attenuation and decay corrected PET images using Inveon Research Workplace 4.2 (Siemens Preclinical Solutions). Tissue uptake values are presented as the percent injected dose per cc (%ID/cc). The spatial resolution of the Inveon PET scanner is 1.4 mm and a previous study showed that %recovery coefficient was 91% for 3 mm-size rod (42). As such, it is suggested that the tracer uptake in target of interest with <3 mm is likely to be underestimated. The maximum value in the targeted region of interest was calculated of day 1 to 5 scans to observe the tracer distribution change over time. Bilateral cervical and axillary lymph nodes plus spleen were regarded as representative lymphoid organs in this study as vaccine was injected near left axillary area and the tumor draining lymph node (TLDN) was right cervical lymph node in our mice. The maximum uptake value of these sites was collected together with representative solid organs such as the heart, the liver, the muscle, and the bilateral brain hemispheres. ROI was drawn to avoid the tracer uptake from surrounding organs referring to corresponding CT scan, when they were intensely seen. The volumetric information of whole-body lymphoid organs was incorporated for the final day 5 scan to observe the total body OX40 activity.

To accurately quantify the extent of tracer binding, we employed a method that has been commonly researched as an index of treatment response in human FDG-PET scans: total lesion glycolysis (TLG; ref. 43). TLG is a multiplication of the metabolic volume which shows pixels above a defined threshold uptake (42% of maximum uptake is the most frequently applied threshold in clinical studies; ref. 44) and mean uptake value of the volume. In our study, we defined the multiplication of the volume above 42% of maximum uptake and mean uptake value of the volume as “total lesion immune response” (TLIR; Supplementary Fig. S1). Once volume of interest (VOI) was set to a targeted lesion, the dedicated software automatically calculated the volume above 42% of maximum uptake, mean uptake in the calculated volume, and their multiplication. This analysis was performed on OsiriX MD version 8.0.1.

Ex vivo studies

Immediately after the day 5 scan, mice were euthanized and blood, tumor, lymph nodes, and all major organ/tissues were collected and weighed. The radioactivity of each tissue was counted in an automated γ-counter (HIDEX), and the tissue accumulations were calculated as %ID/g (mean ± SEM). Flow cytometry was performed to the tumor, lymph nodes, and spleen in several mice immediately after biodistribution for analysis of OX40/CD4 expression.

FACS analyses

Cells were first stained with Live/Dead aqua fixable cell stain (Molecular Probes L34957) following the manufacturer's instructions. Cells were then washed in FACS buffer (PBS and 2% BSA) and stained with a panel of antibodies including CD3-APC (hamster IgG1 k, clone:145-2C11, BD Biosciences), CD4-APC-Cy7 (rat IgG2b k, clone GK1.5:, BioLegend), CD8-PE (rat IgG2a k, clone: 53-6.7, BioLegend), and OX40-BV421 (rat IgG1 k, clone: OX-86, BioLegend). Cells were fixed in 2% paraformaldehyde and stored at 4°C prior to analysis using an LSR II flow cytometer (BD Biosciences). Compensation was performed utilizing control beads (Thermo Fisher Scientific) and data were analyzed using the FlowJo software.

Autoradiography and MRI

To see the uptake of 89Zr-DFO-OX40 mAb tracer reflects the exact brain tumor, brains of a few mice were compared with autoradiograph (ARG) and MRI. To obtain ARG images, mice brains were fresh frozen in optimum cutting temperature compound (O.C.T., Sakura Finetek Inc.) and sliced at 40-μm thickness using a cryostat (Microm). Tissues were exposed to an Imaging Plate (Fujifilm) and kept at −20°C in the dark for 2 weeks prior to scanning using a Typhoon phosphorimager (Amersham Biosciences). MRI T2-weighted images (echo time: 33 ms; retention time: 2,500 ms; 2 averages; 17 slices) were obtained following PET on day 2 using a 7T Varian Magnex Scientific MR system under 1.5% to 2.0% of isoflurane in anesthetized mice as reported previously (45). Fused images were created with Inveon Research Workplace 4.2.

Statistical analysis

The comparison of 89Zr-OX40 mAb uptake between treated and vehicle, or uptake of 89Zr-DFO-OX40 mAb versus isotype control over repeated PET scans, two-way ANOVA was applied as an analyzing method of repeated measures data. Mann–Whitney U test was used in comparison between vaccinated and control groups in terms of the spleen weight, FACS analysis, and best treatment response ratio in BLI. This method was also applied to compare best treatment response ration of BLI and TLIR in low/high–tumor burden classes. Spearman rank correlation test was applied for relationship between PET signals and biodistribution measurements or BLI signal or BLI ratio. GraphPad Prism 8.0.2 (RRID:SCR_002798, GraphPad Software) and R 3.5.1 were used. Two-sided P values under 0.05 was considered as statistically significant.

ImmunoPET enables visualization of stimulated OX40-positive lymphoid organs

Our conceptual study design is described in Fig. 1A. In brief, we prepared orthotopic glioma mice and subsequently imaged them using immunoPET to assess the spatiotemporal dynamics of T-cell activation following administration of a cancer vaccine at a tumor remote site (left shoulder). This strategy is described in Fig. 1B. For 10 to 14 days following glioma inoculation, tumor growth was monitored using BLI. When tumor growth was confirmed with a growing signal of BLI twice within 4 days and a signal above 105 (p/sec/cm2/sr) at second BLI, mice received a combined subcutaneous vaccination of CpG-ODN and glioma tumor lysates, and OX40 mAb, 5 and 6 days respectively, prior to the administration of 89Zr-DFO-OX40 mAb immunoPET tracer. After injection of 89Zr-DFO-OX40 mAb via tail vein, mice underwent PET/CT scans on days 1, 2, and 5. As expected, vaccinated mice exhibited marked 89Zr-DFO-OX40 mAb uptake in multiple lymph nodes (LN) 5 days after tracer administration, which was not seen in control mice, whereas intense tracer uptake was revealed in the brain tumor in controls (Fig. 1C; Supplementary Videos S1 and S2). Vaccinated mice showed enhanced PET signal not only in the left axillary LN, which is the draining lymph node from the vaccinated site, but also in other remote areas including the right cervical LN, which is regarded as the TDLN of inoculated glioma in the ipsilateral hemisphere. This result is in agreement with previous reports that suggest antigens transferred to both superficial and deep areas of the brain drain to cervical lymph nodes (46, 47). The representative time course of 89Zr-DFO-OX40 mAb PET signal in regions of interest from days 1 to 5 in vaccinated and control mice is shown in Fig. 1D. Bilateral lymphatic chains were exhibited in the vaccinated mice, whereas the control mouse showed a substantial tumor in the brain. We considered these organs to play a major role in immune reaction against the tumor in our model: left axillary LN as a treated site, right cervical LN as a TDLN, contralateral sites as routes of migration of activated T cells, and the spleen as a reflection of enhanced systemic T-cell activity. PET uptake in these lymph nodes and the spleen was compared between groups as well as that of the brain tumor (Fig. 1E). The highest PET signal was observed in the left axillary LN and the spleen and was significantly elevated in vaccinated (left axillary LN, P < 0.0001; spleen, P = 0.020) compared with control mice on day 5. In particular, the left axillary LN demonstrated a nearly 3-fold uptake in the vaccinated group relative to control group. Uptake in right cervical LN as a TDLN also showed higher uptake compared with control, while the brain tumor uptake was significantly higher in the control (P = 0.042). Significant splenomegaly was also seen in vaccinated mice (P = 0.019; Fig. 1F). Correlogram of 89Zr-DFO-OX40 mAb accumulation in individual mice revealed that the LN that is closest to the vaccination site exhibited the highest OX40 mAb signals, followed by the spleen, TDLN, right axillary LN, and left cervical LN (Fig. 1G).

Figure 1.

89Zr-DFO-OX40 mAb PET scan revealed stimulated lymphoid organs with cancer vaccination. A, Conceptual sketch of this study. After vaccination therapy in the left shoulder for mice bearing glioma in the right hemisphere, immunoPET with 89Zr-DFO-OX40 mAb was performed to visualize the stimulated immune system, especially focusing on lymph node (LN) of vaccinated area and tumor draining lymph node (TDLN), as well as treatment effect. B, Experimental schedule. After intracranial injection of glioma cells to right hemisphere, mice were monitored by BLI for 10 to 14 days. CpG + tumor lysate and OX40 mAb were injected subcutaneously to left shoulder 5 days and 6 days prior to injection of 89Zr-DFO-OX40 mAb, respectively. PET scans were sequentially acquired on days 1, 2, and 5 thereafter. C, Representative 89Zr-DFO-OX40 mAb PET/CT scan (day 5) of vaccinated and control mice. Whole-body fused maximum intensity projection (MIP) image shows multiple lymphatic organs including left axillary LN (vaccinated LN), right cervical LN (TDLN), and the spleen. Control mouse did not show any stimulated LNs but instead a large tumor in the right hemisphere. Corresponding axial fusion images verify the exact location of tracer accumulation in each site. D, Representative 3D fused PET/CT scans of vaccinated and control mice. Multiple LNs and spleen are clearly visualized from day 2 (arrows). Vehicle demonstrated a large tumor in the right brain on day 5 (arrowhead). E, Time course of the highest PET signal of 89Zr-DFO-OX40 mAb in bilateral axillary, cervical LNs, the spleen, and the brain tumor. Compared with the control group, left axillary LN (vaccinated; P < 0.0001) and the spleen (P = 0.020) showed significantly higher accumulation in vaccinated group on day 5, followed by right cervical LN (TDLN) and right axillary LN with nearly significance. PET signal in the brain tumor was significantly lower in the vaccinated group on day 5 (P = 0.042). Data, mean ± SEM. F, Spleen volume comparison between vaccinated and control groups. In vaccinated group, significant splenomegaly was observed (P = 0.019). G, Correlogram of 89Zr-DFO-OX40 mAb accumulation on day 5. Each color box indicates the PET signal strength from individual mouse. The difference of mean PET signal between vaccinated and control group was larger in this order: left axillary LN (vaccinated) > spleen > right cervical (TDLN) > right axillary LN > left axillary LN. *, P < 0.05; **, P < 0.01; ***, P < 0.001; ****, P < 0.0001.

Figure 1.

89Zr-DFO-OX40 mAb PET scan revealed stimulated lymphoid organs with cancer vaccination. A, Conceptual sketch of this study. After vaccination therapy in the left shoulder for mice bearing glioma in the right hemisphere, immunoPET with 89Zr-DFO-OX40 mAb was performed to visualize the stimulated immune system, especially focusing on lymph node (LN) of vaccinated area and tumor draining lymph node (TDLN), as well as treatment effect. B, Experimental schedule. After intracranial injection of glioma cells to right hemisphere, mice were monitored by BLI for 10 to 14 days. CpG + tumor lysate and OX40 mAb were injected subcutaneously to left shoulder 5 days and 6 days prior to injection of 89Zr-DFO-OX40 mAb, respectively. PET scans were sequentially acquired on days 1, 2, and 5 thereafter. C, Representative 89Zr-DFO-OX40 mAb PET/CT scan (day 5) of vaccinated and control mice. Whole-body fused maximum intensity projection (MIP) image shows multiple lymphatic organs including left axillary LN (vaccinated LN), right cervical LN (TDLN), and the spleen. Control mouse did not show any stimulated LNs but instead a large tumor in the right hemisphere. Corresponding axial fusion images verify the exact location of tracer accumulation in each site. D, Representative 3D fused PET/CT scans of vaccinated and control mice. Multiple LNs and spleen are clearly visualized from day 2 (arrows). Vehicle demonstrated a large tumor in the right brain on day 5 (arrowhead). E, Time course of the highest PET signal of 89Zr-DFO-OX40 mAb in bilateral axillary, cervical LNs, the spleen, and the brain tumor. Compared with the control group, left axillary LN (vaccinated; P < 0.0001) and the spleen (P = 0.020) showed significantly higher accumulation in vaccinated group on day 5, followed by right cervical LN (TDLN) and right axillary LN with nearly significance. PET signal in the brain tumor was significantly lower in the vaccinated group on day 5 (P = 0.042). Data, mean ± SEM. F, Spleen volume comparison between vaccinated and control groups. In vaccinated group, significant splenomegaly was observed (P = 0.019). G, Correlogram of 89Zr-DFO-OX40 mAb accumulation on day 5. Each color box indicates the PET signal strength from individual mouse. The difference of mean PET signal between vaccinated and control group was larger in this order: left axillary LN (vaccinated) > spleen > right cervical (TDLN) > right axillary LN > left axillary LN. *, P < 0.05; **, P < 0.01; ***, P < 0.001; ****, P < 0.0001.

Close modal

Next, we examined whether 89Zr-DFO-OX40 mAb-PET imaging could detect immune activation after vaccination at earlier time points (Supplementary Fig. S1). To investigate this, we administered 89Zr-DFO-OX40 mAb to orthotopic GBM mice the day after vaccination, and performed three subsequent PET scans 1 to 4 days following tracer injection. Interestingly, neither vaccinated nor control group showed enhanced PET signal in lymphoid organs until day 4 (P > 0.05 in all cases).

OX40 immunoPET signal is highly specific for OX40+ CD4+ T cells

To confirm that PET-positive lymphoid organs was due to presence of OX40+ cells, flow cytometry was performed on the left axillary LN, left cervical LN, spleen, and tumor (Fig. 2A). No significant difference in CD4 expression was observed in any organs between vaccinated and control groups. However, OX40+ CD4+ T cells were significantly increased in the left axillary LN (P = 0.017), left cervical LN (P = 0.036), and the spleen (P = 0.036) of vaccinated mice (Fig. 2B), suggesting that OX40 PET signal corresponds to the number of OX40+ CD4+ T cells. No major difference in OX40 expression was seen in the tumor (P > 0.05).

Figure 2.

FACS analysis in left axillary LN, left cervical LN, the spleen, and the brain tumor. A, CD4 and CD8 gated windows. No significant difference was seen in the total CD4-positive population ratio between vaccinated and control groups. B, OX40 and CD4 gated windows. OX40-positive CD4 cells were significantly seen more in vaccinated group in left axillary LN (P = 0.017), left cervical LN (P = 0.036), and the spleen (P = 0.036). No significant difference was observed in the tumor. Data, mean ± SEM. *, P < 0.05.

Figure 2.

FACS analysis in left axillary LN, left cervical LN, the spleen, and the brain tumor. A, CD4 and CD8 gated windows. No significant difference was seen in the total CD4-positive population ratio between vaccinated and control groups. B, OX40 and CD4 gated windows. OX40-positive CD4 cells were significantly seen more in vaccinated group in left axillary LN (P = 0.017), left cervical LN (P = 0.036), and the spleen (P = 0.036). No significant difference was observed in the tumor. Data, mean ± SEM. *, P < 0.05.

Close modal

To further assess the specificity of 89Zr-DFO-OX40 mAb and its binding in lymphoid organs, immunoPET using an 89Zr-labeled isotype control antibody (89Zr-DFO-rat IgG1 mAb) was performed. 89Zr-DFO-OX40 mAb showed significantly higher uptake in bilateral axillary LNs (left, P = 0.0083; right, P = 0.028), and the spleen (P = 0.026) toward day 5, compared with 89Zr-labeled isotype control antibody (Fig. 3). This elevated uptake in LNs between 89Zr-DFO-OX40 mAb and isotype control was not observed in control mice. In the spleen, nonspecific uptake of isotype control was observed in both vaccinated (7.2 ± 1.5 %ID/cc) and control mice (6.4 ± 0.6 %ID/cc); however, it was on average less than half and enough to be differentiated from significant 89Zr-DFO-OX40 mAb uptake in the vaccinated mice (16.7 ± 3.2 %ID/cc). These results indicate that the 89Zr-DFO-OX40 mAb PET signal is specifically visualizing OX40 expression within lymphoid organs. Liver uptake was significantly higher (P = 0.016) and heart showed trend of less uptake on the day 5 scan in 89Zr-DFO-OX40 mAb PET, compared with the isotype control in the vaccinated group, possibly suggesting faster clearance of 89Zr-DFO-OX40 mAb tracer upon vaccination. As for the tumor (right brain) and contralateral (left) brain tissue, no major difference was observed between 89Zr-DFO-OX40 mAb and isotype PET signals in both vaccinated and control groups although in the control group higher mean 89Zr-DFO-OX40 mAb uptake was observed relative to isotype control in the tumor. Biodistribution studies revealed similar trends; the left axillary LN exhibited significantly higher 89Zr-DFO-OX40 mAb signal in vaccinated mice than isotype control (P < 0.0001), and the spleen also showed same tendency (no significance). Liver count was higher and blood count was lower in 89Zr-DFO-OX40 mAb group of the vaccinated mice, suggesting 89Zr-DFO-OX40 mAb distribution kinetics are more rapid within vaccinated animals (vs. nonvaccinated) and compared with the kinetics of the isotype control tracer. PET signal was confirmed to be well correlated with counts from biodistribution in both solid organs and lymph nodes (R2 = 0.83 and 0.55, respectively).

Figure 3.

Specificity of 89Zr-DFO-OX40 mAb PET in lymphoid organs. A, Representative PET images of 89Zr-DFO-OX40 mAb (mAb) and 89Zr-DFO-rat IgG (isotype control), in both vaccinated and control mice, on days 1, 2, and 5. Lymphoid organs were vividly delineated in vaccinated group (arrows) while this was less clear with isotype tracer. B, Time course of 89Zr-DFO-OX40 mAb and 89Zr-DFO-rat IgG mAb (isotype control) in each lymphoid organ and main solid organs. Bilateral axillary LNs and the spleen showed higher uptake of 89Zr-DFO-OX40 mAb than isotype control in vaccinated group. All lymphoid organs showed soaring curves over time with 89Zr-DFO-OX40 mAb in vaccinated group, which was seen neither with isotype control nor non-vaccinated group (left axillary node, P = 0.0083; right axillary node, P = 0.028; spleen, P = 0.026). Heart uptake, which represents blood, showed higher uptake on day 1 with 89Zr-DFO-OX40 mAb compared with isotype control tracer in both vaccinated and control groups, but this phenomenon disappears over time. Liver uptake on day 5 scan was higher with 89Zr-DFO-OX40 mAb in vaccinated group than the control group. C, Biodistribution results after day 5 scan. 89Zr-DFO-OX40 mAb showed significantly higher counts in the liver, spleen, and left axillary LN in vaccinated group than control, while blood count was significantly lower in the same manner. Splenic count was significantly higher in vaccinated mice than control in both 89Zr-DFO-OX40 and isotype group. D, Correlation between PET and biodistribution results. Strong correlations were seen in both solid organs and lymph nodes. Data are shown with mean ± SEM. *, P < 0.05; **, P < 0.01; ***, P < 0.001; ****, P < 0.0001.

Figure 3.

Specificity of 89Zr-DFO-OX40 mAb PET in lymphoid organs. A, Representative PET images of 89Zr-DFO-OX40 mAb (mAb) and 89Zr-DFO-rat IgG (isotype control), in both vaccinated and control mice, on days 1, 2, and 5. Lymphoid organs were vividly delineated in vaccinated group (arrows) while this was less clear with isotype tracer. B, Time course of 89Zr-DFO-OX40 mAb and 89Zr-DFO-rat IgG mAb (isotype control) in each lymphoid organ and main solid organs. Bilateral axillary LNs and the spleen showed higher uptake of 89Zr-DFO-OX40 mAb than isotype control in vaccinated group. All lymphoid organs showed soaring curves over time with 89Zr-DFO-OX40 mAb in vaccinated group, which was seen neither with isotype control nor non-vaccinated group (left axillary node, P = 0.0083; right axillary node, P = 0.028; spleen, P = 0.026). Heart uptake, which represents blood, showed higher uptake on day 1 with 89Zr-DFO-OX40 mAb compared with isotype control tracer in both vaccinated and control groups, but this phenomenon disappears over time. Liver uptake on day 5 scan was higher with 89Zr-DFO-OX40 mAb in vaccinated group than the control group. C, Biodistribution results after day 5 scan. 89Zr-DFO-OX40 mAb showed significantly higher counts in the liver, spleen, and left axillary LN in vaccinated group than control, while blood count was significantly lower in the same manner. Splenic count was significantly higher in vaccinated mice than control in both 89Zr-DFO-OX40 and isotype group. D, Correlation between PET and biodistribution results. Strong correlations were seen in both solid organs and lymph nodes. Data are shown with mean ± SEM. *, P < 0.05; **, P < 0.01; ***, P < 0.001; ****, P < 0.0001.

Close modal

89Zr-DFO-OX40 mAb binding corresponds with glioma location

To confirm that the location of 89Zr-DFO-OX40 mAb uptake in the brain corresponded to inoculated glioma, coronal PET/CT was compared with MRI on day 2 and ARG on day 5. PET signal corresponded with the exact location of tumor as confirmed by MRI and ARG (Fig. 4). PET signal in the tumor on day 5 also correlated with the BLI signal of the right hemisphere in both vaccinated (R2 = 0.44) and control mice (R2 = 0.85). These results indicate that 89Zr-DFO-OX40 mAb accumulated in the glioma itself as well. This may be attributed to blood brain barrier (BBB) permeability, and the extent of tracer accumulation might depend on the actual tumor volume.

Figure 4.

89Zr-DFO-OX40 mAb accumulation in the brain tumor was verified by MRI (A) and ARG (B). A, Coronal scan of 89Zr-DFO-OX40 mAb PET/CT (left), T2WI MRI/CT (right), and PET/MRI/CT fusion (middle) on day 2 after tracer injection. B, Coronal scan of 89Zr-DFO-OX40 mAb PET/CT on day 5 was compared with ARG of the same day. Top row, good response with vaccination; bottom row, huge tumor in control group. In the vaccinated mice, both PET and ARG showed small signals at the right bottom of the skull, while the mice who did not receive vaccine or the vaccine effect was not insufficient, large tumor was visualized in the colocalized right hemisphere. Top, vaccinated; bottom, vehicle. C, Correlation between BLI signals in the brain tumor and 89Zr-DFO-OX40 mAb accumulation in PET on day 5. Significant correlation was seen in both treated (n = 15) and vehicle-treated mice (n = 11).

Figure 4.

89Zr-DFO-OX40 mAb accumulation in the brain tumor was verified by MRI (A) and ARG (B). A, Coronal scan of 89Zr-DFO-OX40 mAb PET/CT (left), T2WI MRI/CT (right), and PET/MRI/CT fusion (middle) on day 2 after tracer injection. B, Coronal scan of 89Zr-DFO-OX40 mAb PET/CT on day 5 was compared with ARG of the same day. Top row, good response with vaccination; bottom row, huge tumor in control group. In the vaccinated mice, both PET and ARG showed small signals at the right bottom of the skull, while the mice who did not receive vaccine or the vaccine effect was not insufficient, large tumor was visualized in the colocalized right hemisphere. Top, vaccinated; bottom, vehicle. C, Correlation between BLI signals in the brain tumor and 89Zr-DFO-OX40 mAb accumulation in PET on day 5. Significant correlation was seen in both treated (n = 15) and vehicle-treated mice (n = 11).

Close modal

Treatment effect after cancer vaccination

Brain tumor burden was monitored with BLI after a combined vaccination. Representative images of BLI from treatment days 0 to 8 are shown in Fig. 5A. We divided the mice into two classes according to the tumor burden on the day of vaccination: low burden with mean signal of lower than 106 (p/sec/cm2/sr; vaccinated, n = 12; control, n = 9), and high burden with higher than 106 (p/sec/cm2/sr; vaccinated, n = 9; control, n = 7). Treatment response was most effective in the low-burden class with one third of vaccinated mice (n = 4) demonstrating >90% decrease in tumor volume, which was not observed in the control mice (Fig. 5B). In contrast, vaccinated mice in the high-burden class exhibited <25% decrease in tumor signal. Accordingly, treatment response was found to be significant in low-burden (P = 0.015) but not in high-burden mice when compared with control mice (Fig. 5C). In addition, we investigated whether administering 89Zr-DFO-OX40 mAb affected treatment response in any way. Importantly, there was no significant difference in BLI signal change between mice that received 89Zr-DFO-OX40 mAb tracer dose injection (20 μg antibody) with those that received the isotype control mAb (Fig. 5D).

Figure 5.

Tumor size tracking with BLI to see therapeutic effect of cancer vaccine. A, Representative BLI images on 0, 4, and 8 days after cancer vaccination. Top left, vaccinated, low tumor burden [<106 (p/sec/cm2/sr)]; bottom left, vaccinated, high tumor burden [> 106 (p/sec/cm2/sr)]; top right, control, low burden; bottom right, control, high burden. B, Waterfall plot of best treatment response (the ratio of lowest signal after vaccination to baseline in BLI) in classes of low and high burden. C, Comparison between vaccinated and control groups in both low- and high-burden classes. In low-burden mice, vaccinated mice indicated significant shrinking of the brain tumor with BLI (P = 0.015). Data are shown with mean ± SEM. D, Time course of BLI signals after vaccination in 89Zr-DFO-OX40 mAb PET group and isotype control PET group of low tumor burden. There was no significant difference in tumor signals between 89Zr-DFO-OX40 mAb and isotype control, indicating 89Zr-DFO-OX40 mAb itself does not affect the treatment response. *, P < 0.05.

Figure 5.

Tumor size tracking with BLI to see therapeutic effect of cancer vaccine. A, Representative BLI images on 0, 4, and 8 days after cancer vaccination. Top left, vaccinated, low tumor burden [<106 (p/sec/cm2/sr)]; bottom left, vaccinated, high tumor burden [> 106 (p/sec/cm2/sr)]; top right, control, low burden; bottom right, control, high burden. B, Waterfall plot of best treatment response (the ratio of lowest signal after vaccination to baseline in BLI) in classes of low and high burden. C, Comparison between vaccinated and control groups in both low- and high-burden classes. In low-burden mice, vaccinated mice indicated significant shrinking of the brain tumor with BLI (P = 0.015). Data are shown with mean ± SEM. D, Time course of BLI signals after vaccination in 89Zr-DFO-OX40 mAb PET group and isotype control PET group of low tumor burden. There was no significant difference in tumor signals between 89Zr-DFO-OX40 mAb and isotype control, indicating 89Zr-DFO-OX40 mAb itself does not affect the treatment response. *, P < 0.05.

Close modal

Lower tumor burden was effectively treated with less T-cell activation in lymphoid organs

Because we observed that vaccinated mice showed distinct patterns of 89Zr-DFO-OX40 mAb binding in lymphoid organs (LNs and the spleen), we compared total 89Zr-DFO-OX40 mAb accumulation in lymphoid organs of vaccinated mice with either low (n = 4) and high tumor burden (n = 5). An anatomic depiction of LNs and other lymphoid tissues is shown in Fig. 6A. We observed enhanced PET signal in brachial and inguinal LNs in several vaccinated mice. 89Zr-DFO-OX40 mAb PET MIP images of mice on day 5 are shown in Fig. 6B. All mice exhibited more than two visually detectable LNs and/or spleen, which indicates that the vaccine consistently works by stimulating lymphoid organs. To quantify the strength of the immune response, we introduced an index named TLIR, which was defined as a mixed index of volume and intensity of 89Zr-DFO-OX40 mAb PET signal (Supplementary Fig. S2). TLIR in bilateral LNs of axillary, brachial, cervical, and inguinal area, and the spleen were calculated and compared between low– and high–tumor burden groups (Fig. 6C). TLIR was significantly higher in the spleen (P = 0.032) and right inguinal LN (P = 0.016) of high-burden mice, while TLIR of right cervical LN (TDLN) was higher on average for low-burden mice. TLIR ratio of TDLN/other lymphoid organs was 0.27 and 0.08 in low- and high-burden mice, respectively. The sum of TLIR and percentage decrease of BLI signal per individual mouse are shown in Fig. 6D as indicators of whole-body OX40-powered lymphatic tissue and treatment response of vaccine. Within the low–tumor burden group, two out of four mice had an almost complete response (≥99% decrease in BLI signal) along with whole-body (WB)-TLIR > 1,500 × 10−3 %ID. In contrast, only one mouse showed a weak treatment response (i.e., 19% decrease). Nevertheless, all mice exhibited >2,000 × 10–3 %ID of WB-TLIR in the high–tumor burden group of mice. Taken together, these results demonstrate that when the tumor burden is low, even a modest amount of OX40+ activated T cells in lymphatic tissues is expected to assure a favorable treatment response after a single shot of mixed vaccine. Alternatively, if the tumor burden is already high at the time of vaccination, even a large systemic OX40-positive immune response is not sufficient to induce tumor shrinkage.

Figure 6.

Evaluating the therapeutic effect of vaccination with 89Zr-DFO-OX40 mAb PET/CT. A, Whole-body lymphoid organs potentially stimulated by vaccine. B, MIP images of 89Zr-DFO-OX40 mAb PET/CT in low tumor burden (n = 4) and high tumor burden (n = 5; right). C, TLIR in 89Zr-DFO-OX40 mAb PET in each stimulated lymphoid organs of vaccinated mice (bilateral axillary, brachial, cervical, inguinal LNs, and the spleen). Significantly higher uptake was seen in the spleen and right inguinal LN in high-burden group. D, WB-TLIR and percentage decrease of BLI signal in low and high burden of vaccinated mice. In low-burden group, higher WB-TLIR (>1,500 × 10–3 %ID) was associated with good treatment response (≥99%), whereas only one mouse exhibited weak treatment effect (<20%) in high-burden group despite moderate to intense immune response (>2,000 × 10–3 %ID). Data are shown with mean ± SEM. E, Correlation of 89Zr-DFO-OX40 mAb accumulation and BLI ratio as treatment response in low- and high-burden mice. Left axillary LN (vaccinated) showed a good negative correlation in low-burden group, whereas no correlation was seen in high-burden group. *, P < 0.05.

Figure 6.

Evaluating the therapeutic effect of vaccination with 89Zr-DFO-OX40 mAb PET/CT. A, Whole-body lymphoid organs potentially stimulated by vaccine. B, MIP images of 89Zr-DFO-OX40 mAb PET/CT in low tumor burden (n = 4) and high tumor burden (n = 5; right). C, TLIR in 89Zr-DFO-OX40 mAb PET in each stimulated lymphoid organs of vaccinated mice (bilateral axillary, brachial, cervical, inguinal LNs, and the spleen). Significantly higher uptake was seen in the spleen and right inguinal LN in high-burden group. D, WB-TLIR and percentage decrease of BLI signal in low and high burden of vaccinated mice. In low-burden group, higher WB-TLIR (>1,500 × 10–3 %ID) was associated with good treatment response (≥99%), whereas only one mouse exhibited weak treatment effect (<20%) in high-burden group despite moderate to intense immune response (>2,000 × 10–3 %ID). Data are shown with mean ± SEM. E, Correlation of 89Zr-DFO-OX40 mAb accumulation and BLI ratio as treatment response in low- and high-burden mice. Left axillary LN (vaccinated) showed a good negative correlation in low-burden group, whereas no correlation was seen in high-burden group. *, P < 0.05.

Close modal

Elevated 89Zr-DFO-OX40 mAb PET signal in vaccinated LN is a predictor of treatment response

To evaluate whether LN response can predict vaccine treatment response, correlation between the uptake of 89Zr-DFO-OX40 mAb on day 5 in each bilateral axillary LN/cervical LN/spleen and the best treatment response was calculated according to low– and high–tumor burden groups (Fig. 6A). In the low-burden class, a significant correlation was observed in left axillary LN (r = −0.94, P = 0.017), which was the closest LN from vaccination site, while the other LNs and the spleen did not show any correlation. No correlation was seen in the high-burden class at all sites. This indicates that 89Zr-DFO-OX40 mAb accumulation in the vaccinated site can be utilized as a treatment predictor in the context of this vaccine treatment strategy.

The difficulties of treating GBM have motivated the investigation of different promising immunotherapies and new combination treatment strategies. As such, antitumor effects of therapeutics targeting various immune-cell surface markers are being assessed. OX40 is one such promising target for T-cell activating agonistic antibodies, together with 4-1BB, GITR, ICOS, CD27, and CD30 (4, 48). Several anti-OX40 agonists are already being implemented in phase I or II cancer clinical trials (49). However, there are fewer tools under investigation to directly image the expression of these immune-cell markers; thus, the evaluation of immunotherapy efficacy has been limited. We previously reported the successful development of two highly specific immune system visualization techniques. The first involved the generation of 64Cu-DOTA-OX40 mAb, which we showed was sensitive and specific for imaging OX40+ activated T cells in a mouse model of syngeneic lymphoma (39) and acute graft-versus-host disease (50) using PET. The second involved the development of a PET tracer, known as 89Zr-DFO-ICOS mAb, for detecting ICOS+ T cells in a murine lung cancer model (51) and CAR T cells (52). Following these promising results, we extended our pursuit of imaging T-cell activation markers to a more challenging type of tumor, GBM, and set out to quantify the characteristics of activated immune cells in the whole body after remote vaccination.

The current work demonstrates that 89Zr-DFO-OX40 mAb PET specifically delineates stimulated lymphoid organs following administration of a cancer vaccine applied in an orthotopic GBM model. After receiving combined cancer vaccine in the remote site, multiple LNs and spleen, particularly the LN proximal to the vaccination site, exhibited robust 89Zr-DFO-OX40 mAb PET signals, reflecting an increased number of CD4+ OX40+ T cells. When GBM burden was low, a single pair of mixed vaccines led to a significant decrease in the tumor signal in BLI while requiring less boost of immune stimulation indicated by TLIR in whole-body lymphoid organs. Of note, the tumor shrinkage was correlated with the 89Zr-DFO-OX40 mAb PET signals at LN nearest to the vaccinated site.

Consequently, immunoPET with 89Zr-DFO-OX40 mAb was quite successful in visualizing stimulated LNs and the spleen, with a combined vaccination. Five days after tracer injection, all vaccinated mice showed vivid 89Zr-DFO-OX40 mAb uptake in the vaccinated LN with a high contrast to blood pool. Accumulation within other LNs and the spleen varied among individual mice, but overall, every animal exhibited multiple hot regions of marked tracer uptake within lymphoid organs (i.e., lymph nodes and/or spleen). This directly shows that the immune activation triggered by the remote vaccination not only occurred at the vaccinated site but also systemically. Calzascia and colleagues demonstrated that the anatomic location in which antigen-specific lymphocytes are activated may “imprint” their ability to traffic to their ultimate destination (53); specifically, naïve T cells activated in the cervical lymph nodes demonstrate a “central nervous system homing” phenotype that may be influenced by T-cell expression of the α4β1 integrin (54). In this study, we found that the vaccination induced a trend toward higher 89Zr-DFO-OX40 mAb uptake in tumor-draining LN, following by the nearest LN from vaccinated site and the spleen. In addition, we observed that a single vaccination worked efficiently in mice with low-burden GBM with less total volume of immune activity compared with large tumors by calculating WB-TLIR. Conversely, when a tumor was large [>106 (p/sec/cm2/sr)], little tumor shrinkage was induced nevertheless even stronger immune reaction quantified as TLIR was introduced, especially at the spleen and remote LNs from the brain such as inguinal LN, as shown in 89Zr-DFO-OX40 mAb PET. TDLN was the only lymphoid organ that demonstrated higher 89Zr-DFO-OX40 mAb uptake in the low–tumor burden group, compared with high–tumor burden group. The reason of the higher uptake in TDLN in the low–tumor burden group is potentially explained as follows. Low tumor burden showed a significant response to immunotherapy, which was in line with previous clinical studies. That is, the low baseline tumor volume was associated with better overall survival in immune checkpoint blockade users with other cancers such as melanoma (55) and non–small cell cancer (56). Moreover, PD-1 inhibitor pembrolizumab brought survival benefit to recurrent patients with GBM when the tumor burden was reduced with surgical resection (57). These clinical studies suggest that the smaller tumor volume is related to better response to immunotherapy. On the other hand, recent evidence points to the need for the recruitment of newly primed and peripherally (e.g., in TDLN) expanded effector T cells to ensure efficacy of immunotherapy (58). Clinical efficacy and durability of antitumor immunity were reported a relation with elevated frequencies of central-memory or early-effector T cells with the ability to home to lymph nodes (59). These reports imply the accumulation of primed cytotoxic OX40+ T cells in TDLN is a factor for good therapy response. Taken altogether, we consider low–tumor burden group showed higher uptake in TDLN because both factors were associated with responders. However, this result is of limited value because there was a large overlap in TDLN uptake between low– and high–tumor burden groups without any significance. We should validate if the responders truly show high 89Zr-DFO-OX40 mAb uptake in TDLN in a larger number of cohorts in further studies. We also briefly showed the correlation of higher 89Zr-DFO-OX40 mAb uptake in the LN nearest from the vaccinated site and the better treatment response when the tumor was small. We assume this correlation implies that the PET signals at the vaccinated LN reflect the combined effect of T-cell priming and migration to the tumor sites. Conversely, in nonresponders, the vaccination stimulated the lymphoid organs but supposedly failed in priming and clonal expansion of T cells sufficiently. Considering above, vaccination may be preferred at the tumor-draining LN to facilitate the priming of T cells for the best therapeutic response, although we need to verify this in future studies. However, it is meaningful to confirm that the vaccination at a remote LN was effective, as the tumor-draining LN is not always accessible in the clinical practice, especially in cases of GBM. All in all, our data indicate that the strongest immune response in the whole body occurs at the nearest LN from the vaccinated site, which may help to predict the real treatment response. These phenomena might be a potential reference for future perspective of how immunotherapies help contribute to cancer therapies.

Several limitations are included in this study. We utilized only the GL26 glioma cells that were labeled with the Luc2-eGFP reporter to avoid the issue of immunogenicity. This subline does not respond to innate immune system that may affect the interpretation of acquired data. Another limitation of this study was that the remote vaccination site was fixed at the subcutaneous contralateral shoulder. If the vaccination was tried in other parts of the body, 89Zr-DFO-OX40 mAb PET may have shown a different distribution of enhanced OX40-positive lymphoid organs. Moreover, we did not test different doses or optimize the number of shots of the vaccination protocol or conduct a follow-up study to assess overall survival, as it takes several days to observe animals with PET while the tumor is rather progressive when the initial vaccination was not effective. Consequently, the treatment effect was limited to the “low” tumor burden group, and it was not explored if the higher tumor burden group responds to more intensive treatment regimens. Furthermore, direct evaluation of intratumoral lymphocytes in between responders and nonresponders were unachievable due to the rapid shrinkage of tumor in responders; therefore, we could not corroborate the response resulting from the migrated cytotoxic T cells. Finally, we included OX40 mAb in the vaccination regimen but did not verify in this study if our OX40 imaging approach could also be employed to monitor other types of vaccination; however, OX40-targeted immunoPET will be implemented in other vaccination regimens without OX40 mAb for other diseases, considering our previous study (39) showing the clear 64Cu-DOTA-OX40 mAb uptake in the lymphoma in in situ CpG treatment group, followed by tumor shrinkage. Validation of OX40 PET imaging is warranted in future studies. The aim of this study was focused on evaluating the utility of OX40 PET imaging to visualize how the systemic lymphoid organs respond following vaccination and how that response predicts and contributes to the therapeutic effect in a murine glioma model. Further analysis will be required to confirm the optimal strategy in the context of vaccination therapy.

As OX40 imaging was capable of monitoring lymphoma model without OX40 mAb vaccination in our previous study (39), this imaging approach is expected to be applied to various clinical occasions including the monitoring of other types of vaccination to malignancy, but also other benign diseases associated with immune rejections such as GVHD. Dosimetry studies will be performed when translated into clinics. One of major caveats upon translation to clinics is a potential unknown biological effect of 89Zr-DFO-OX40 mAb to the patient immune system. To avoid any immunogenicity, engineering of OX40 antibody may be needed in the future. The rising clinical demand of immunotherapies highlights the importance of an accurate imaging strategy to noninvasively evaluate the distribution of stimulated immune-related organs longitudinally. The successful observation of stimulated lymphoid organs in this murine glioma model further supports the value of 89Zr-DFO-OX40 mAb PET and encourages future human study.

S.S. Gambhir reports grants from NIH and grants from Ben and Catherine Ivy Foundation during the conduct of the study. No disclosures were reported by the other authors.

T.W. Nobashi: Conceptualization, data curation, formal analysis, investigation, visualization, methodology, writing–original draft, writing–review and editing. A.T. Mayer: Conceptualization, data curation, supervision, investigation, visualization, methodology, writing–review and editing. Z. Xiao: Data curation, formal analysis, methodology. C.T. Chan: Supervision, methodology, writing–review and editing. A.M. Chaney: Resources, data curation, software, visualization, methodology. M.L. James: Resources, supervision, methodology, writing–review and editing. S.S. Gambhir: Resources, supervision, funding acquisition, project administration.

S.S. Gambhir reports grants from National Cancer Institute (R01 CA201719-05). The research was supported in part by the Ben & Catherine Ivy Foundation (to S.S. Gambhir). T.W. Nobashi was supported by Wagner-Torizuka Fellowship 2017-2019 sponsored by Nihon Medi-Physics Co., Ltd. We presented a portion of this study and received 2nd place of Young Investigator Award in the Center for Molecular Imaging Innovation & Translation at SNMMI 2020. The authors would like to thank the Stanford Center for Innovation in In-Vivo Imaging (SCi3) and the Stanford shared FACS facility for their support. In addition, we are grateful to Drs. Israt S. Alam and Gayatri Gowrishankar for supporting in vivo imaging studies. We also would like to dedicate this work to the memories of Dr. Sanjiv Sam Gambhir (1962-2020) and the valuable mentorship he provided.

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1.
Ostrom
QT
,
Gittleman
H
,
Truitt
G
,
Boscia
A
,
Kruchko
C
,
Barnholtz-Sloan
JS
. 
CBTRUS statistical report: primary brain and other central nervous system tumors diagnosed in the United States in 2011–2015
.
Neuro Oncol
2018
;
20
:
iv1
86
.
2.
Wilcox
JA
,
Ramakrishna
R
,
Magge
R
. 
Immunotherapy in glioblastoma
.
World Neurosurg
2018
;
116
:
518
28
.
3.
Korja
M
,
Raj
R
,
Seppä
K
,
Luostarinen
T
,
Malila
N
,
Seppälä
M
, et al
Glioblastoma survival is improving despite increasing incidence rates: a nationwide study between 2000 and 2013 in Finland
.
Neuro Oncol
2019
;
21
:
370
9
.
4.
Waldman
AD
,
Fritz
JM
,
Lenardo
MJ
. 
A guide to cancer immunotherapy: from T cell basic science to clinical practice
.
Nat Rev Immunol
2020
;
20
:
651
68
.
5.
Patel
AP
,
Tirosh
I
,
Trombetta
JJ
,
Shalek
AK
,
Gillespie
SM
,
Wakimoto
H
, et al
Single-cell RNA-seq highlights intratumoral heterogeneity in primary glioblastoma
.
Science
2014
;
344
:
1396
401
.
6.
Dix
AR
,
Brooks
WH
,
Roszman
TL
,
Morford
LA
. 
Immune defects observed in patients with primary malignant brain tumors
.
J Neuroimmunol
1999
;
100
:
216
32
.
7.
Woroniecka
K
,
Chongsathidkiet
P
,
Rhodin
K
,
Kemeny
H
,
Dechant
C
,
Farber
SH
, et al
T-cell exhaustion signatures vary with tumor type and are severe in glioblastoma
.
Clin Cancer Res
2018
;
24
:
4175
86
.
8.
Agarwalla
P
,
Barnard
Z
,
Fecci
P
,
Dranoff
G
,
Curry
WT
. 
Sequential immunotherapy by vaccination with GM-CSF-expressing glioma cells and CTLA-4 blockade effectively treats established murine intracranial tumors
.
J Immunother
2012
;
35
:
385
9
.
9.
Fecci
PE
,
Ochiai
H
,
Mitchell
DA
,
Grossi
PM
,
Sweeney
AE
,
Archer
GE
, et al
Systemic CTLA-4 blockade ameliorates glioma-induced changes to the CD4+ T cell compartment without affecting regulatory T-cell function
.
Clin Cancer Res
2007
;
13
:
2158
67
.
10.
Wainwright
DA
,
Chang
AL
,
Dey
M
,
Balyasnikova
IV
,
Kim
CK
,
Tobias
A
, et al
Durable therapeutic efficacy utilizing combinatorial blockade against IDO, CTLA-4, and PD-L1 in mice with brain tumors
.
Clin Cancer Res
2014
;
20
:
5290
301
.
11.
Okada
H
,
Kalinski
P
,
Ueda
R
,
Hoji
A
,
Kohanbash
G
,
Donegan
TE
, et al
Induction of CD8+ T-cell responses against novel glioma-associated antigen peptides and clinical activity by vaccinations with {alpha}-type 1 polarized dendritic cells and polyinosinic-polycytidylic acid stabilized by lysine and carboxymethylcellulose in patients with recurrent malignant glioma
.
J Clin Oncol
2011
;
29
:
330
6
.
12.
Kirkin
AF
,
Dzhandzhugazyan
KN
,
Guldberg
P
,
Fang
JJ
,
Andersen
RS
,
Dahl
C
, et al
Adoptive cancer immunotherapy using DNA-demethylated T helper cells as antigen-presenting cells
.
Nat Commun
2018
;
9
:
785
.
13.
Houot
R
,
Schultz
LM
,
Marabelle
A
,
Kohrt
H
. 
T-cell-based immunotherapy: adoptive cell transfer and checkpoint inhibition
.
Cancer Immunol Res
2015
;
3
:
1115
22
.
14.
Pardoll
DM
. 
The blockade of immune checkpoints in cancer immunotherapy
.
Nat Rev Cancer
2012
;
12
:
252
64
.
15.
Postow
MA
,
Callahan
MK
,
Wolchok
JD
. 
Immune checkpoint blockade in cancer therapy
.
J Clin Oncol
2015
;
33
:
1974
82
.
16.
Mallett
S
,
Fossum
S
,
Barclay
AN
. 
Characterization of the MRC OX40 antigen of activated CD4 positive T lymphocytes–a molecule related to nerve growth factor receptor
.
EMBO J
1990
;
9
:
1063
8
.
17.
Calderhead
DM
,
Buhlmann
JE
,
van den Eertwegh
AJ
,
Claassen
E
,
Noelle
RJ
,
Fell
HP
. 
Cloning of mouse Ox40: a T cell activation marker that may mediate T-B cell interactions
.
J Immunol
1993
;
151
:
5261
71
.
18.
Croft
M
,
So
T
,
Duan
W
,
Soroosh
P
. 
The significance of OX40 and OX40L to T-cell biology and immune disease
.
Immunol Rev
2009
;
229
:
173
91
.
19.
Weinberg
AD
,
Rivera
M-M
,
Prell
R
,
Morris
A
,
Ramstad
T
,
Vetto
JT
, et al
Engagement of the OX-40 receptor in vivo enhances antitumor immunity
.
J Immunol
2000
;
164
:
2160
9
.
20.
Kjaergaard
J
,
Tanaka
J
,
Kim
JA
,
Rothchild
K
,
Weinberg
A
,
Shu
S
. 
Therapeutic efficacy of OX-40 receptor antibody depends on tumor immunogenicity and anatomic site of tumor growth
.
Cancer Res
2000
;
60
:
5514
21
.
21.
Kjaergaard
J
,
Wang
L-X
,
Kuriyama
H
,
Shu
S
,
Plautz
GE
. 
Active immunotherapy for advanced intracranial murine tumors by using dendritic cell-tumor cell fusion vaccines
.
J Neurosurg
2005
;
103
:
156
64
.
22.
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
:
eaan4488
.
23.
Carpentier
A
,
Metellus
P
,
Ursu
R
,
Zohar
S
,
Lafitte
F
,
Barrie
M
, et al
Intracerebral administration of CpG oligonucleotide for patients with recurrent glioblastoma: a phase II study
.
Neuro Oncol
2010
;
12
:
401
8
.
24.
Ardon
H
,
Van Gool
SW
,
Verschuere
T
,
Maes
W
,
Fieuws
S
,
Sciot
R
, et al
Integration of autologous dendritic cell-based immunotherapy in the standard of care treatment for patients with newly diagnosed glioblastoma: results of the HGG-2006 phase I/II trial
.
Cancer Immunol Immunother
2012
;
61
:
2033
44
.
25.
Parney
IF
,
Gustafson
MP
,
Peterson
T
,
Steinmetz
SM
,
Dietz
AB
. 
ATIM-13. Allogeneic tumor lysate/autologous dendritic cell vaccines in newly diagnosed glioblastoma: results of clinical trial MC1272
.
Neuro Oncol
2017
;
19
:
vi28
9
.
26.
Murphy
KA
,
Lechner
MG
,
Popescu
FE
,
Bedi
J
,
Decker
SA
,
Hu
P
, et al
An in vivo immunotherapy screen of costimulatory molecules identifies Fc-OX40L as a potent reagent for the treatment of established murine gliomas
.
Clin Cancer Res
2012
;
18
:
4657
68
.
27.
Vander Heiden
MG
,
Cantley
LC
,
Thompson
CB
. 
Understanding the Warburg effect: the metabolic requirements of cell proliferation
.
Science
2009
;
324
:
1029
33
.
28.
Li
X
,
Rosenkrans
ZT
,
Wang
J
,
Cai
W
. 
PET imaging of macrophages in cardiovascular diseases
.
Am J Transl Res
2020
;
12
:
1491
514
.
29.
Rashidian
M
,
Ingram
JR
,
Dougan
M
,
Dongre
A
,
Whang
KA
,
LeGall
C
, et al
Predicting the response to CTLA-4 blockade by longitudinal noninvasive monitoring of CD8 T cells
.
J Exp Med
2017
;
214
:
2243
55
.
30.
Antonios
JP
,
Soto
H
,
Everson
RG
,
Moughon
DL
,
Wang
AC
,
Orpilla
J
, et al
Detection of immune responses after immunotherapy in glioblastoma using PET and MRI
.
Proc Natl Acad Sci U S A
2017
;
114
:
10220
5
.
31.
Seo
JW
,
Tavaré
R
,
Mahakian
LM
,
Silvestrini
MT
,
Tam
S
,
Ingham
ES
, et al
CD8 + T-cell density imaging with 64 Cu-labeled Cys-diabody informs immunotherapy protocols
.
Clin Cancer Res
2018
;
24
:
4976
87
.
32.
Weist
MR
,
Starr
R
,
Aguilar
B
,
Chea
J
,
Miles
JK
,
Poku
E
, et al
PET of adoptively transferred chimeric antigen receptor T cells with 89Zr-Oxine
.
J Nucl Med
2018
;
59
:
1531
7
.
33.
Perrin
J
,
Capitao
M
,
Mougin-Degraef
M
,
Guérard
F
,
Faivre-Chauvet
A
,
Rbah-Vidal
L
, et al
Cell tracking in cancer immunotherapy
.
Front Med
2020
;
7
:
34
.
34.
Griessinger
CM
,
Kehlbach
R
,
Bukala
D
,
Wiehr
S
,
Bantleon
R
,
Cay
F
, et al
In vivo tracking of Th1 cells by PET reveals quantitative and temporal distribution and specific homing in lymphatic tissue
.
J Nucl Med
2014
;
55
:
301
7
.
35.
Man
F
,
Lim
L
,
Volpe
A
,
Gabizon
A
,
Shmeeda
H
,
Draper
B
, et al
In vivo PET tracking of 89 Zr-labeled Vγ9Vδ2 T cells to mouse xenograft breast tumors activated with liposomal alendronate
.
Mol Ther
2019
;
27
:
219
29
.
36.
Nisar
S
,
Bhat
AA
,
Hashem
S
,
Yadav
SK
,
Rizwan
A
,
Singh
M
, et al
Non-invasive biomarkers for monitoring the immunotherapeutic response to cancer
.
J Transl Med
2020
;
18
:
471
.
37.
van de Donk
PP
,
Kist de Ruijter
L
,
Lub-de Hooge
MN
,
Brouwers
AH
,
van der Wekken
AJ
,
Oosting
SF
, et al
Molecular imaging biomarkers for immune checkpoint inhibitor therapy
.
Theranostics
2020
;
10
:
1708
18
.
38.
Willoughby
J
,
Griffiths
J
,
Tews
I
,
Cragg
MS
. 
OX40: structure and function - What questions remain?
Mol Immunol
2017
;
83
:
13
22
.
39.
Alam
IS
,
Mayer
AT
,
Sagiv-Barfi
I
,
Wang
K
,
Vermesh
O
,
Czerwinski
DK
, et al
Imaging activated T cells predicts response to cancer vaccines
.
J Clin Invest
2018
;
128
:
2569
80
.
40.
Barfi
IS
,
Czerwinski
DK
,
Shree
T
,
Levy
R
. 
In situ vaccination with Cpg and anti-OX40 antibody: preclinical optimization for clinical translation
.
Blood
2018
;
132
:
2943
.
41.
Holliger
P
,
Hudson
PJ
. 
Engineered antibody fragments and the rise of single domains
.
Nat Biotechnol
2005
;
23
:
1126
36
.
42.
Teuho
J
,
Riehakainen
L
,
Honkaniemi
A
,
Moisio
O
,
Han
C
,
Tirri
M
, et al
Evaluation of image quality with four positron emitters and three preclinical PET/CT systems
.
EJNMMI Res
2020
;
10
:
155
.
43.
Larson
SM
,
Erdi
Y
,
Akhurst
T
,
Mazumdar
M
,
Macapinlac
HA
,
Finn
RD
, et al
Tumor treatment response based on visual and quantitative changes in global tumor glycolysis using PET-FDG imaging. the visual response score and the change in total lesion glycolysis
.
Clin Positron Imaging
1999
;
2
:
159
71
.
44.
Gallicchio
R
,
Mansueto
G
,
Simeon
V
,
Nardelli
A
,
Guariglia
R
,
Capacchione
D
, et al
F-18 FDG PET/CT quantization parameters as predictors of outcome in patients with diffuse large B-cell lymphoma
.
Eur J Haematol
2014
;
92
:
382
9
.
45.
Chaney
AM
,
Johnson
EJ
,
Cropper
HC
,
James
ML
. 
PET imaging of neuroinflammation using [11C]DPA-713 in a mouse model of ischemic stroke
.
J Vis Exp
2018
;
136
:
57243
.
46.
Bradbury
MW
,
Cserr
HF
,
Westrop
RJ
. 
Drainage of cerebral interstitial fluid into deep cervical lymph of the rabbit
.
Am J Physiol
1981
;
240
:
F329
36
.
47.
Cserr
HF
,
Knopf
PM
. 
Cervical lymphatics, the blood-brain barrier and the immunoreactivity of the brain: a new view
.
Immunol Today
1992
;
13
:
507
12
.
48.
Buchan
SL
,
Rogel
A
,
Al-Shamkhani
A
. 
The immunobiology of CD27 and OX40 and their potential as targets for cancer immunotherapy
.
Blood
2018
;
131
:
39
48
.
49.
Cebada
J
,
Perez-Santos
M
,
Bandala
C
,
Lara-Padilla
E
,
Herrera-Camacho
I
,
Rosas-Murrieta
NH
, et al
OX40 agonists for cancer treatment: a patent review
.
Expert Opin Ther Pat
2021
;
31
:
81
90
.
50.
Alam
IS
,
Simonetta
F
,
Scheller
L
,
Mayer
AT
,
Murty
S
,
Vermesh
O
, et al
Visualization of activated T cells by OX40-ImmunoPET as a strategy for diagnosis of acute graft-versus-host disease
.
Cancer Res
2020
;
80
:
4780
90
.
51.
Xiao
Z
,
Mayer
AT
,
Nobashi
TW
,
Gambhir
SS
. 
ICOS is an indicator of T-cell-mediated response to cancer immunotherapy
.
Cancer Res
2020
;
80
:
3023
32
.
52.
Simonetta
F
,
Alam
IS
,
Lohmeyer
JK
,
Sahaf
B
,
Good
Z
,
Chen
W
, et al
Molecular imaging of chimeric antigen receptor T cells by ICOS-ImmunoPET
.
Clin Cancer Res
2021
;
27
:
1058
68
.
53.
Calzascia
T
,
Masson
F
,
Di Berardino-Besson
W
,
Contassot
E
,
Wilmotte
R
,
Aurrand-Lions
M
, et al
Homing phenotypes of tumor-specific CD8 T cells are predetermined at the tumor site by crosspresenting APCs
.
Immunity
2005
;
22
:
175
84
.
54.
Dunn
GP
,
Fecci
PE
,
Curry
WT
. 
Cancer immunoediting in malignant glioma
.
Neurosurgery
2012
;
71
:
201
22
.
55.
Joseph
RW
,
Elassaiss-Schaap
J
,
Kefford
R
,
Hwu
W-J
,
Wolchok
JD
,
Joshua
AM
, et al
Baseline tumor size is an independent prognostic factor for overall survival in patients with melanoma treated with pembrolizumab
.
Clin Cancer Res
2018
;
24
:
4960
7
.
56.
Chardin
D
,
Paquet
M
,
Schiappa
R
,
Darcourt
J
,
Bailleux
C
,
Poudenx
M
, et al
Baseline metabolic tumor volume as a strong predictive and prognostic biomarker in patients with non-small cell lung cancer treated with PD1 inhibitors: a prospective study
.
J Immunother Cancer
2020
;
8
:
e000645
.
57.
Cloughesy
TF
,
Mochizuki
AY
,
Orpilla
JR
,
Hugo
W
,
Lee
AH
,
Davidson
TB
, et al
Neoadjuvant anti-PD-1 immunotherapy promotes a survival benefit with intratumoral and systemic immune responses in recurrent glioblastoma
.
Nat Med
2019
;
25
:
477
86
.
58.
Wu
TD
,
Madireddi
S
,
de Almeida
PE
,
Banchereau
R
,
Chen
Y-JJ
,
Chitre
AS
, et al
Peripheral T cell expansion predicts tumour infiltration and clinical response
.
Nature
2020
;
579
:
274
8
.
59.
van Pul
KM
,
Fransen
MF
,
van de Ven
R
,
de Gruijl
TD
. 
Immunotherapy goes local: the central role of lymph nodes in driving tumor infiltration and efficacy
.
Front Immunol
2021
;
12
:
643291
.

Supplementary data