89Zr-Labeled Anti-PD-L1 Antibody PET Monitors Gemcitabine Therapy-Induced Modulation of Tumor PD-L1 Expression

Visual Abstract We developed an 89Zr-labeled anti–programmed death ligand 1 (anti-PD-L1) immune PET that can monitor chemotherapy-mediated modulation of tumor PD-L1 expression in living subjects. Methods: Anti-PD-L1 underwent sulfohydryl moiety–specific conjugation with maleimide-deferoxamine followed by 89Zr radiolabeling. CT26 colon cancer cells and PD-L1–overexpressing CT26/PD-L1 cells underwent binding assays, flow cytometry, and Western blotting. In vivo pharmacokinetics, biodistribution, and PET imaging were evaluated in mice. Results: 89Zr-anti-PD-L1 synthesis was straightforward and efficient. Sodium dodecyl sulfate polyacrylamide gel electrophoresis showed that reduction produced half-antibody fragments, and matrix-assisted laser desorption ionization time-of-flight analysis estimated 2.18 conjugations per antibody, indicating specific conjugation at the hinge-region disulfide bonds. CT26/PD-L1 cells showed 102.2 ± 6.7-fold greater 89Zr-anti-PD-L1 binding than that of weakly expressing CT26 cells. Excellent target specificity was confirmed by a drastic reduction in binding by excess cold antibody. Intravenous 89Zr-anti-PD-L1 followed biexponential blood clearance. PET/CT image analysis demonstrated decreases in major organ activity over 7 d, whereas high CT26/PD-L1 tumor activity was maintained. Again, this was suppressed by excess cold antibody. Treatment of CT26 cells with gemcitabine for 24 h augmented PD-L1 protein to 592.4% ± 114.2% of the control level and increased 89Zr-anti-PD-L1 binding, accompanied by increased AKT (protein kinase B) activation and reduced phosphatase and tensin homolog (PTEN). In CT26 tumor–bearing mice, gemcitabine treatment substantially increased tumor uptake from 1.56% ± 0.48% to 6.24% ± 0.37% injected dose per gram (tumor-to-blood ratio, 34.7). Immunoblots revealed significant increases in tumor PD-L1 and activated AKT and a decrease in PTEN. Conclusion: 89Zr-anti-PD-L1 showed specific targeting with favorable imaging properties. Gemcitabine treatment upregulated cancer cell and tumor PD-L1 expression and increased 89Zr-anti-PD-L1 uptake. 89Zr-anti-PD-L1 PET may thus be useful for monitoring chemotherapy-mediated tumor PD-L1 modulation in living subjects.

[1]  Tao Yu,et al.  Application of molecular imaging technology in tumor immunotherapy. , 2020, Cellular immunology.

[2]  C. H. Nielsen,et al.  Quantitative PET imaging of PD-L1 expression in xenograft and syngeneic tumour models using a site-specifically labelled PD-L1 antibody , 2019, European Journal of Nuclear Medicine and Molecular Imaging.

[3]  Kyung-Han Lee,et al.  Troglitazone exerts metabolic and antitumor effects on T47D breast cancer cells by suppressing mitochondrial pyruvate availability. , 2019, Oncology reports.

[4]  J. Bussink,et al.  Tracers for non-invasive radionuclide imaging of immune checkpoint expression in cancer , 2019, EJNMMI Radiopharmacy and Chemistry.

[5]  E. Giovannetti,et al.  PTEN Alterations as a Potential Mechanism for Tumor Cell Escape from PD-1/PD-L1 Inhibition , 2019, Cancers.

[6]  B. Brodská,et al.  High PD-L1 Expression Predicts for Worse Outcome of Leukemia Patients with Concomitant NPM1 and FLT3 Mutations , 2019, International journal of molecular sciences.

[7]  S. Perkins,et al.  Prognostic Significance of the Dynamic Change of Programmed Death-ligand 1 Expression in Patients with Multiple Myeloma , 2019, Cureus.

[8]  Zhaoming Li,et al.  Predictive biomarkers for PD-1 and PD-L1 immune checkpoint blockade therapy. , 2019, Immunotherapy.

[9]  I. C. Kok,et al.  89Zr-atezolizumab imaging as a non-invasive approach to assess clinical response to PD-L1 blockade in cancer , 2018, Nature Medicine.

[10]  R. Stahel,et al.  Gemcitabine Synergizes with Immune Checkpoint Inhibitors and Overcomes Resistance in a Preclinical Model and Mesothelioma Patients , 2018, Clinical Cancer Research.

[11]  T. Schumacher,et al.  Regulation and Function of the PD-L1 Checkpoint. , 2018, Immunity.

[12]  Y. Naito,et al.  The JAK/STAT pathway is involved in the upregulation of PD-L1 expression in pancreatic cancer cell lines. , 2017, Oncology reports.

[13]  A. Ribas,et al.  Combination cancer immunotherapies tailored to the tumour microenvironment , 2016, Nature Reviews Clinical Oncology.

[14]  P. Millner,et al.  Considerations in producing preferentially reduced half-antibody fragments. , 2016, Journal of immunological methods.

[15]  S. Murphy,et al.  Chemotherapy Induces Programmed Cell Death-Ligand 1 Overexpression via the Nuclear Factor-κB to Foster an Immunosuppressive Tumor Microenvironment in Ovarian Cancer. , 2015, Cancer research.

[16]  H. Ishwaran,et al.  Radiation and Dual Checkpoint Blockade Activates Non-Redundant Immune Mechanisms in Cancer , 2015, Nature.

[17]  Shohei Koyama,et al.  Loss of Lkb1 and Pten leads to lung squamous cell carcinoma with elevated PD-L1 expression. , 2014, Cancer cell.

[18]  Antoni Ribas,et al.  Effects of MAPK and PI3K Pathways on PD-L1 Expression in Melanoma , 2014, Clinical Cancer Research.

[19]  I. Endo,et al.  Changes in the immune cell population and cell proliferation in peripheral blood after gemcitabine-based chemotherapy for pancreatic cancer , 2014, Clinical and Translational Oncology.

[20]  Jun Hu,et al.  PTEN Loss Increases PD-L1 Protein Expression and Affects the Correlation between PD-L1 Expression and Clinical Parameters in Colorectal Cancer , 2013, PloS one.

[21]  David C. Smith,et al.  Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. , 2012, The New England journal of medicine.

[22]  G. Freeman,et al.  The Programmed Death-1 Ligand 1:B7-1 Pathway Restrains Diabetogenic Effector T Cells In Vivo , 2011, The Journal of Immunology.

[23]  C. Liu,et al.  Cisplatin induces programmed death-1-ligand 1(PD-L1) over-expression in hepatoma H22 cells via Erk /MAPK signaling pathway. , 2010, Cellular and molecular biology.

[24]  Judith E. Flores,et al.  Site-specifically 89Zr-labeled monoclonal antibodies for ImmunoPET. , 2010, Nuclear medicine and biology.

[25]  Tae Woo Kim,et al.  Activation of Akt as a mechanism for tumor immune evasion. , 2009, Molecular therapy : the journal of the American Society of Gene Therapy.

[26]  G. Freeman,et al.  Programmed death-1 ligand 1 interacts specifically with the B7-1 costimulatory molecule to inhibit T cell responses. , 2007, Immunity.

[27]  S. Albelda,et al.  Gemcitabine has significant immunomodulatory activity in murine tumor models independent of its cytotoxic effects , 2007, Cancer biology & therapy.

[28]  Haidong Dong,et al.  Tumor-associated B7-H1 promotes T-cell apoptosis: A potential mechanism of immune evasion , 2002, Nature Medicine.

[29]  R. Herbst,et al.  Quantitative Assessment of the Heterogeneity of PD-L1 Expression in Non-Small-Cell Lung Cancer. , 2016, JAMA oncology.

[30]  P. Mischel,et al.  Loss of tumor suppressor PTEN function increases B7-H1 expression and immunoresistance in glioma , 2007, Nature Medicine.