ABBV-011, A Novel, Calicheamicin-Based Antibody-Drug Conjugate, Targets SEZ6 to Eradicate Small Cell Lung Cancer Tumors

Wolf R Wiedemeyer, Julia Gavrilyuk, Alexander Schammel, Xi Zhao, Hetal Sarvaiya, Marybeth Pysz, Christine Gu, Monica You, Kumiko Isse, Theodore Sullivan, Dorothy French, Christina Lee, Angeline T Dang, Zhaomei Zhang, Monette Aujay, Alexander J Bankovich, Philip Vitorino, Wolf R Wiedemeyer, Julia Gavrilyuk, Alexander Schammel, Xi Zhao, Hetal Sarvaiya, Marybeth Pysz, Christine Gu, Monica You, Kumiko Isse, Theodore Sullivan, Dorothy French, Christina Lee, Angeline T Dang, Zhaomei Zhang, Monette Aujay, Alexander J Bankovich, Philip Vitorino

Abstract

In the past year, four antibody-drug conjugates (ADC) were approved, nearly doubling the marketed ADCs in oncology. Among other attributes, successful ADCs optimize targeting antibody, conjugation chemistry, and payload mechanism of action. Here, we describe the development of ABBV-011, a novel SEZ6-targeted, calicheamicin-based ADC for the treatment of small cell lung cancer (SCLC). We engineered a calicheamicin conjugate that lacks the acid-labile hydrazine linker that leads to systemic release of a toxic catabolite. We then screened a patient-derived xenograft library to identify SCLC as a tumor type with enhanced sensitivity to calicheamicin ADCs. Using RNA sequencing (RNA-seq) data from primary and xenograft SCLC samples, we identified seizure-related homolog 6 (SEZ6) as a surface-expressed SCLC target with broad expression in SCLC and minimal normal tissue expression by both RNA-seq and IHC. We developed an antibody targeting SEZ6 that is rapidly internalized upon receptor binding and, when conjugated to the calicheamicin linker drug, drives potent tumor regression in vitro and in vivo. These preclinical data suggest that ABBV-011 may provide a novel treatment for patients with SCLC and a rationale for ongoing phase I studies (NCT03639194).

©2022 The Authors; Published by the American Association for Cancer Research.

Figures

Figure 1.
Figure 1.
A, Modified linker drug chemistry in LD19.10 to eliminate toxic DMZ catabolite from calicheamicin linker drug. Chemical structure and metabolic pathway for the 19.10 linker drug in ABBV-011 compared with the acid-labile linker drug used in clinically approved calicheamicin-based ADCs. B, RNA expression of ubiquitously expressed surface antigen CD46 across PDX of varying indications by microarray analysis. C, PDX tumor response to a single dose of anti-CD46 antibody conjugated to LD19.10 calicheamicin linker drug in murine tumor models derived from multiple tumor indications. Animals were dosed with 2 (SCLC PDX top half-filled circles), 4 (SCLC PDX filled circles), or 8 mg/kg (all others) as indicated.
Figure 2.
Figure 2.
SEZ6 is expressed in NE SCLC with minimal expression in normal tissues. A, Comparison of pairwise Pearson correlation between CHGA and all cell surface markers in PDX setting (y-axis) and in human primary setting combined across three published human cohorts (refs. ; x-axis). B, Microarray data (Affymetrix Clariom D platform) of PDX shows SEZ6 expression is high in SCLC PDX, but low in normal tissues and other cancers. C, SEZ6 had upregulated expression in human SCLC tumors (n = 79) compared with the normal reference (n = 7) from the SCLC79 cohort (16). D, Hierarchical clustering with Euclidean distance and Ward linkage on expression of genes of interest in the Ucologne cohort (15).
Figure 3.
Figure 3.
SEZ6 is expressed in primary SCLC samples by IHC. A, Representative images of SEZ6 IHC analysis of primary human SCLC tumor samples with a range of target expression (from left to right: sez6 negative, weakly positive, moderately positive, and strongly positive). Arrows indicate areas of brown SEZ6 staining, with arrow heads indicating specific membranous staining. B, Quantitative analysis of percent positivity (left) and H-score (right) across 73 primary human SCLC samples. Scatter plot indicates distribution, and horizontal bar indicates mean. C, Representative images of SEZ6 IHC staining across normal human tissues. Arrows indicate staining in parasympathetic ganglia of the large intestine.
Figure 4.
Figure 4.
SC17 is a SEZ6-specific antibody that internalizes when bound to SEZ6 on the surface of SCLC cells. A, Parental 293T and 293T cells overexpressing human SEZ6, H69 cells and SEZ6 KO H69 cells, and SCLC PDX models, LU149, LU64, LU95, and LU505, were stained with SC17 or control IgG antibody conjugated to the fluorophore PE. Specific SEZ6 cell surface staining is observed with SC17-PE but not with IgG-PE in 293T cells overexpressing SEZ6, in naïve H69 cells, and in the two SCLC PDX models. B, Time course fluorescence microscopy with SC17-A647 in A549 cells engineered to express human SEZ6 and lysosomal protein LAMP1 fused to mRUBY (red signal). At time 0, SC17 localized to the outer membrane of the cell (green signal). After a 4-hour incubation period, we observed strong intracellular staining of SC17-A647 and colocalization with lysosomes (yellow signal). C, Time course quantitation of the lysosomal colocalization of SC17-A647 compared with CD46-A647 and IgG-A647 (negative control).
Figure 5.
Figure 5.
ABBV-011 potently and selectively kills SEZ6-positive cells in vitro. A, Schematic depiction of the ABBV-011 ADC. ABBV-011 consists of mAb SC17 conjugated to two molecules of LD19.10 linker drug (DAR2). B, 293T-SEZ6 cells were exposed to increasing concentrations of ABBV-011, nontargeted control ADC, unconjugated SC17 antibody targeting SEZ6, a nontargeted control antibody, or free calicheamicin. Percentage of live cells was determined by a luminometric viability assay. C, Parental NCI-H69 cells and SEZ6-KO NCI-H69 cells were exposed to increasing concentrations of ABBV-011 or nontargeted control ADC. Percentage of live cells was determined by a luminometric viability assay.
Figure 6.
Figure 6.
ABBV-011 induces dose-dependent tumor regression in SEZ6-positive SCLC PDX models. Antitumor activity in SEZ6-positive LU64 (A), LU95 (B), and LU149 (C) SCLC PDX models when mice are treated with indicated doses of ABBV-011 (red), cisplatin + etoposide (blue), or controls (gray). D, Lack of antitumor response in SEZ6-negative LU505 SCLC PDX. Mice treated with ABBV-011 (red), cisplatin + etoposide (blue), or CD46-targeted calicheamicin ADC (black).

References

    1. Lee M, Ellestad GA, Borders DB. Calicheamicins: discovery, structure, chemistry, and interaction with DNA. Acc Chem Res 1991;24.8:235–43.
    1. Nicolaou KC, Rigol S. The role of organic synthesis in the emergence and development of antibody-drug conjugates as targeted cancer therapies. Angew Chem Int Ed Engl 2019;58:11206–41.
    1. Appelbaum FR, Bernstein ID. Gemtuzumab ozogamicin for acute myeloid leukemia. Blood 2017;130:2373–6.
    1. Hamann PR, Hinman LM, Hollander I, Beyer CF, Lindh D, Holcomb R, et al. . Gemtuzumab ozogamicin, a potent and selective anti-CD33 antibody-calicheamicin conjugate for treatment of acute myeloid leukemia. Bioconjug Chem 2002;13:47–58.
    1. Dowell JA, Korth-Bradley J, Liu H, King SP, Berger MS. Pharmacokinetics of gemtuzumab ozogamicin, an antibody-targeted chemotherapy agent for the treatment of patients with acute myeloid leukemia in first relapse. J Clin Pharmacol 2001;41:1206–14.
    1. Giles FJ, Kantarjian HM, Kornblau SM, Thomas DA, Garcia-Manero G, Waddelow TA, et al. . Mylotarg (gemtuzumab ozogamicin) therapy is associated with hepatic venoocclusive disease in patients who have not received stem cell transplantation. Cancer 2001;92:406–13.
    1. Garrido-Laguna I, Krop I, Burris HA 3rd, Hamilton E, Braiteh F, Weise AM, et al. . First-in-human, phase I study of PF-06647263, an anti-EFNA4 calicheamicin antibody-drug conjugate, in patients with advanced solid tumors. Int J Cancer 2019;145:1798–808.
    1. Epenetos AA, Snook D, Durbin H, Johnson PM, Taylor-Papadimitriou J. Limitations of radiolabeled monoclonal antibodies for localization of human neoplasms. Cancer Res 1986;46:3183–91.
    1. Rekhtman N. Neuroendocrine tumors of the lung: an update. Arch Pathol Lab Med 2010;134:1628–38.
    1. Foster NR, Qi Y, Shi Q, Krook JE, Kugler JW, Jett JR, et al. . Tumor response and progression-free survival as potential surrogate endpoints for overall survival in extensive stage small-cell lung cancer: findings on the basis of North Central Cancer Treatment Group trials. Cancer 2011;117:1262–71.
    1. Augustyn A, Borromeo M, Wang T, Fujimoto J, Shao C, Dospoy PD, et al. . ASCL1 is a lineage oncogene providing therapeutic targets for high-grade neuroendocrine lung cancers. Proc Natl Acad Sci U S A 2014;111:14788–93.
    1. Kasprzak A, Zabel M, Biczysko W. Selected markers (chromogranin A, neuron-specific enolase, synaptophysin, protein gene product 9.5) in diagnosis and prognosis of neuroendocrine pulmonary tumours. Pol J Pathol 2007;58:23–33.
    1. Jensen SM, Gazdar AF, Cuttitta F, Russell EK, Linnoila RI. A comparison of synaptophysin, chromogranin, and L-dopa decarboxylase as markers for neuroendocrine differentiation in lung cancer cell lines. Cancer Res 1990;50:6068–74.
    1. Irizarry RA, Hobbs B, Collin F, Beazer-Barclay YD, Antonellis KJ, Scherf U, et al. . Exploration, normalization, and summaries of high density oligonucleotide array probe level data. Biostatistics 2003;4:249–64.
    1. George J, Lim JS, Jang SJ, Cun Y, Ozretic L, Kong G, et al. . Comprehensive genomic profiles of small cell lung cancer. Nature 2015;524:47–53.
    1. Jiang L, Huang J, Higgs BW, Hu Z, Xiao Z, Yao X, et al. . Genomic landscape survey identifies SRSF1 as a key oncodriver in small cell lung cancer. PLoS Genet 2016;12:e1005895.
    1. Clinical Lung Cancer Genome Project, Network Genomic Medicine. A genomics-based classification of human lung tumors. Sci Transl Med 2013;5:209ra153.
    1. Team RC. R: A language and environment for statistical computing. Vienna, Austria: R Foundation for Statistical Computing; 2013.
    1. Diehl KH, Hull R, Morton D, Pfister R, Rabemampianina Y, Smith D, et al. . A good practice guide to the administration of substances and removal of blood, including routes and volumes. J Appl Toxicol 2001;21:15–23.
    1. Anderson WC, Boyd MB, Aguilar J, Pickell B, Laysang A, Pysz MA, et al. . Initiation and characterization of small cell lung cancer patient-derived xenografts from ultrasound-guided transbronchial needle aspirates. PLoS One 2015;10:e0125255.
    1. AbbVie Stemcentrx LLC, assignee. Anti-Sez6 antibody drug conjugates and methods of use patent WO/2019/232241; 2019.
    1. AbbVie Stemcentrx LLC, assignee. Anti-Sez6 antibody drug conjugates and methods of use; 2021.
    1. Larson RA, Sievers EL, Stadtmauer EA, Lowenberg B, Estey EH, Dombret H, et al. . Final report of the efficacy and safety of gemtuzumab ozogamicin (Mylotarg) in patients with CD33-positive acute myeloid leukemia in first recurrence. Cancer 2005;104:1442–52.
    1. Yurkiewicz IR, Muffly L, Liedtke M. Inotuzumab ozogamicin: a CD22 mAb-drug conjugate for adult relapsed or refractory B-cell precursor acute lymphoblastic leukemia. Drug Des Devel Ther 2018;12:2293–300.
    1. Beck A, Goetsch L, Dumontet C, Corvaia N. Strategies and challenges for the next generation of antibody-drug conjugates. Nat Rev Drug Discov 2017;16:315–37.
    1. Elvington M, Liszewski MK, Atkinson JP. CD46 and oncologic interactions: Friendly fire against cancer. Antibodies 2020;9:59.
    1. Eichhorn JH, Lawrence WD, Young RH, Scully RE. Ovarian neuroendocrine carcinomas of non-small-cell type associated with surface epithelial adenocarcinomas. A study of five cases and review of the literature. Int J Gynecol Pathol 1996;15:303–14.
    1. Travis WD, Linnoila RI, Tsokos MG, Hitchcock CL, Cutler GB Jr, Nieman L, et al. . Neuroendocrine tumors of the lung with proposed criteria for large-cell neuroendocrine carcinoma. An ultrastructural, immunohistochemical, and flow cytometric study of 35 cases. Am J Surg Pathol 1991;15:529–53.
    1. Shimizu-Nishikawa K, Kajiwara K, Sugaya E. Cloning and characterization of seizure-related gene, SEZ-6. Biochem Biophys Res Commun 1995;216:382–9.
    1. Gunnersen JM, Kim MH, Fuller SJ, De Silva M, Britto JM, Hammond VE, et al. . Sez-6 proteins affect dendritic arborization patterns and excitability of cortical pyramidal neurons. Neuron 2007;56:621–39.
    1. Kudoh S, Tenjin Y, Kameyama H, Ichimura T, Yamada T, Matsuo A, et al. . Significance of achaete-scute complex homologue 1 (ASCL1) in pulmonary neuroendocrine carcinomas; RNA sequence analyses using small cell lung cancer cells and Ascl1-induced pulmonary neuroendocrine carcinoma cells. Histochem Cell Biol 2020;153:443–56.
    1. Rudin CM, Poirier JT, Byers LA, Dive C, Dowlati A, George J, et al. . Molecular subtypes of small cell lung cancer: a synthesis of human and mouse model data. Nat Rev Cancer 2019;19:289–97.
    1. Pigoni M, Hsia HE, Hartmann J, Rudan Njavro J, Shmueli MD, Muller SA, et al. . Seizure protein 6 controls glycosylation and trafficking of kainate receptor subunits GluK2 and GluK3. EMBO J 2020;39:e103457.
    1. Okamoto H, Oitate M, Hagihara K, Shiozawa H, Furuta Y, Ogitani Y, et al. . Pharmacokinetics of trastuzumab deruxtecan (T-DXd), a novel anti-HER2 antibody-drug conjugate, in HER2-positive tumour-bearing mice. Xenobiotica 2020;50:1242–50.
    1. Gazdar AF, Bunn PA, Minna JD. Small-cell lung cancer: what we know, what we need to know and the path forward. Nat Rev Cancer 2017;17:765.
    1. Ireland AS, Micinski AM, Kastner DW, Guo B, Wait SJ, Spainhower KB, et al. . MYC drives temporal evolution of small cell lung cancer subtypes by reprogramming neuroendocrine fate. Cancer Cell 2020;38:60–78.
    1. Walker S, Landovitz R, Ding WD, Ellestad GA, Kahne D. Cleavage behavior of calicheamicin gamma 1 and calicheamicin T. Proc Natl Acad Sci U S A 1992;89:4608–12.
    1. Tang CH, Parham C, Shocron E, McMahon G, Patel N. Picoplatin overcomes resistance to cell toxicity in small-cell lung cancer cells previously treated with cisplatin and carboplatin. Cancer Chemother Pharmacol 2011;67:1389–400.

Source: PubMed

3
Se inscrever