Preclinical Characterization of HPN536, a Trispecific, T-Cell− Activating Protein Construct for the Treatment of Mesothelin-Expressing Solid Tumors
Total Page:16
File Type:pdf, Size:1020Kb
Published OnlineFirst December 1, 2020; DOI: 10.1158/1078-0432.CCR-20-3392 CLINICAL CANCER RESEARCH | TRANSLATIONAL CANCER MECHANISMS AND THERAPY Preclinical Characterization of HPN536, a Trispecific, T-Cell–Activating Protein Construct for the Treatment of Mesothelin-Expressing Solid Tumors A C Mary Ellen Molloy1, Richard J. Austin1, Bryan D. Lemon1, Wade H. Aaron1, Vaishnavi Ganti1, Adrie Jones1, Susan D. Jones1, Kathryn L. Strobel1, Purbasa Patnaik1, Kenneth Sexton1, Laurie Tatalick1, Timothy Z. Yu1, Patrick A. Baeuerle1,2,3, Che-Leung Law1, and Holger Wesche1 ABSTRACT ◥ Purpose: Mesothelin (MSLN) is a glycophosphatidylinositol- ity, and half-life of HPN536 in in vitro assays, rodent models, and in linked tumor antigen overexpressed in a variety of malignancies, nonhuman primates (NHP). including ovarian, pancreatic, lung, and triple-negative breast can- Results: HPN536 binds to MSLN-expressing tumor cells and to cer. Early signs of clinical efficacy with MSLN-targeting agents CD3e on T cells, leading to T-cell activation and potent redirected have validated MSLN as a promising target for therapeutic inter- target cell lysis. A third domain of HPN536 binds to serum albumin vention, but therapies with improved efficacy are still needed to for extension of plasma half-life. In cynomolgus monkeys, HPN536 address the significant unmet medical need posed by MSLN- at doses ranging from 0.1 to 10 mg/kg demonstrated MSLN- expressing cancers. dependent pharmacologic activity, was well tolerated, and showed Experimental Design: We designed HPN536, a 53-kDa, tri- pharmacokinetics in support of weekly dosing in humans. specific, T-cell–activating protein-based construct, which binds to Conclusions: HPN536 is potent, is well tolerated, and exhibits MSLN-expressing tumor cells, CD3e on T cells, and to serum extended half-life in NHPs. It is currently in phase I clinical testing albumin. Experiments were conducted to assess the potency, activ- in patients with MSLN-expressing malignancies (NCT03872206). Introduction a tumor antigen, a sdAb specific for serum albumin for half-life extension, and a single-chain fragment variable (scFv) specific for the Redirection of cytotoxic T cells with bispecific antibody constructs CD3e subunit of the TCR complex (11). Their molecular size of 53 kDa for cancer therapy has been validated in the clinic (1–6). Blinatumo- is about one-third of that of an IgG. Binding of TriTACs to tumor mab is the first and thus far the only bispecific T-cell engager (BiTE) antigen and CD3e is monovalent, which minimizes off-target CD3e approved by the FDA (7). T-cell–engaging biologics function by clustering that can potentially lead to nonspecific T-cell activation. The forming an immunologic cytolytic synapse between cancer target cells absence of an Fc-gamma domain for half-life extension is functionally and T cells, which leads to target cell lysis independent of T-cell compensated by an albumin-binding domain. HPN424 (11) and receptor (TCR) specificity, peptide antigen presentation by HLA, and HPN536, the first two TriTACs are in phase I clinical testing in T-cell costimulation. Despite the clinical success of blinatumomab for hormone refractory prostate cancer and mesothelin (MSLN)-over- treating relapsed and refractory acute lymphoblastic leukemia, other expressing solid tumors, respectively. molecules, including BiTE antibodies, showed only limited activity in Human MSLN is produced as a 71-kDa precursor of 628 amino the treatment of solid tumors (8, 9). Their short plasma half-life acids, which is expressed as a glycophosphatidylinositol-linked cell required continuous intravenous infusion limiting their utility for surface glycoprotein. Its 31-kDa N-terminal domain is released as a most solid tumor indications. Novel designs for T-cell–engaging soluble protein, termed as the megakaryocyte potentiating factor antibodies aim at overcoming limitations of the first generation and (MPF), while the 40-kDa C-terminal domain remains attached to the are already being tested in clinical trials (10). plasma membrane as mature MSLN (12–14). MSLN expression on The Trispecific T-cell–Activating Construct (TriTAC) design has normal tissue is confined to the single-cell mesothelial layer covering been specifically developed to treat solid tumors (11). TriTACs consist the surface of tissues and organs of the pleural, pericardial, and of a single polypeptide chain aligning three humanized, antibody- peritoneal cavities (13, 15). MUC16/CA125 is a binding partner for derived binding domains: a single-domain antibody (sdAb) specific for MSLN, implicating a role for MSLN in cell adhesion (16, 17). However, the precise physiologic functions of MSLN have not been defined, and MSLN-knockout mice exhibit no detectable phenotype or develop- 1Harpoon Therapeutics, South San Francisco, California. 2MPM Capital, Cam- bridge, Massachusetts. 3Institute for Immunology, Ludwig-Maximilians Univer- mental abnormality (18). sity Munich, Planegg- Martinsried, Munich, Germany. MSLN is overexpressed in many malignancies, including ovarian cancer (13, 15, 19), pancreatic cancer (20, 21), non–small cell lung Note: Supplementary data for this article are available at Clinical Cancer – Research Online (http://clincancerres.aacrjournals.org/). cancer (22 26), triple-negative breast cancer (26, 27), and mesothe- lioma (28, 29). In triple-negative breast cancer (25) and in lung and Corresponding Author: Mary Ellen Molloy, Harpoon Therapeutics, 131 Oyster Point Boulevard, 300, South San Francisco, CA 94080. Phone: 773-318-0796; pancreatic adenocarcinomas (22, 23, 30), overexpression of MSLN E-mail: [email protected] correlates with poor prognosis. Differential expression of MSLN in cancer versus normal tissue has made it an attractive target for MSLN- Clin Cancer Res 2020;XX:XX–XX directed imaging agents and therapeutics (10, 31–33). A challenge in doi: 10.1158/1078-0432.CCR-20-3392 developing MSLN-directed therapeutics is the expression of MSLN on Ó2020 American Association for Cancer Research. normal mesothelial cells, potentially leading to dose-limiting toxicities. AACRJournals.org | OF1 Downloaded from clincancerres.aacrjournals.org on October 1, 2021. © 2020 American Association for Cancer Research. Published OnlineFirst December 1, 2020; DOI: 10.1158/1078-0432.CCR-20-3392 Molloy et al. loads, appropriate serial dilutions of known analyte concentrations, Translational Relevance and association and dissociation times were then fit globally to a one- Patients with mesothelin (MSLN)-overexpressing tumors, to-one binding model using Octet DataAnalysis 9.0 software. including ovarian, pancreatic, lung, and triple-negative breast cancer, have a high unmet clinical need. A number of MSLN- In vitro T-cell–dependent cell cytotoxicity and T-cell activation targeted therapeutics have been developed that show limited assays efficacy and safety in clinical trials. HPN536 is a novel, MSLN- T cells from healthy donors were purified from leukopaks (leuka- targeted, trispecific, T-cell–activating protein construct that can pheresis samples, StemCell Technologies) using EasySep Human potently redirect T cells to lyse tumor cells and was remarkably well T Cell Isolation Kits (StemCell Technologies, 17951) following the tolerated in nonhuman primates at single doses up to 10 mg/kg, manufacturer's instructions. All cancer cell lines were obtained from which is far above the expected therapeutic dose level. Our findings the ATCC, with the exception of OVCAR8 cells, which were obtained suggest that HPN536 has the potential for high clinical activity from the NCI (Bethesda, MD). Cell lines were passaged a maximum of and a wide therapeutic window. Its long serum half-life supports 36 times after being received from the ATCC. Cell line authentication once-weekly dosing in humans. Currently, HPN536 is the only and Mycoplasma testing were not performed. T-cell–dependent cell MSLN-targeting, T-cell–engaging biologic in clinical testing. cytotoxicity (TDCC) assays were performed as described previous- ly (35). Briefly, luciferase-expressing target cells and purified human T cells were seeded per well of a 384-well plate at a 10:1 T cell-to-target cell ratio. Target cell killing was assessed following incubation for fi o HPN536 speci cally redirects T cells for potent redirected lysis of 48 hours at 37 C and 5% CO2. Target cell viability was assessed by MSLN-expressing cancer cells with concomitant T-cell activation. In incubation with the SteadyGlo Reagent (Promega). Luminescence was three different mouse xenograft models, HPN536 induced durable measured using a PerkinElmer EnVision Detection System. Activated antitumor activity at very low doses. In cynomolgus monkeys, T cells were identified by CD69 and CD25 surface expression (BD HPN536 was well tolerated, showed a long serum half-life, and elicited Biosciences). Samples were analyzed on a FACSCelesta Flow Cyt- signs of target engagement on mesothelial structures. ometer (BD Biosciences). Flow cytometry data were processed using FlowJo v10 Software (FlowJo, LLC). Materials and Methods Binding of HPN536 on MSLN-expressing OVCAR and T cells Protein production Cultured cells were incubated with 1 mg/mL HPN536 or anti-GFP Sequences of TriTACs, sdAbs, and extracellular domains of target TriTAC (control) for 1 hour. Binding was detected using Alexa647- proteins fused to an Fc domain or a hexahistidine tag were cloned into anti-TriTAC antibody using a FACSCelesta Flow Cytometer (BD mammalian expression vector, pcDNA 3.4 (Invitrogen), preceded by a Biosciences).