⁸⁹Zr-radiopharmaceuticals to study whole-body distribution and response to antibody-based cancer immunotherapies

Purpose: Probody therapeutic CX-072 is a protease-activatable antibody that is cross-reactive with murine and human programmed death-ligand 1 (PD-L1). CX-072 can be activated in vivo by proteases present in the tumor microenvironment, thereby potentially reducing peripheral, anti–PD-L1-mediated toxicities. To study its targeting of PD-L1–expressing tissues, we radiolabeled CX-072 with the PET isotope zirconium-89 ( 89 Zr). Experimental Design: 89 Zr-labeled CX-072, nonspecific Probody control molecule (PbCtrl) and CX-072 parental antibody (CX-075) were injected in BALB/c nude mice bearing human MDA-MB-231 tumors or C57BL/6J mice bearing syngeneic MC38 tumors. Mice underwent serial PET imaging 1, 3, and 6 days after intravenous injection (pi), followed by ex vivo biodistribution. Intratumoral 89 Zr-CX-072 distribution was studied by autoradiography on tumor tissue sections, which were subsequently stained for PD-L1 by IHC. Activated CX-072 species in tissue lysates were detected by Western capillary electrophoresis. Results: PET imaging revealed 89 Zr-CX-072 accumulation in MDA-MB-231 tumors with 2.1-fold higher tumor-to-blood ratios at 6 days pi compared with 89 Zr-PbCtrl. Tumor tissue autoradiography showed high 89 Zr-CX-072 uptake in high PD-L1–expressing regions. Activated CX-072 species were detected in these tumors, with 5.3-fold lower levels found in the spleen. Furthermore, 89 Zr-CX-072 uptake by lymphoid tissues of immune-competent mice bearing MC38 tumors was low compared with 89 Zr-CX-075, which lacks the Probody design. Conclusions: 89 Zr-CX-072 accumulates specifically in PD-L1–expressing tumors with limited uptake in murine peripheral lymphoid tissues. Our data may enable clinical evaluation of 89 Zr-CX-072 whole-body distribution as a tool to support CX-072 drug development (NCT03013491). We performed a PET imaging study in murine models with 89 Zr-labeled CX-072 to reveal its whole-body distribution. Also, we compared 89 Zr-CX-072 targeting of tumor and lymphoid tissues in both an immune-compromised and an immune-competent setting. To enable clinical PET imaging of 89 Zr-CX-072 distribution to tumor and lymphoid tissues in patients, we characterized and developed a good manufacturing practice (GMP)–compliant tracer. These irAEs may be caused by immune checkpoint blockade in healthy tissues, and immune checkpoint–inhibiting antibodies with tumor-restricted activity are therefore of interest. Recently, imaging of 89 Zr-atezolizumab whole-body distribution in patients with cancer showed high uptake in healthy lymphoid tissues, including spleen, lymph nodes, and Waldeyer's ring. Our preclinical imaging study in mice reveals anti– PD-L1 Probody therapeutic CX-072 is preferentially activated in tumors, followed by PD-L1–mediated uptake, whereas accumulation in spleen and other PD-L1–expressing peripheral lymphoid tissues is limited. These findings demonstrate CX-072 may reduce anti–PD-L1-mediated toxicities in healthy tissues, thereby potentially expanding its use in combination therapies. We developed and characterized clinical grade 89 Zr-CX-072, which is currently studied in patients as part of a phase I/II clinical trial (NCT03013491) to support CX-072 drug development. internalization in MDA-MB-231 and MC38 cell lines 89 Zr-CX-075 was used for internalization experiments because it acts as a surrogate for potential internalization of 89 Zr-CX-072 after removal of its masking peptide by tumor-associated proteases. Internalization in MDA-MB-231 and MC38 cells was assessed by adding 50 ng 89 Zr-CX-075 (200 MBq/mg) to 1×10 6 cells. For control of binding without internalization, cells were incubated for 1 hour on ice. Cells were subsequently washed with ice-cold 1% human serum albumin (Sanquin) in PBS. Total PD-L1–bound 89 Zr-CX-075 was determined by measuring cell-associated activity in a calibrated well-type gamma counter (LKB instruments) followed by incubation for 1 and 2 hours at 37 °C, whereas controls were kept on ice. Cells were stripped of cell-surface–bound antibody using 0.05 mol/L glycine and 0.1 mol/L sodium chloride (pH 2.8) and subsequently counted for remaining cell-associated activity, ie, internalized PD-L1– bound 89 Zr-CX-075, in a calibrated well-type gamma counter. Internalization was expressed as percentage of total PD-L1–bound 89 Zr-CX-075, corrected for internalization in 4 °C controls. PD-L1–expressing xenografts limited uptake in lymphoid hypothesis that CX-072 anti–PD-L1-mediated toxicities in

[1]  O. Vasiljeva,et al.  The multifaceted roles of tumor-associated proteases and harnessing their activity for prodrug activation , 2019, Biological chemistry.

[2]  P. Choyke,et al.  Immuno-PET Imaging of the Programmed Cell Death-1 Ligand (PD-L1) Using a Zirconium-89 Labeled Therapeutic Antibody, Avelumab , 2019, Molecular imaging.

[3]  J. Bussink,et al.  PD-L1 microSPECT/CT Imaging for Longitudinal Monitoring of PD-L1 Expression in Syngeneic and Humanized Mouse Models for Cancer , 2018, Cancer Immunology Research.

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

[5]  Xuelei Ma,et al.  Safety and efficacy of durvalumab (MEDI4736) in various solid tumors , 2018, Drug design, development and therapy.

[6]  A. Bertaut,et al.  Phase Ib/II trial evaluating the safety, tolerability and immunological activity of durvalumab (MEDI4736) (anti-PD-L1) plus tremelimumab (anti-CTLA-4) combined with FOLFOX in patients with metastatic colorectal cancer , 2018, ESMO Open.

[7]  J. Szustakowski,et al.  Nivolumab plus Ipilimumab in Lung Cancer with a High Tumor Mutational Burden , 2018, The New England journal of medicine.

[8]  Bohuslav Melichar,et al.  Nivolumab plus Ipilimumab versus Sunitinib in Advanced Renal‐Cell Carcinoma , 2018, The New England journal of medicine.

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

[10]  Dan Li,et al.  Immuno-PET Imaging of 89Zr Labeled Anti-PD-L1 Domain Antibody. , 2018, Molecular pharmaceutics.

[11]  M. Sawyer,et al.  Durable Clinical Benefit With Nivolumab Plus Ipilimumab in DNA Mismatch Repair-Deficient/Microsatellite Instability-High Metastatic Colorectal Cancer. , 2018, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[12]  Matthew D. Hellmann,et al.  Immune‐Related Adverse Events Associated with Immune Checkpoint Blockade , 2018, The New England journal of medicine.

[13]  C. Craik,et al.  Imaging PD-L1 Expression with ImmunoPET , 2017, Bioconjugate chemistry.

[14]  D. Schadendorf,et al.  Overall Survival with Combined Nivolumab and Ipilimumab in Advanced Melanoma , 2017, The New England journal of medicine.

[15]  R. Ferris,et al.  Preclinical immunoPET/CT imaging using Zr-89-labeled anti-PD-L1 monoclonal antibody for assessing radiation-induced PD-L1 upregulation in head and neck cancer and melanoma , 2017, Oncoimmunology.

[16]  G. Sgouros,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 , 2017, The Journal of Nuclear Medicine.

[17]  S. Dow,et al.  Regulation of PD-L1 expression on murine tumor-associated monocytes and macrophages by locally produced TNF-α , 2017, Cancer Immunology, Immunotherapy.

[18]  R. Bourgon,et al.  Atezolizumab as first-line treatment in cisplatin-ineligible patients with locally advanced and metastatic urothelial carcinoma: a single-arm, multicentre, phase 2 trial , 2017, The Lancet.

[19]  Wojciech G. Lesniak,et al.  PD-L1 Detection in Tumors Using [(64)Cu]Atezolizumab with PET. , 2016, Bioconjugate chemistry.

[20]  M. Bartholomä,et al.  High-Resolution PET Imaging with Therapeutic Antibody-based PD-1/PD-L1 Checkpoint Tracers , 2016, Theranostics.

[21]  Wojciech G. Lesniak,et al.  A humanized antibody for imaging immune checkpoint ligand PD-L1 expression in tumors , 2016, Oncotarget.

[22]  George Sgouros,et al.  Imaging, Biodistribution, and Dosimetry of Radionuclide-Labeled PD-L1 Antibody in an Immunocompetent Mouse Model of Breast Cancer. , 2016, Cancer research.

[23]  O. Vasiljeva,et al.  In vivo imaging of protease activity by Probody therapeutic activation , 2015, Biochimie.

[24]  W. Oyen,et al.  Noninvasive Imaging of Tumor PD-L1 Expression Using Radiolabeled Anti-PD-L1 Antibodies. , 2015, Cancer research.

[25]  Paul H. Bessette,et al.  Tumor-Specific Activation of an EGFR-Targeting Probody Enhances Therapeutic Index , 2013, Science Translational Medicine.

[26]  S. Hanash,et al.  Imaging a functional tumorigenic biomarker in the transformed epithelium , 2012, Proceedings of the National Academy of Sciences.

[27]  Simon-Peter Williams Tissue Distribution Studies of Protein Therapeutics Using Molecular Probes: Molecular Imaging , 2012, The AAPS Journal.

[28]  Z. Werb,et al.  Matrix Metalloproteinases: Regulators of the Tumor Microenvironment , 2010, Cell.

[29]  J. Belizário Immunodeficient Mouse Models: An Overview , 2009 .

[30]  S. Sorrenti,et al.  The urokinase plasminogen activator system: a target for anti-cancer therapy. , 2009, Current cancer drug targets.

[31]  H. Hollema,et al.  In Vivo VEGF Imaging with Radiolabeled Bevacizumab in a Human Ovarian Tumor Xenograft , 2007, Journal of Nuclear Medicine.

[32]  R. Boellaard,et al.  89Zr immuno-PET: comprehensive procedures for the production of 89Zr-labeled monoclonal antibodies. , 2003, Journal of nuclear medicine : official publication, Society of Nuclear Medicine.

[33]  M. M. Bradford A rapid and sensitive method for the quantitation of microgram quantities of protein utilizing the principle of protein-dye binding. , 1976, Analytical biochemistry.

[34]  M. Kavanaugh,et al.  Abstract A081: A PD-L1-targeted Probody provides antitumor efficacy while minimizing induction of systemic autoimmunity , 2016 .

[35]  J. Wolchok,et al.  Combined Nivolumab and Ipilimumab or Monotherapy in Untreated Melanoma. , 2015, The New England journal of medicine.