Skip to main content

Peptide-based PET imaging agent of tumor TIGIT expression

Abstract

Background

Accumulating studies have demonstrated that elevated TIGIT expression in tumor microenvironment correlates with better therapeutic response to TIGIT-based immunotherapy in pre-clinical studies. Therefore, a non-invasive method to detect tumor TIGIT expression is crucial to predict the therapeutic effect.

Methods

In this study, a peptide-based PET imaging agent, 68Ga-DOTA-DTBP-3, was developed to non-invasively detect TIGIT expression by micro-PET in tumor-bearing BALB/c mice. DTBP-3, a D-peptide comprising of 12 amino acids, was radiolabeled with 68Ga through a DOTA chelator. In vitro studies were performed to evaluate the affinity of 68Ga-DOTA-DTBP-3 to TIGIT and its stability in fetal bovine serum. In vivo studies were assessed by micro-PET, biodistribution, and immunohistochemistry on tumor-bearing BALB/c mice.

Results

The in vitro studies showed the equilibrium dissociation constant of 68Ga-DOTA-DTBP-3 for TIGIT was 84.21 nM and its radiochemistry purity was 89.24 ± 1.82% in FBS at 4 h in room temperature. The results of micro-PET, biodistribution and immunohistochemistry studies indicated that 68Ga-DOTA-DTBP-3 could be specifically targeted in 4T1 tumor-bearing mice, with a highest uptake at 0.5 h.

Conclusion

68Ga-DOTA-DTBP-3 holds potential for non-invasively detect tumor TIGIT expression and for timely assessment of the therapeutic effect of immune checkpoint blockade.

Introduction

Cancer cells utilize multiple pathways to evade immune-mediated recognition, one of powerful mechanism is the activation of immune checkpoint. Following continuous antigen stimulation, T cells become exhausted and upregulate the immune checkpoint molecules including CTLA-4 and PD-1, which severely limit the survival and function of T cells [1]. At the same time, ligands of these immune checkpoints receptors are overexpressed on antigen-presenting cells and cancer cells in the tumor microenvironment (TME). Over the last decade, antibody-based immunotherapy targeting immune checkpoints proteins like CTLA-4 and PD-1 have been remarkably successful, with obvious therapeutic effects in more than 15 types of advanced human malignancies [2,3,4]. Despite the success of immune checkpoint blockade therapies, only a small proportion of patients (approximately 20%) benefit from the current available immunotherapies [5]. Therefore, it is essential to discover novel immune checkpoints with high anti-tumor efficacy for various malignancies.

In addition to CTLA-4 and PD-1, TIGIT is an immune checkpoint expressed on CD4+ Th cells, CD8+ cytotoxic T lymphocytes, FOXP3+ regulatory T cells, and NK cells that is a potential target for future cancer immunotherapy [6,7,8]. TIGIT has two ligands, CD155 (PVR) and CD112 (PVRL2, nectin-2), which are expressed by antigen-presenting cells and tumor cells in TME to decrease the activity of T cells and NK cells. Studies have indicated that elevated TIGIT expression in the TME may be a marker of poor prognosis and that TIGIT+CD8+ T cells could be an independent prognostic indicator for TIGIT-based immunotherapy and a potential biomarker for responsiveness [9,10,11,12]. Furthermore, it has been demonstrated that TIGIT blockade, especially combined with other immune checkpoint inhibitors, may protect against various hematological and solid tumors, and several antibody-based immunotherapies that block the inhibitory activity of TIGIT have been developed [13, 14]. Several clinical trials are currently being developed to evaluate the effect of anti-TIGIT therapies. Therefore, it is necessary to analyze TIGIT expression in tumors before treatment to screen for the population that will benefit from immunotherapy and avoid the unnecessary financial burden for patients who will not respond.

Several approaches including immunohistochemistry (IHC), Western blotting, and PCR have been used to analyze TIGIT expression by various types, including follicular lymphoma, Hodgkin’s lymphoma, small cell lung cancer, hepatocellular carcinoma, squamous cell cancers, renal cell carcinoma, and melanoma [15,16,17,18,19,20,21]. IHC is the most commonly used method to detect tumoral TIGIT expression; however, it is limited by the highly heterogeneous and dynamic expression of immune checkpoint molecules [22]. Therefore, a non-invasive method is necessary to detect and quantify tumoral TIGIT expression in the TME, which will aid in screening the population most likely to benefit from anti-TIGIT immunotherapies [23]. This approach has been performed using single-photon emission computed tomography and positron emission tomography (PET) radiotracers for CTLA-4, PD-1, PD-L1, with some showing promising results in pre-clinical models [24,25,26]. PET is widely used to evaluate the characteristic of new drugs in pre-clinical studies due to its non-invasive, high-resolution and timely assessments of therapeutic effect.

Antibody-based TIGIT agents for PET imaging with zirconium-89 (89Zr) and copper-64 (64Cu) have been previously reported [27]; however, due to the long half-life of monoclonal antibodies and the relatively expensive production costs of 89Zr and 64Cu, the long retention of radioactive compounds may cause potential radiation damage to the body. Therefore, it is essential to develop low molecular weight-based radioactive compounds. Wang Xb [28] et al. utilized a 68Ga-labeled D-peptide antagonist, 68Ga-GP12, was developed and validated for PET imaging of TIGIT expression, and the potential of 68Ga-GP12 for PET/CT imaging of TIGIT expression were also evaluated in a pilot study with advanced NSCLC patients. However, only two clinical volunteers were recruited in the clinical trial, and whether this probe can be applied in clinical need to further validation. On the other hand, the tumor uptake of GP12 (SUVmax) and the ratio of target/non-target was lower compared to 18F-FDG, indicating that the radiotracer needs to be further optimized for clinical transformation. 68Ga is one of the most commonly used positron nuclides in clinical practice, and its availability from a generator and its proper half-life of 67.6 min, which matches well with the pharmacokinetic of a peptide, make it a promising candidate for in vivo imaging [25]. An increasing number of 68Ga-based radiotracers including 68Ga-DOTATATE and 68Ga-PSMA have gained widespread use in clinical practice.

In this study, a novel D-peptide TBP-3, that was first reported by Zhou et al. [29], was conjugated with 68Ga to non-invasively detect TIGIT expression in tumor-bearing BALB/c mice. Micro-PET and biodistribution studies demonstrated that 68Ga-DOTA-DTBP-3 is a promising radiotracer for non-invasive detecting of TIGIT in the TME.

Materials and methods

Cell lines and culture conditions

The murine mammary cancer cell line 4T1 and human melanoma cell line A375 were purchased from Shanghai Cell Bank of Chinese Academy of Sciences (Shanghai, China) and cultured in RPMI 1640 medium supplemented with 10% fetal bovine serum (FBS) (Gibco), penicillin (100 U/ml), and streptomycin (0.1 mg/ml) (Gibco, Grand Island, NY, USA) in a T25 flasks, which was placed in a humidified incubator at 37℃ in an atmosphere containing 5% CO2.

Reagents and instruments

DOTA-(COOt-Bu)3 was purchased from Macrocylics (Plano, TX, USA). A [68Ge/68Ga] generator (ITG GmbH, Berlin, Germany) was used to elute [68Ga] GaCl3. [68Ga] GaCl3 was eluted from the generator using 5 ml of 0.1 M HCI, and then passed through a cation exchange cartridge to trap the 68Ga ions alone. Finally, a concentrated NaCI/HCI solution was used to obtain the trapped pure 68Ga. High-performance liquid chromatography (HPLC) and mass spectrometry were performed on LC-20AT (Shimadzu Corporation, Tokyo, Japan) and LCMS-2020 (Shimadzu Corporation, Tokyo, Japan), respectively.

Synthesis of 68Ga-DOTA-DTBP-3

DTBP-3 and DOTA-DTBP-3 were synthesized by Shanghai Apeptide Co. (Shanghai, China). Briefly, 1 mg of DOTA-DTBP-3 was dissolved in 250 μL of 0.25 M sodium acetate buffer (pH 8.6). Subsequently, the pH of the peak fraction of the 1.5 mL 68GaCl3 (140–360 MBq) in 0.05 M HCI was adjusted to approximately 4.0 by adding 450 μL 0.25 M sodium acetate (pH 8.6). Then, DOTA-DTBP-3 (3.78–9.72 μL) was incubated with 140–360 MBq of 68 Ga for 10 min at 100 °C with a final pH of 4.0. The compounds were purified using a preconditional C-18 Sep-Pak cartridge column, rinsed with 10 mL of ultrapure water, and eluted with 1.0 mL of ethanol. Radio-HPLC was performed to assess the radiochemical purity of the radiotracer. A semipreparative C-18 Luna column (5, 10 × 250 mm) was used with methanol (0.1% TFA) and water (0.1% TFA) as the mobile phase, with a flow rate of 4.5 mL/min, the linear gradient from 65/35 (methanol/water) to 85/15 (methanol/water) exceeding 30 min for peak separation.

In vitro binding assay

The equilibrium dissociation constant of 68Ga-DOTA-DTBP-3 for TIGIT was determined by a cell-saturated assay. Briefly, 1 × 106 4T1 cells/well were grown to confluence in 24-well plates. After incubating the cells for 30 min 150 μL 1% BSA/PBS, serial dilutions of 68Ga-DOTA-DTBP-3 (from 1 × 10–4 to 5 × 102 nM) preparations were added to each well in triplicate and then co-incubated at 37℃ for 0.5 h. The wells were washed 3 times with cold PBS, and then 0.1 M NaOH solution was used to lyse the cells. The supernatant was collected, precipitated separately, and the radioactivity was measured using an automated γ counter. Non-specific cell binding was determined by adding 40-fold DTBP-3 (about 120.0 μg), incubating for 0.5 h, and then adding the 68Ga-DOTA-DTBP-3. Equilibrium dissociation constant KD was analyzed using a computer program.

For cell uptake experiments, 4T1 and A375 cells were incubated with 68Ga-DOTA-DTBP-3 at different time points (15, 30, 60 and 120 min), respectively. Then, the cells were washed 3 times with cold PBS, and 0.1 M NaOH solution was used to lyse the cells and collected for γ-count.

Animal studies

All animal procedures were strictly implemented in accordance with the guidelines approved by the Institutional Animal Care and Use Committee of Wuhan University.

Female BALB/c mice of 4–5 weeks were purchased from Beijing Biotechnology Co., Ltd. (Beijing, China). The 4T1 murine cancer model was established by the injection of 3 × 105 cells/150 μl into the front right limbs of female BALB/c mice. Experiments started when the tumors reached a volume of approximately 250–350 mm3.

Micro-PET studies

PET images were acquired on a micro-PET scanner (InliView-3000B, Minfound, Hangzhou, China). Mice bearing subcutaneous tumors were anesthetized with isoflurane (1.0–1.5%) and then were injected with about 5.55 MBq of 68Ga-DOTA-DTBP-3 via the tail vein. In the blocking group, tumor-bearing BALB/c were pretreated with the 40-fold excess of DTBP-3 for 0.5 h, and then injected equal dose of radiotracer with the above groups. Static imaging (15 min) was collected at different time points (0.5, 1 and 2 h post-injection).

Biodistribution and pharmacokinetics studies

Female BALB/c tumor-bearing mice with subcutaneous 4T1 xenografts were randomly divided into 2 groups (n = 9 for the experimental group, and n = 3 for the blocking group), and then were injected with 3.70 MBq of 68Ga-DOTA-DTBP-3 via the tail vein. At different time points (0.5, 1 and 2 h post-injection for the experimental group, 0.5 h post-injection for the blocking group), the mice were euthanized, and the tumor, main organs (muscle, bone, large intestine, small intestine, stomach, kidney, spleen, liver, lung, heart, brain) and blood were weighed and counted by a γ counter. The results are expressed as percentage injected dose per gram of tissue (% ID/g), and the values were calculated based on signal decay correction and normalization to external 68Ga standards.

HE&IHC for TIGIT expression

For IHC, harvested tumors were fixed in 4% paraformaldehyde and then embedded. After deparaffinization with xylene and an alcohol gradient, antigen retrieval was performed using 10 mM citrate buffer (pH 6.0). An anti-mouse TIGIT monoclonal antibody (catalog NO. BE0274, Bio X cell) was used as primary antibody. Tumor sections were incubated with the primary anti-mouse TIGIT antibody with a dilution of 1:500 at 4℃ overnight, washed with PBS for twice, and then incubated with the secondary antibody for 1 h at room temperature. The slides were counterstained with hematoxylin, dehydrated with an alcohol gradient, washed with xylene, and fixed with a cover slip.

Statistical analysis

All data were analyzed with the software Graphpad Prism 8.0 (Graphpad Software Inc., San Diego, CA, USA), with P < 0.05 being considered statistically significant.

Results

Synthesis and characterization of 68Ga-DOTA-DTBP-3

The low molecular weight D-peptide TBP-3 that targets TIGIT was reported to penetrate and accumulate in the tumor tissues. To utilize this characteristic for non-invasive PET imaging of TIGIT expression, the DOTA-DTBP-3 was synthesized. DOTA-DTBP-3 was obtained with its chemical purity more than 95% (Fig. 1a). The retention time of 68Ga-DOTA-DTBP-3 was about 13.4 min (Fig. 1b). The radiochemical purity of 68Ga-DOTA-DTBP-3 in FBS was 97.23 ± 0.59% after purification, and the molar activity was calculated to be 88.34 ± 4.56 MBq/nmol. The stability of 68Ga-DOTA-DTBP-3 in FBS was demonstrated as a radiochemical purity of 89.24 ± 1.82% at 4 h in room temperature, which revealed its relative stable.

Fig. 1
figure 1

The characterization of DOTA-DTBP-3 was identified by mass spectrometer (a). The retention time of 68 Ga-DOTA-DTBP-3 was about 13.4 min identified by radio-HPLC (b)

In vitro studies

The TIGIT expression of 4T1 and A375 cells was demonstrated by western blot (Fig. 2a), and 4T1 cell has a high TIGIT expression while A375 cell has a low TIGIT expression. A cell-saturated binding assay was performed to determine the affinity of radiotracer to murine TIGIT. The radiotracer showed a relative high affinity to murine TIGIT with the equilibrium dissociation constant KD was 84.21 nM (Fig. 2b). 4T1 and A375 cells have a highest uptake of radiotracer with 8.41 ± 1.22% and 2.56 ± 0.19% at 0.5 h, respectively (Fig. 2c). The 4T1 cellular uptake of the radiotracer could be blocked by excess unlabeled DTBP-3, indicating their specific binding.

Fig. 2
figure 2

The TIGIT expression of 4T1 and A375 cells was demonstrated by western blot (a). Binding curve of 68 Ga-DOTA-DTBP-3 to TIGIT (b), showing a relative high affinity to murine TIGIT with the equilibrium dissociation constant KD was 84.21 nM. Cell uptake curve of 4T1 and A375 cells with different time points (15, 30, 60 and 120 min), 4T1 and A375 cells have a highest uptake of radiotracer with 8.41 ± 1.22% and 2.56 ± 0.19% at 0.5 h, respectively (c)

Micro-PET imaging

The TIGIT-targeting ability of 68Ga-DOTA-DTBP-3 was performed on micro-PET in 4T1 tumor-bearing mice (Fig. 3a). As shown in Fig. 3a, 4T1 tumor-bearing BALB/c mice showed rapid and high uptake (3.49 ± 0.14) of 68Ga-DOTA-DTBP-3 in the tumors at 0.5 h following intravenously injection, with the highest tumor/muscle ratio of 5.20 ± 0.16. There was no obvious uptake of 68Ga-DOTA-DTBP-3 by tumors in 4T1 tumor-bearing mice pretreated with the unlabeled DTBP-3. This result was further confirmed by HE and IHC (Fig. 4a and b).

Fig. 3
figure 3

PET images of tumor-bearing mice. Static images at 0.5, 1, and 2 h post-injection of 4.44 MBq 68 Ga-DOTA-DTBP3 (n = 3), 68 Ga-DOTA-DTBP3 could be blocked by DTBP3 at 0.5, 1 and 2 h (While the blocking group at 1, 2 h were not shown) (a), and tumor/muscle rations were shown in b

Fig. 4
figure 4

HE and IHC of 4T1 tumor (a and b)

Ex vivo studies

The biodistribution data of 68Ga-DOTA-DTBP-3 in 4T1 tumor-bearing BALB/c mice are presented in Fig. 5. At 0.5 h post-injection, the kidney had relatively high radiotracer uptake, while the liver and remaining organs showed low radiotracer uptake. The biodistribution results indicated that 68Ga-DOTA-DTBP-3 could be quickly cleared from the blood, which resulted in a high tumor-to-muscle ratio and a high tumor-to-blood ratio at 0.5 h post-injection in 4T1 tumor-bearing mice.

Fig. 5
figure 5

Biodistribution analysis at 0.5, 1 and 2 h post-injection of 68 Ga-DOTA-DTBP3 in 4T1 tumor-bearing mice model (n = 3) (Fig. 4)

Discussion

TIGIT is expected as a promising therapeutic target for the next generation of immune checkpoints, and the efficacy of anti-TIGIT immunotherapy is correlated with the expression of TIGIT in the TME. However, detection by IHC relies on samples taken by biopsy, which is invasive and does not provide a comprehensive quantification of expression levels in the whole body. PET is a non-invasive imaging technology that can be performed in real time, is repeatable, and allows dynamic detection of all lesions [27]. Thus, it could greatly facilitate the screening of patients who may benefit from specific immunotherapies, evaluations of treatment efficacy, and treatment adjustments. In this study, a novel D-peptide TBP-3, was conjugated with 68 Ga to non-invasively detect TIGIT expression in tumor-bearing BALB/c mice.

PET has been widely used to image other immune checkpoints proteins including PD-1/PD-L1 [30,31,32], LAG-3 [33], and TIM-3 [34] with radionuclide-labeled antibodies. Apart from the shortcomings of immune-related side effects, poor tissue infiltration is feature of antibody themselves; additionally, their relatively long half-lives means than imaging can only be performed 4–7 d to allow for better tissue contrast [35]. In addition, low molecular weight peptide drugs are ideal candidates for PET imaging because they have the appropriate half-lives and strong tissue infiltration. High-affinity peptides targeting PD-L1, especially TPP-1 and WL12 have been widely used for PET imaging with various radionuclide label, includes 68Ga, 18F, 64Cu and 89Zr [25, 36,37,38,39,40,41,42]. PET imaging of TIGIT has been explored using 64Cu and 89Zr-labeled TIGIT antibody and remains to be further developed. Here, we conducted PET imaging with DTBP-3, which is a high-affinity D-peptide for TIGIT with proteolysis-resistance and strong tissue infiltration [29]. As to the radionuclide label, 68Ga was selected. 68Ga is widely used in clinical centers and has the benefit of low radiation exposure. More importantly, the half-life of 68Ga has comparative pharmacokinetic properties with peptides [25].

For PET imaging of TIGIT, a murine breast cancer cell line with high expression of TIGIT was used [43]. As expected, BALB/c tumor-bearing mice had the highest uptake of PET radiotracer at 0.5 h, and this could be blocked by excess unlabeled DTBP-3, which demonstrated that 68Ga-DOTA-DTBP-3 could specifically bind to TIGIT in vivo. Fortunately, Wang Xb [28] et al. also utilized a 68Ga-labeled D-peptide antagonist, 68Ga-GP12, was developed and validated for PET imaging of TIGIT expression, and the potential of 68Ga-GP12 for PET/CT imaging of TIGIT expression were also evaluated in a pilot study with advanced NSCLC patients. Although the same D-peptide was used in both studies, there were still some differences of the probe. They modified the peptide with polyethylene glycol and then coupled with NOTA while our study directly used DOTA to couple the D-peptide. The affinity of 68Ga-GP12 to TIGIT and the stability of 68Ga-GP12 in FBS were better than 68Ga-DOTA-DTBP-3 may attribute to the following reasons. Firstly, the structure of the NOTA ring matches 68Ga better than DOTA; secondly, PEG modified peptide increases the water solubility of the probe. In the experiment, we found that the half-life of 68Ga-DOTA-DTBP-3 was fast in vivo, and the peak uptake of tumor tissue was about 0.5 h which was quickly than them. On the other hand, liver uptake was obvious lower than our study which due to the PEG modified peptide. In addition, there are some limitations of this study. TIGIT is mainly expressed in tumor infiltrating lymphocytes, while a cell line with tumor cells overexpressed TIGIT was used in our study which may not reflect the true situation of the TME. On the other hand, we did not compare it with 18-F-FDG.

Single domain antibody is particularly suitable for molecular imaging due to its small molecular weight and high affinity with receptor, and 68Ga-labeled PD-L1 single domain antibody showed better pharmacokinetic in pre-clinical or clinical practice. In the future, NOTA conjugated TIGIT single domain antibody would be used to monitor tumor TIGIT expression, and human TIGIT would be explored for further study.

Conclusions

This work validates the TIGIT-specific peptide for quantification of the expression level of TIGIT in the TME. We demonstrate that 68Ga-DOTA-DTBP-3 could serve as a specific molecular probe for detecting TIGIT expression in vivo. 68Ga-DOTA-DTBP-3 was evaluated in BALB/C mice with 4T1 breast cancer and showed the potential to quantitatively detect TIGIT expression from 0.5 min to 1 h post-injection. These results indicate 68Ga-DOTA- DTBP-3 as a potential companion diagnosis or evidence for patient stratification of TIGIT blocking-based cancer immunotherapy.

Availability of data and materials

All data generated or analyzed during the current study are included in this published article.

References

  1. Chauvin JM, Zarour HM. TIGIT in cancer immunotherapy. J Immunother Cancer. 2020. https://doi.org/10.1136/jitc-2020-000957.

    Article  PubMed  PubMed Central  Google Scholar 

  2. Kruger S, Ilmer M, Kobold S, Cadilha BL, Endres S, Ormanns S, et al. Advances in cancer immunotherapy 2019—latest trends. J Exp Clin Cancer Res CR. 2019;38:268. https://doi.org/10.1186/s13046-019-1266-0.

    Article  PubMed  Google Scholar 

  3. Smyth MJ, Ngiow SF, Ribas A, Teng MW. Combination cancer immunotherapies tailored to the tumour microenvironment. Nat Rev Clin Oncol. 2016;13:143–58. https://doi.org/10.1038/nrclinonc.2015.209.

    Article  CAS  PubMed  Google Scholar 

  4. Martins F, Sofiya L, Sykiotis GP, Lamine F, Maillard M, Fraga M, et al. Adverse effects of immune-checkpoint inhibitors: epidemiology, management and surveillance. Nat Rev Clin Oncol. 2019;16:563–80. https://doi.org/10.1038/s41571-019-0218-0.

    Article  CAS  PubMed  Google Scholar 

  5. Darvin P, Toor SM, Sasidharan Nair V, Elkord E. Immune checkpoint inhibitors: recent progress and potential biomarkers. Exp Mol Med. 2018;50:1–11. https://doi.org/10.1038/s12276-018-0191-1.

    Article  CAS  PubMed  Google Scholar 

  6. Dougall WC, Kurtulus S, Smyth MJ, Anderson AC. TIGIT and CD96: new checkpoint receptor targets for cancer immunotherapy. Immunol Rev. 2017;276:112–20. https://doi.org/10.1111/imr.12518.

    Article  CAS  PubMed  Google Scholar 

  7. Solomon BL, Garrido-Laguna I. TIGIT: a novel immunotherapy target moving from bench to bedside. Cancer Immunol Immunother CII. 2018;67:1659–67. https://doi.org/10.1007/s00262-018-2246-5.

    Article  CAS  PubMed  Google Scholar 

  8. Johnston RJ, Comps-Agrar L, Hackney J, Yu X, Huseni M, Yang Y, et al. The immunoreceptor TIGIT regulates antitumor and antiviral CD8(+) T cell effector function. Cancer Cell. 2014;26:923–37. https://doi.org/10.1016/j.ccell.2014.10.018.

    Article  CAS  PubMed  Google Scholar 

  9. Liu Z, Zhou Q, Wang Z, Zhang H, Zeng H, Huang Q, et al. Intratumoral TIGIT(+) CD8(+) T-cell infiltration determines poor prognosis and immune evasion in patients with muscle-invasive bladder cancer. J Immunother Cancer. 2020. https://doi.org/10.1136/jitc-2020-000978.

    Article  PubMed  PubMed Central  Google Scholar 

  10. Chiu DK, Yuen VW, Cheu JW, Wei LL, Ting V, Fehlings M, et al. Hepatocellular carcinoma cells up-regulate PVRL1, stabilizing PVR and inhibiting the cytotoxic T-cell response via TIGIT to mediate tumor resistance to PD1 inhibitors in mice. Gastroenterology. 2020;159:609–23. https://doi.org/10.1053/j.gastro.2020.03.074.

    Article  CAS  PubMed  Google Scholar 

  11. Sun Y, Luo J, Chen Y, Cui J, Lei Y, Cui Y, et al. Combined evaluation of the expression status of CD155 and TIGIT plays an important role in the prognosis of LUAD (lung adenocarcinoma). Int Immunopharmacol. 2020;80:106198. https://doi.org/10.1016/j.intimp.2020.106198.

    Article  CAS  PubMed  Google Scholar 

  12. Xu D, Zhao E, Zhu C, Zhao W, Wang C, Zhang Z, et al. TIGIT and PD-1 may serve as potential prognostic biomarkers for gastric cancer. Immunobiology. 2020;225:151915. https://doi.org/10.1016/j.imbio.2020.151915.

    Article  CAS  PubMed  Google Scholar 

  13. Burugu S, Dancsok AR, Nielsen TO. Emerging targets in cancer immunotherapy. Semin Cancer Biol. 2018;52:39–52. https://doi.org/10.1016/j.semcancer.2017.10.001.

    Article  CAS  PubMed  Google Scholar 

  14. Liu XG, Hou M, Liu Y. TIGIT, a novel therapeutic target for tumor immunotherapy. Immunol Invest. 2017;46:172–82. https://doi.org/10.1080/08820139.2016.1237524.

    Article  CAS  PubMed  Google Scholar 

  15. Blessin NC, Simon R, Kluth M, Fischer K, Hube-Magg C, Li W, et al. Patterns of TIGIT expression in lymphatic tissue, inflammation, and cancer. Dis Markers. 2019;2019:5160565. https://doi.org/10.1155/2019/5160565.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Duan X, Liu J, Cui J, Ma B, Zhou Q, Yang X, et al. Expression of TIGIT/CD155 and correlations with clinical pathological features in human hepatocellular carcinoma. Mol Med Rep. 2019;20:3773–81. https://doi.org/10.3892/mmr.2019.10641.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Josefsson SE, Huse K, Kolstad A, Beiske K, Pende D, Steen CB, et al. T cells expressing checkpoint receptor TIGIT are enriched in follicular lymphoma tumors and characterized by reversible suppression of T-cell receptor signaling. Clin Cancer Res Off J Am Assoc Cancer Res. 2018;24:870–81. https://doi.org/10.1158/1078-0432.ccr-17-2337.

    Article  CAS  Google Scholar 

  18. Li W, Blessin NC, Simon R, Kluth M, Fischer K, Hube-Magg C, et al. Expression of the immune checkpoint receptor TIGIT in Hodgkin’s lymphoma. BMC Cancer. 2018;18:1209. https://doi.org/10.1186/s12885-018-5111-1.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Hong X, Wang X, Wang T, Zhang X. Correlation of T cell immunoglobulin and ITIM domain (TIGIT) and programmed death 1 (PD-1) with clinicopathological characteristics of renal cell carcinoma may indicate potential targets for treatment. Med Sci Monit Int Med J Exp Clin Res. 2018;24:6861–72. https://doi.org/10.12659/msm.910388.

    Article  CAS  Google Scholar 

  20. Xu Y, Cui G, Jiang Z, Li N, Zhang X. Survival analysis with regard to PD-L1 and CD155 expression in human small cell lung cancer and a comparison with associated receptors. Oncol Lett. 2019;17:2960–8. https://doi.org/10.3892/ol.2019.9910.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Inozume T, Yaguchi T, Furuta J, Harada K, Kawakami Y, Shimada S. Melanoma cells control antimelanoma CTL responses via Interaction between TIGIT and CD155 in the effector phase. J Invest Dermatol. 2016;136:255–63. https://doi.org/10.1038/jid.2015.404.

    Article  CAS  PubMed  Google Scholar 

  22. Natarajan A, Mayer AT, Xu L, Reeves RE, Gano J, Gambhir SS. Novel radiotracer for ImmunoPET imaging of PD-1 checkpoint expression on tumor infiltrating lymphocytes. Bioconjug Chem. 2015;26:2062–9. https://doi.org/10.1021/acs.bioconjchem.5b00318.

    Article  CAS  PubMed  Google Scholar 

  23. Ehlerding EB, England CG, McNeel DG, Cai W. Molecular imaging of immunotherapy targets in cancer. J Nucl Med Off Publ Soc Nucl Med. 2016;57:1487–92. https://doi.org/10.2967/jnumed.116.177493.

    Article  CAS  Google Scholar 

  24. Lv G, Sun X, Qiu L, Sun Y, Li K, Liu Q, et al. PET imaging of tumor PD-L1 expression with a highly specific nonblocking single-domain antibody. J Nucl Med Off Publ Soc Nucl Med. 2020;61:117–22. https://doi.org/10.2967/jnumed.119.226712.

    Article  CAS  Google Scholar 

  25. De Silva RA, Kumar D, Lisok A, Chatterjee S, Wharram B, Venkateswara Rao K, et al. Peptide-based (68)Ga-PET radiotracer for imaging PD-L1 expression in cancer. Mol Pharm. 2018;15:3946–52. https://doi.org/10.1021/acs.molpharmaceut.8b00399.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Robu S, Richter A, Gosmann D, Seidl C, Leung D, Hayes W, et al. Synthesis and preclinical evaluation of (68)Ga-labeled adnectin, (68)Ga-BMS-986192 as a PET agent for imaging PD-L1 expression. J Nucl Med Offi Publ Soc Nucl Med. 2021. https://doi.org/10.2967/jnumed.120.258384.

    Article  Google Scholar 

  27. Shaffer T, Natarajan A, Gambhir SS. PET imaging of TIGIT expression on tumor-infiltrating lymphocytes. Clin Cancer Res Offi J Am Assoc Cancer Res. 2021;27:1932–40. https://doi.org/10.1158/1078-0432.ccr-20-2725.

    Article  CAS  Google Scholar 

  28. Wang X, Zhou M, Chen B, Liu H, Fang J, Xiang S, et al. Preclinical and exploratory human studies of novel (68)Ga-labeled D-peptide antagonist for PET imaging of TIGIT expression in cancers. Eur J Nucl Med Mol Imaging. 2022;49:2584–94. https://doi.org/10.1007/s00259-021-05672-x.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Zhou X, Zuo C, Li W, Shi W, Zhou X, Wang H, et al. A novel d-peptide identified by mirror-image phage display blocks TIGIT/PVR for cancer immunotherapy. Angew Chem Int Ed Engl. 2020;59:15114–8. https://doi.org/10.1002/anie.202002783.

    Article  CAS  PubMed  Google Scholar 

  30. England CG, Ehlerding EB, Hernandez R, Rekoske BT, Graves SA, Sun H, et al. Preclinical pharmacokinetics and biodistribution studies of 89Zr-labeled pembrolizumab. J Nucl Med Off Publ Soc Nucl Med. 2017;58:162–8. https://doi.org/10.2967/jnumed.116.177857.

    Article  CAS  Google Scholar 

  31. Du Y, Liang X, Li Y, Sun T, Jin Z, Xue H, et al. Nuclear and fluorescent labeled PD-1-Liposome-DOX-(64)Cu/IRDye800CW allows improved breast tumor targeted imaging and therapy. Mol Pharm. 2017;14:3978–86. https://doi.org/10.1021/acs.molpharmaceut.7b00649.

    Article  CAS  PubMed  Google Scholar 

  32. Truillet C, Oh HLJ, Yeo SP, Lee CY, Huynh LT, Wei J, et al. Imaging PD-L1 expression with ImmunoPET. Bioconjug Chem. 2018;29:96–103. https://doi.org/10.1021/acs.bioconjchem.7b00631.

    Article  CAS  PubMed  Google Scholar 

  33. Lecocq Q, Zeven K, De Vlaeminck Y, Martens S, Massa S, Goyvaerts C, et al. Noninvasive imaging of the immune checkpoint LAG-3 using nanobodies, from development to pre-clinical use. Biomolecules. 2019. https://doi.org/10.3390/biom9100548.

    Article  PubMed  PubMed Central  Google Scholar 

  34. Wei W, Jiang D, Lee HJ, Engle JW, Akiba H, Liu J, et al. ImmunoPET imaging of TIM-3 in murine melanoma models. Adv Therapeutics. 2020. https://doi.org/10.1002/adtp.202000018.

    Article  Google Scholar 

  35. Wang GX, Guo LQ, Gainor JF, Fintelmann FJ. Immune checkpoint inhibitors in lung cancer: imaging considerations. AJR Am J Roentgenol. 2017;209:567–75. https://doi.org/10.2214/ajr.16.17770.

    Article  PubMed  Google Scholar 

  36. Wang W, Hu Z. Targeting peptide-based probes for molecular imaging and diagnosis. Adv Mater (Deerfield Beach, Fla). 2019;31:e1804827. https://doi.org/10.1002/adma.201804827.

    Article  CAS  Google Scholar 

  37. Zhang X, Zhao H, Shi X, Jia X, Yang Y. Identification and validation of an immune-related gene signature predictive of overall survival in colon cancer. Aging. 2020;12:26095–120. https://doi.org/10.18632/aging.202317.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Chatterjee S, Lesniak WG, Miller MS, Lisok A, Sikorska E, Wharram B, et al. Rapid PD-L1 detection in tumors with PET using a highly specific peptide. Biochem Biophys Res Commun. 2017;483:258–63. https://doi.org/10.1016/j.bbrc.2016.12.156.

    Article  CAS  PubMed  Google Scholar 

  39. Lesniak WG, Mease RC, Chatterjee S, Kumar D, Lisok A, Wharram B, et al. Development of [(18)F]FPy-WL12 as a PD-L1 specific PET imaging peptide. Mol Imaging. 2019;18:1536012119852189. https://doi.org/10.1177/1536012119852189.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Kumar D, Lisok A, Dahmane E, McCoy M, Shelake S, Chatterjee S, et al. Peptide-based PET quantifies target engagement of PD-L1 therapeutics. J Clin Investig. 2019;129:616–30. https://doi.org/10.1172/jci122216.

    Article  PubMed  PubMed Central  Google Scholar 

  41. Hu K, Kuan H, Hanyu M, Masayuki H, Xie L, Zhang Y, et al. Developing native peptide-based radiotracers for PD-L1 PET imaging and improving imaging contrast by pegylation. Chem Commun (Camb). 2019;55:4162–5. https://doi.org/10.1039/c9cc00445a.

    Article  CAS  PubMed  Google Scholar 

  42. Jiang J, Li D, Liu T, Xia L, Guo X, Meng X, et al. Noninvasive evaluation of PD-L1 expression using Copper 64 labeled peptide WL12 by micro-PET imaging in Chinese hamster ovary cell tumor model. Bioorganic Med Chem Lett. 2021;40:127901. https://doi.org/10.1016/j.bmcl.2021.127901.

    Article  CAS  Google Scholar 

  43. Zhou XM, Li WQ, Wu YH, Han L, Cao XG, Yang XM, et al. Intrinsic expression of immune checkpoint molecule TIGIT could help tumor growth in vivo by suppressing the function of NK and CD8(+) T cells. Front Immunol. 2018;9:2821. https://doi.org/10.3389/fimmu.2018.02821.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

This work was financially supported by the “Fundamental Research Funds for the Central Universities” (Grant No. 2042018kf0179) and the “Research Fund from medical Sci-Tech innovation platform of Zhongnan Hospital, Wuhan University” (Grant No. PTXM2022029). Currently, there are no similar reports to this study.

Author information

Authors and Affiliations

Authors

Contributions

DW and RG were responsible for labeling peptide, Micro-PET imaging and essay writing. ZZ, YG and ZG were responsible for cell culture and animal feeding. DW, RG and RA were responsible for data analysis. DW and XZ were responsible for synthesis of the peptide. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Dinghu Weng.

Ethics declarations

Ethics approval and consent to participate

All animal procedures were strictly implemented in accordance with the guidelines approved by the Institutional Animal Care and Use Committee of Wuhan University. The study was carried out in compliance with the ARRIVE guidelines.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Weng, D., Guo, R., Zhu, Z. et al. Peptide-based PET imaging agent of tumor TIGIT expression. EJNMMI Res 13, 38 (2023). https://doi.org/10.1186/s13550-023-00982-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s13550-023-00982-7

Keywords