Summary

Development of a 68Gallium-Labeled D-Peptide PET Tracer for Imaging Programmed Death-Ligand 1 Expression

Published: February 03, 2023
doi:

Summary

This study developed a noninvasive and real-time method to evaluate the distribution of programmed death-ligand 1 in the whole body, based on positron emission tomographic imaging of [68Ga] D-dodecapeptide antagonist. This technique has advantages over conventional immunohistochemistry and improves the efficiency of identifying appropriate patients who will benefit from immune checkpoint blockade therapy.

Abstract

The development of immune checkpoint blockade therapy based on programmed cell death-protein 1 (PD-1)/programmed death-ligand 1 (PD-L1) has revolutionized cancer therapies in recent years. However, only a fraction of patients responds to PD-1/PD-L1 inhibitors, owing to the heterogeneous expression of PD-L1 in tumor cells. This heterogeneity presents a challenge in the precise detection of tumor cells by the commonly used immunohistochemistry (IHC) approach. This situation calls for better methods to stratify patients who will benefit from immune checkpoint blockade therapy, to improve treatment efficacy. Positron emission tomography (PET) enables real-time visualization of the whole-body PD-L1 expression in a noninvasive way. Therefore, there is a need for the development of radiolabeled tracers to detect PD-L1 distribution in tumors through PET imaging.

Compared to their L-counterparts, dextrorotary (D)-peptides have properties such as proteolytic resistance and remarkably prolonged metabolic half-lives. This study designed a new method to detect PD-L1 expression based on PET imaging of 68Ga-labeled PD-L1-targeted D-peptide, a D-dodecapeptide antagonist (DPA), in tumor-bearing mice. The results showed that the [68Ga]DPA can specifically bind to PD-L1-overexpressing tumors in vivo, and showed favorable stability as well as excellent imaging ability, suggesting that [68Ga]DPA-PET is a promising approach for the assessment of PD-L1 status in tumors.

Introduction

The discovery of immune checkpoint proteins was a breakthrough in tumor therapy, and has led to major advances in the development of immune checkpoint blockade therapy1. Programmed cell death-protein 1 (PD-1) and programmed death-ligand 1 (PD-L1) are potential drug targets with several antibodies approved by the Food and Drug Administration (FDA). PD-1 is expressed by tumor-infiltrating immune cells, such as CD4+, CD8+ T cells, and regulatory T cells. PD-L1 is one of the PD-1 ligands, which is overexpressed in a variety of tumor cells2,3. The interaction between PD-1 and PD-L1 inactivates PD-1, thus suppressing the antitumor immune response4. These findings suggest that the inhibition of PD-L1 can improve the killing effect of immune cells and eliminate tumor cells5. Currently, chromogenic immunohistochemistry (IHC) is the most commonly used approach to identify patients who are most likely to respond to immune checkpoint therapy6,7. However, due to the heterogeneous expression of PD-L1 in tumor cells, IHC results from biopsies cannot provide accurate information about PD-L1 expression in patients8. Previous studies have reported that only 20%-40% of patients gain long-term benefits from immune checkpoint blockade therapy1,9,10. There is, therefore, an urgent need to develop a new method to circumvent the false-negative results caused by the heterogeneous expression of these immune checkpoint proteins.

Molecular imaging technology, such as positron emission tomography (PET), enables real-time visualization of the whole body in a noninvasive way, and thus can outperform the conventional IHC method11,12,13. Radiolabeled antibodies, peptides, and small molecules are promising tracers for monitoring PD-L1 expression in cancer patients14,15,16,17,18,19,20,21,22,23,24,25. The FDA has approved three PD-L1 therapeutic monoclonal antibodies: avelumab, atezolizumab, and durvalumab26. Immuno-PET tracers based on these antibodies have been well documented27,28,29,30,31,32. Early-phase clinical trials have revealed limited value for clinical application, because of the unfavorable pharmacokinetics30. Compared with antibodies, peptides exhibit faster blood and organ clearance from healthy organs, and can be easily chemically modified33. Multiple peptides with high affinities for PD-1/PD-L1 have been reported2; WL12 is a reported peptide that shows specific binding to PD-L134. Radiolabeled tracers, [64Cu]WL12, [68Ga]WL12, and [18F]FPyWL12, have been reported to show high in vivo specific tumor-targeting ability, which allows for the harvest of high-quality images of PD-L1 expression in tumors26,35,36,37. Moreover, the first in-human evaluation of radiolabeled WL12 has demonstrated that [68Ga]WL12 (chelated by NOTA) has a safe and efficient potential for clinical tumor imaging38. Due to its high hydrophobicity and high uptake in the healthy liver, WL12 has limited clinical use. Other radiolabeling peptides, such as TPP1 and SETSKSF, which specifically bind to PD-L1, have also showed potential stability and specificity to visualize whole-body PD-L1 expression39,40. However, unmodified peptides are easily degraded by proteases, and are rapidly metabolized by the kidney. Dextrorotary(D)-peptides have been widely used as effective mediators, due to the poor stability of left-handed (L)-peptides41,42,43. D-peptides are hyper-resistant to proteolytic degradation and have remarkably prolonged metabolic half-lives. Compared with their L-counterparts, D-peptides mostly show specific binding abilities44,45,46.

This study designed a new method to detect PD-L1 expression, based on PET imaging of a 68Ga-labeled PD-L1-targeted D-peptide, D-dodecapeptide antagonist (DPA), in a tumor-bearing mouse model47. The stability of [68Ga]DPA was first studied in phosphate-buffered saline (PBS) and mouse serum, after which the binding affinity of [68Ga]DPA in PD-L1-overexpressing tumors was tested. Thereafter, PET imaging was performed in glioblastoma xenograft models to confirm whether [68Ga]DPA was an ideal PET tracer to monitor PD-L1 expression in tumors. The combination of PET imaging and DPA not only provides a new approach to overcome challenges associated with the heterogeneous expression of PD-L1, but also lays the basis for the development of D-peptide-based radiotracers.

Protocol

The animal experimental procedures were approved by the Animal Ethics Committee of Nanjing Medical University or the National Institutes of Quantum Science and Technology. Mice experiments were strictly performed in compliance with the institutional guidelines of the Committee for the Care and Use of Laboratory Animals. 1. Peptide synthesis Swell 100 mg of 4-methylbenzhydrylamine (MBHA) resin (loading capacity of 0.37 mmol/g) in 1 mL of N-methyl-2-pyr…

Representative Results

[68Ga]DPA radiolabeling and stability The model peptide, DPA, is an effective PD-L1 antagonist. DOTA-DPA was obtained with >95% purity and a yield of 68%. The mass of DOTA-DPA is experimentally observed at 1,073.3 ([M+2H]2+). 68Gallium is considered a suitable radionuclide to label peptides for PET imaging, and therefore was chosen for this study. To radiolabel DPA with 68Ga (half-life: 68 min), DOTA-PEG3-DPA was synt…

Discussion

The critical steps described in this method include the efficient labeling of 68Ga to DPA and choosing a suitable time window for PET imaging, which must perfectly match the pharmacodynamic pattern of DPA in the tumor.

In contrast to IHC, PET imaging enables real-time detection of whole-body PD-L1 expression in a noninvasive manner, allowing the visualization of each positive area in a heterogeneous tumor6,7. Peptides were c…

Divulgations

The authors have nothing to disclose.

Acknowledgements

This study was supported by the Non-profit Central Research Institute Fund of Chinese Academy of Medical Sciences (no. 2022-RC350-04) and the CAMS Innovation Fund for Medical Sciences (nos. 2021-I2M-1-026, 2022-I2M-1-026-1, 02120101, 02130101, and 2022-I2M-2-002).

Materials

1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid (DOTA) Merck 60239-18-1 68Ga  chelation
3,3-diaminobenzidine (DAB) Kit Sigma-Aldrich D7304-1SET Immunohistochemistry
anti-PD-L1 monoclonal antibody Wuhan Proteintech 17952-1-ap Immunohistochemistry: primary antibody
BMS202 Selleck 1675203-84-5 Competitive binding assay: inhibitor
BSA Merck V900933 Immunofluorescent : blocking 
DAPI Merck D9542 Immunofluorescent: staining of nucleus
Dichloromethane (DCM) Merck 34856 Solvent
DIPEA Merck 3439 Peptide coupling
EDC·HCl Merck E6383 Activation of DOTA
FBS Gibco 10099 Cell culture: supplement
FITC-conjugated anti-human IgG Fc Antibody Biolegend 409310 Immunofluorescent: secondary antibody
FITC-conjugated anti PD-L1 antibody Biolegend 393606 Flow cytometry: direct antibody
HCTU Energy Chemical E070004-25g Peptide coupling
HRP labeled goat anti-rabbit antibody Servicebio GB23303 Immunohistochemistry: secondary antibody
Hydroxysuccinimide (NHS) Merck 130672 Activation of DOTA
MeCN Merck PHR1551 Solvent
Morpholine Merck 8.06127 Fmoc- deprotection
NMP Merck 8.06072 Solevent
Paraformaldehyde Merck 30525-89-4 Fixation of tissues
PBS Gibco 10010023 Cell culture: buffer
Penicillin-streptomycin  Gibco 10378016 Cell culture: supplement
RIA tube PolyLab P10301A As tissue sample container
RPMI-1640 medium Gibco 11875093 Cell culture: basic medium
Sodium acetate Merck 1.06264 Salt for buffer
Trypsin-EDTA Gibco 25200056 Cell culture: dissociation agent
U87MG cell line Procell Life Science & Technology Co CL-0238 Cell model
Equipment
68Ge/68Ga generator Isotope Technologies Munich, ITM Not applicable Generation of [68Ga]
Autogamma counter Perkin Elmer  Wizard2 Detection of radioactivity
Confocal fluorescent microscopy Keyence Observation of immunofluorescent results
Flow cytometer Becton Dickinson, BD LSRII Monitoring the PD-L1 positive cells
High-performance liquid chromatography (HPLC) SHIMAZU LC-20AT  Purification of DPA peptide
PET scanner Siemens Medical Solutions Inveon MultiModality System PET imaging
Optical microscopy Nikon  Eclipse E100 Observation of immunohistochemistry results
Solid phase peptide synthesizer Promega Vac-Man Laboratory Vacuum Manifold LOT#11101 Synthesis of DPA-DOTA peptide
Software
ASIPro Siemens Medical Solutions Not applicable Analysis of PET-CT results
FlowJo Becton Dickinson, BD FlowJo 7.6.1 Analysis of the flow cytometer results
Inveon Acquisition Workplace (IAW) Siemens Medical Solutions Not applicable Management of PET mechine
Prism Graphpad Prism 8.0 Analysis of the data 

References

  1. Doroshow, D. B., et al. PD-L1 as a biomarker of response to immune-checkpoint inhibitors. Natire Reviews Clinical Oncology. 18 (6), 345-362 (2021).
  2. Krutzek, F., Kopka, K., Stadlbauer, S. Development of radiotracers for imaging of the PD-1/PD-L1 axis. Pharmaceuticals. 15 (6), 747 (2022).
  3. Topalian, S. L., Drake, C. G., Pardoll, D. M. Immune checkpoint blockade: a common denominator approach to cancer therapy. Cancer Cell. 27 (4), 450-461 (2015).
  4. Pardoll, D. M. The blockade of immune checkpoints in cancer immunotherapy. Nature Reviews Cancer. 12 (4), 252-264 (2012).
  5. Xiao, Z., et al. PEIGel: A biocompatible and injectable scaffold with innate immune adjuvanticity for synergized local immunotherapy. Materials Today Bio. 15, 100297 (2022).
  6. Teng, M. W. L., Ngiow, S. F., Ribas, A., Smyth, M. J. Classifying cancers based on T-cell infiltration and PD-L1. Recherche en cancérologie. 75 (11), 2139-2145 (2015).
  7. Dolled-Filhart, M., et al. Development of a companion diagnostic for pembrolizumab in non-small cell lung cancer using immunohistochemistry for programmed death ligand-1. Archives of Pathology & Laboratory Medicine. 140 (11), 1243-1249 (2016).
  8. Meng, X. J., Huang, Z. Q., Teng, F. F., Xing, L. G., Yu, J. M. Predictive biomarkers in PD-1/PD-L1 checkpoint blockade immunotherapy. Cancer Treatment Reviews. 41 (10), 868-876 (2015).
  9. Hakozaki, T., Hosomi, Y., Kitadai, R., Kitagawa, S., Okuma, Y. Efficacy of immune checkpoint inhibitor monotherapy for patients with massive non-small-cell lung cancer. Journal of Cancer Research and Clinical Oncology. 146 (11), 2957-2966 (2020).
  10. Haslam, A., Prasad, V. Estimation of the percentage of US patients with cancer who are eligible for and respond to checkpoint inhibitor immunotherapy drugs. Jama Network Open. 2 (5), 192535 (2019).
  11. Willmann, J. K., van Bruggen, N., Dinkelborg, L. M., Gambhir, S. S. Molecular imaging in drug development. Nature Reviews Drug Discovery. 7 (7), 591-607 (2008).
  12. Zhang, L., et al. Recent developments on PET radiotracers for TSPO and their applications in neuroimaging. Acta Pharmaceutica Sinica B. 11 (2), 373-393 (2021).
  13. Sun, J., et al. Imaging-guided targeted radionuclide tumor therapy: From concept to clinical translation. Advanced Drug Delivery Reviews. 190, 114538 (2022).
  14. Xu, M., et al. Preclinical study of a fully human Anti-PD-L1 antibody as a theranostic agent for cancer immunotherapy. Molecular Pharmaceutics. 15 (10), 4426-4433 (2018).
  15. Niemeijer, A. N., et al. Whole body PD-1 and PD-L1 positron emission tomography in patients with non-small-cell lung cancer. Nature Communications. 9 (1), 4664 (2018).
  16. Mayer, A. T., et al. Practical immuno-PET radiotracer design considerations for human immune checkpoint imaging. Journal of Nuclear Medicine. 58 (4), 538-546 (2017).
  17. Lv, G., et al. PET Imaging of tumor PD-L1 expression with a highly specific nonblocking single-domain antibody. Journal of Nuclear Medicine. 61 (1), 117-122 (2020).
  18. Li, D., et al. Immuno-PET imaging of 89Zr labeled anti-PD-L1 domain antibody. Molecular Pharmaceutics. 15 (4), 1674-1681 (2018).
  19. Lesniak, W. G., et al. PD-L1 detection in tumors using [(64)Cu]Atezolizumab with PET. Bioconjugate Chemistry. 27 (64), 2103-2110 (2016).
  20. Kristensen, L. K., et al. CD4(+) and CD8a(+) PET imaging predicts response to novel PD-1 checkpoint inhibitor: studies of Sym021 in syngeneic mouse cancer models. Theranostics. 9 (26), 8221-8238 (2019).
  21. Christensen, C., Kristensen, L. K., Alfsen, M. Z., Nielsen, C. H., Kjaer, A. Quantitative PET imaging of PD-L1 expression in xenograft and syngeneic tumour models using a site-specifically labelled PD-L1 antibody. European Journal of Nuclear Medicine and Molecular Imaging. 47 (5), 1302-1313 (2020).
  22. Bensch, F., et al. 89Zr-atezolizumab imaging as a non-invasive approach to assess clinical response to PD-L1 blockade in cancer. Nature Medicine. 24 (12), 1852-1858 (2018).
  23. Gonzalez Trotter, D. E., et al. In vivo imaging of the programmed death ligand 1 by 18F PET. Journal of Nuclear Medicine. 58 (11), 1852-1857 (2017).
  24. Lv, G., et al. Promising potential of a 18F-labelled small-molecular radiotracer to evaluate PD-L1 expression in tumors by PET imaging. Bioorganic Chemistry. 115, 105294 (2021).
  25. Miao, Y., et al. One-step radiosynthesis and initial evaluation of a small molecule PET tracer for PD-L1 imaging. Bioorganic & Medicinal Chemical Letters. 30 (24), 127572 (2020).
  26. Kumar, D., et al. Peptide-based PET quantifies target engagement of PD-L1 therapeutics. The Journal of Clinical Investigation. 129 (2), 616-630 (2019).
  27. Powles, T., et al. MPDL3280A (anti-PD-L1) treatment leads to clinical activity in metastatic bladder cancer. Nature. 515 (7528), 558-562 (2014).
  28. Herbst, R. S., et al. Predictive correlates of response to the anti-PD-L1 antibody MPDL3280A in cancer patients. Nature. 515 (7528), 563-567 (2014).
  29. Marciscano, A. E., Gulley, J. L. Avelumab demonstrates promise in advanced NSCLC. Oncotarget. 8 (61), 102767-102768 (2017).
  30. Vaddepally, R. K., Kharel, P., Pandey, R., Garje, R., Chandra, A. B. Review of indications of FDA-approved immune checkpoint inhibitors per NCCN guidelines with the level of evidence. Cancers. 12 (3), 738 (2020).
  31. Antonia, S. J., et al. Durvalumab after chemoradiotherapy in stage III non-small-cell lung cancer. The New England Journal of Medicine. 377 (20), 1919-1929 (2017).
  32. Wang, D. Y., et al. Fatal toxic effects associated with immune checkpoint inhibitors: a systematic review and meta-analysis. JAMA Oncology. 4 (12), 1721-1728 (2018).
  33. Sgouros, G., Bodei, L., McDevitt, M. R., Nedrow, J. R. Radiopharmaceutical therapy in cancer: clinical advances and challenges. Nature Reviews Drug Discovery. 19 (9), 589-608 (2020).
  34. (US) M. M. M, et al. Macrocyclic inhibitors of the pd-1/pd-l1 and cd80(b7-1)/pd-l1 protein/protein interactions. United States patent. , (2014).
  35. Chatterjee, S., et al. Rapid PD-L1 detection in tumors with PET using a highly specific peptide. Biochemical and Biophysical Research Communications. 483 (1), 258-263 (2017).
  36. De Silva, R. A., et al. Peptide-based 68Ga-PET radiotracer for imaging PD-L1 expression in cancer. Molecular Pharmaceutics. 15 (9), 3946-3952 (2018).
  37. Lesniak, W. G., et al. Development of [18F]FPy-WL12 as a PD-L1 specific PET imaging peptide. Molecular Imaging. 18, 1536012119852189 (2019).
  38. Zhou, X., et al. First-in-humans evaluation of a PD-L1-binding peptide PET radiotracer in non-small cell lung cancer patients. Journal of Nuclear Medicine. 63 (4), 536-542 (2022).
  39. Hu, K., et al. Developing native peptide-based radiotracers for PD-L1 PET imaging and improving imaging contrast by pegylation. Chemical Communications. 55 (29), 4162-4165 (2019).
  40. Liu, H., et al. A novel small cyclic peptide-based 68Ga-Radiotracer for positron emission tomography imaging of PD-L1 expression in tumors. Molecular Pharmaceutics. 19 (1), 138-147 (2022).
  41. Rabideau, A. E., Pentelute, B. L. A D-amino acid at the N-terminus of a protein abrogates its degradation by the N-end rule pathway. ACS Central Science. 1 (8), 423-430 (2015).
  42. Uppalapati, M., et al. A potent D-protein antagonist of VEGF-A is nonimmunogenic, metabolically stable, and longer-circulating in vivo. ACS Chemical Biology. 11 (4), 1058-1065 (2016).
  43. Garton, M., et al. Method to generate highly stable D-amino acid analogs of bioactive helical peptides using a mirror image of the entire PDB. Proceedings of the National Academy of Sciences. 115 (7), 1505-1510 (2018).
  44. Jia, F. J., et al. D-amino acid substitution enhances the stability of antimicrobial peptide polybia-CP. Acta Biochimica et Biophysica Sinica. 49 (10), 916-925 (2017).
  45. Carmona, G., Rodriguez, A., Juarez, D., Corzo, G., Villegas, E. Improved protease stability of the antimicrobial peptide Pin2 substituted with D-amino acids. Protein Journal. 32 (6), 456-466 (2013).
  46. Feng, Z., Xu, B. Inspiration from the mirror: D-amino acid containing peptides in biomedical approaches. Biomolecular Concepts. 7 (3), 179-187 (2016).
  47. Hu, K., et al. Whole-body PET tracking of a d-dodecapeptide and its radiotheranostic potential for PD-L1 overexpressing tumors. Acta Pharmaceutica Sinica. B. 12 (3), 1363-1376 (2022).
  48. Qiu, X. Y., et al. PD-L1 confers glioblastoma multiforme malignancy via Ras binding and Ras/Erk/EMT activation. Biochimica Et Biophysica Acta. Molecular Basis of Disease. 1864, 1754-1769 (2018).
  49. Hu, K., et al. Development of a stable peptide-based PET tracer for detecting CD133-expressing cancer cells. ACS Omega. 7 (1), 334-341 (2021).
  50. Jin, Z. -. H., et al. Radiotheranostic agent 64Cu-cyclam-RAFT-c(-RGDfK-)4 for management of peritoneal metastasis in ovarian cancer. Clinical Cancer Research. 26 (23), 6230-6241 (2020).
  51. Hu, K., et al. Harnessing the PD-L1 interface peptide for positron emission tomography imaging of the PD-1 immune checkpoint. RSC Chemical Biology. 1 (4), 214-224 (2020).
  52. Hu, K., et al. PET imaging of VEGFR with a novel 64Cu-labeled peptide. ACS Omega. 5 (15), 8508-8514 (2020).
  53. Hu, K., et al. An in-tether chiral center modulates the helicity, cell permeability, and target binding affinity of a peptide. Angewandte Chemie International Edition. 55 (28), 8013-8017 (2016).
  54. Zhao, J., et al. Concurrent injection of unlabeled antibodies allows positron emission tomography imaging of programmed cell death ligand 1 expression in an orthotopic pancreatic tumor model. ACS Omega. 5 (15), 8474-8482 (2020).
  55. Moroz, A., et al. A preclinical assessment of 89Zr-atezolizumab identifies a requirement for carrier added formulations not observed with 89Zr-C4. Bioconjugate Chemistry. 29 (10), 3476-3482 (2018).
  56. Nedrow, J. R., et al. Imaging of programmed cell death ligand 1: impact of protein concentration on distribution of anti-PD-L1 SPECT agents in an immunocompetent murine model of melanoma. Journal of Nuclear Medicine. 58 (10), 1560-1566 (2017).
check_url/fr/65047?article_type=t

Play Video

Citer Cet Article
Zhang, L., Zhang, S., Wu, W., Wang, X., Shen, J., Wang, D., Hu, K., Zhang, M., Wang, F., Wang, R. Development of a 68Gallium-Labeled D-Peptide PET Tracer for Imaging Programmed Death-Ligand 1 Expression. J. Vis. Exp. (192), e65047, doi:10.3791/65047 (2023).

View Video