Abstract Adenoid cystic carcinoma (ACC) is an aggressive salivary gland malignancy with limited treatment options for recurrent or metastatic disease. Due to chemotherapy resistance and lack of targeted therapeutic approaches, current treatment options for the localized disease are limited to surgery and radiation, which fails to prevent locoregional recurrences and distant metastases in over 50% of patients. Approximately 20% of patients with ACC carry NOTCH-activating mutations that are associated with a distinct phenotype, aggressive disease, and poor prognosis. Given the role of NOTCH signaling in regulating tumor cell behavior, NOTCH inhibitors represent an attractive potential therapeutic strategy for this subset of ACC. AL101 (osugacestat) is a potent γ-secretase inhibitor that prevents activation of all four NOTCH receptors. While this investigational new drug has demonstrated antineoplastic activity in several preclinical cancer models and in patients with advanced solid malignancies, we are the first to study the therapeutic benefit of AL101 in ACC. Here, we describe the antitumor activity of AL101 using ACC cell lines, organoids, and patient-derived xenograft models. Specifically, we find that AL101 has potent antitumor effects in in vitro and in vivo models of ACC with activating NOTCH1 mutations and constitutively upregulated NOTCH signaling pathway, providing a strong rationale for evaluation of AL101 in clinical trials for patients with NOTCH-driven relapsed/refractory ACC. Subject terms: Head and neck cancer, Targeted therapies Introduction Adenoid cystic carcinoma (ACC) is a relatively uncommon secretory gland malignancy with a high propensity for perineural invasion, locoregional recurrence, and distant metastasis despite curative-intent treatment. Due to its insidious infiltrative growth pattern, ACC is often advanced by the time of clinical recognition. The therapeutic management for patients with locoregional and recurrent/metastatic ACC is limited to surgery and radiation, as no systemic agent has been found to be effective in improving long-term disease control [[62]1]. The mortality rate of ACC remains high, with over 60% of the patients succumbing to the disease within 15 years of diagnosis. Therefore, new therapeutic approaches for treating ACC are urgently needed. Like other malignancies, ACC is thought to arise through genetic and epigenetic aberrations that lead to the mis-expression of tumor suppressor genes and oncogenes. Molecular and genetic characterization of ACC has begun to reveal the most common driver mutations in this still incompletely understood cancer [[63]2–[64]5]. One common driver mutation in ACC consists of chromosomal rearrangements that produce MYB-NFIB (~70% of the tumors) or MYBL1-NFIB fusion genes, which appear to have a central role in the genesis of ACC. While representing useful markers in the diagnosis of ACC, MYB family members, like other transcription factors, remain difficult therapeutic targets, and the prognostic and biologic significance of MYB fusion genes are uncertain [[65]6]. Other driver mutations in ACC involve genes that encode epigenetic regulators (e.g., MLL2, MLL3, EP300, SMARCA2, SMARCC1, CREBBP, and KDM6A), pro-growth factors (e.g., FGFR2, PIK3CA, MYC, KRAS, BRAF), and DNA damage/checkpoint regulators (ATM, CDKN2A, TP53) [[66]2–[67]5, [68]7]. Recurrent copy number losses (e.g., of chromosome 1p36) and amplifications (e.g., of chromosome 7p14.1 and 14q11.2) have also been noted [[69]2–[70]5]. However, the fraction of ACC with mutations in these genes that present opportunities for targeted therapy is small. By contrast, recently emerging data suggest that the NOTCH pathway is a tractable, rational therapeutic target in ACC [[71]7, [72]8]. The NOTCH gene family contains four paralogs encoding large type 1 transmembrane receptor signaling proteins, NOTCH1, 2, 3, and 4. Normal NOTCH signaling is initiated by binding the receptor to a ligand (JAG1, JAG2, DLL1, or DLL4) on an adjacent cell. This event elicits conformational changes in the extracellular domain of NOTCH that makes it susceptible to successive cleavages by ADAM10 and γ-secretase. The latter cleavage releases the NOTCH intracellular domain (NICD), which migrates to the nucleus and forms a transcription complex that upregulates the expression of downstream target genes. These upregulated genes, in turn, regulate diverse cellular functions in a context-specific fashion, many of which have the potential to influence the behavior of cancers through both cell autonomous and non-autonomous mechanisms [[73]9–[74]13]. Particular cancers are marked by the presence of NOTCH-activating mutations, which either disrupt the extracellular NOTCH negative regulatory region (NRR), leading to ligand-independent proteolysis and NICD generation, or a C-terminal PEST domain, leading to stabilization of NICD [[75]9]. Among these cancers is ACC, in which activating NOTCH mutations are found in ~20% of tumors [[76]7, [77]8, [78]14, [79]15]. According to the largest analysis (n = 1045) conducted across several retrospective sequencing datasets [[80]8], these mutations predominantly occur in NOTCH1, although mutations in NOTCH2, NOTCH3, and NOTCH4 have also been identified [[81]2, [82]5, [83]7, [84]8]. A retrospective analysis of three independent cohorts demonstrated that ACCs with NOTCH1 activating mutations are associated with a poorer median overall survival and progress four times faster than those patients without activating mutations [[85]8, [86]15, [87]16]. The first retrospective study of 102 ACCs revealed that tumors with NOTCH1 mutations are associated with solid histology, advanced-stage disease at presentation, liver and bone metastases, and shorter relapse-free (12.5 vs 33.9 months) and median overall survival (29.6 vs 121.9 months) compared to tumors with wild-type (WT) NOTCH1 [[88]15]. The second retrospective study in 84 patients with recurrent/metastatic ACC found that median overall survival was significantly shorter in those with NOTCH1-mutated tumors compared to NOTCH1 WT malignancies (55.1 vs 204.5 months) [[89]8]. Furthermore, among NOTCH1 mutant tumors, activating mutations were associated with significantly poorer survival (31.1 vs 73.8 months) [[90]8]. These findings were recently supported in an independent cohort in which activating NOTCH1 mutations were again associated with poorer overall survival (48 vs 195.6 months) [[91]16]. Recurrent mutations in genes encoding proteins that regulate the activity of the NOTCH transcription complex, such as SPEN and FBXW7, have also been identified in ACC genomes, further implicating the NOTCH pathway in ACC tumorigenesis [[92]2, [93]7, [94]15]. Based on the proposed multifaceted pro-oncogenic roles of NOTCH signaling in cancer [[95]17–[96]20], several γ-secretase inhibitors (GSIs) have been developed [[97]15, [98]21–[99]26] and tested in preclinical studies [[100]27–[101]32] and Phase I/II trials in patients with advanced solid tumors, including ACC [[102]15, [103]33], either as a single agent or in combination with targeted therapeutics or chemotherapy [[104]33]. However, the overall response rates to NOTCH-targeted therapy have been suboptimal. Factors limiting success to date include use of GSIs with suboptimal pharmacokinetic (PK)/pharmacodynamic properties, dose-limiting toxicity, and the failure to use biomarkers of NOTCH activation as selection criteria for clinical trial entry [[105]33–[106]36]. Thus, the potential of NOTCH inhibitors such as GSIs has yet to be fully explored. AL101 is an investigational small molecule GSI that potently inhibits all four NOTCH paralogs and prevents the upregulation of NOTCH target genes [[107]22]. While a number of recent preclinical studies have shown robust AL101 antitumor activity in several in vivo cancer models [[108]22, [109]37], a comprehensive evaluation of its effect in ACC is lacking, in part due to the scarcity of experimental model systems for ACC. Here we demonstrate that AL101 monotherapy inhibits the proliferation of NOTCH-activated ACC organoids at nanomolar concentrations. Furthermore, AL101 is well tolerated in vivo and has significant antitumor effects in ACC patient-derived xenograft (PDX) models with NOTCH1 activating mutations, but not in PDX models with WT NOTCH genes. Taken together, our results provide a strong foundation for the clinical development of AL101 as a targeted monotherapy in patients with NOTCH-activated ACC. Methods Cell lines and reagents The human ACC cell line HACC2A was received from Dr Jacques Nȍr (University of Michigan), the UFH2 cell line was received from Dr Frederic Kaye (University of Florida), and the ACC52 cell line was received from Dr Lurdes Quiemado (University of Oklahoma). Cells were monitored for mycoplasma using the MycoDetect kit (Greiner Bio-One). HACC2A cells were cultured in DMEM medium (Gibco) supplemented with 10% FBS, 200 mM L-glutamine, antibiotic-antimycotic (100X) (Gibco), 400 ng/ml hydrocortisone, 20 ng/ml epidermal growth factor, 5 µg/ml insulin and bovine brain extract (Lonza). UFH2 cells were cultured in DMEM+GlutaMAX medium (Gibco) supplemented with 10% FBS and 5000 U/ml penicillin-streptomycin (Gibco 15070063). ACC52 cells were cultured in RPMI 1640 medium (Corning) supplemented with 10% FBS, antibiotic-antimycotic (100X), 20 ng/ml epidermal growth factor, 400 ng/ml hydrocortisone, and 5 μg/ml insulin. AL101 (batch: 3H66027) was obtained from Ayala Pharmaceuticals. All other chemicals used in this study were purchased from Sigma and prepared according to the manufacturer’s recommendations. Organoid preparation Freshly obtained surgical samples were digested first with a mixture of collagenase and dispase, followed by TrypLE (ThermoFisher). After passing through a mesh, cells were embedded in Matrigel (Corning) and cultured in four different organoid media formulations in parallel. These formulations have the same base media containing the growth factors EGF, Noggin (Sigma), R-spondin (Sigma), and FGF10 (ThermoFisher) as well as N-acetylcysteine (ThermoFisher), nicotinamide, Y-27632 (Rho kinase inhibitor), A83-01 (TGF-β signaling inhibitor), N2, and B27 (all from Sigma). Additional components of these media include forskolin (adenylyl cyclase activator), CHIR99021 (Wnt activator), gastrin I, prostaglandin E2, FGF2, hydrocortisone, and heregulin β1 (all from Sigma). The cells that demonstrated the most robust growth were further expanded, and fractions of cells were harvested for histology, DNA and RNA isolation, or cryopreserved at low passage numbers. IHC staining Sections (4-micron) prepared from FFPE PDX tumors were stained for NOTCH1 intracellular domain (NICD1) (Cell Signaling, 4147S, 1:200), MYC (Abcam, clone Y69, catalog ab32072; 0.56 μg/ml), or Ki67 (BioCare, clone SP6, catalog CRM326, 1:100) using a Bond III automated immunostainer (Leica). Staining was carried out in the Dana Farber/Harvard Cancer Center Specialized Histopathology Core Laboratory, which is certified by the College of American Pathologists and meets Clinical Laboratory Improvement Amendments standards. Staining was developed using the Bond Polymer Refine Detection Kit (Leica). Slides were counterstained with hematoxylin and reviewed by a board-certified anatomic pathologist (JCA). Cell viability assays For cell lines, relative viability was determined using an Alamar Blue assay as outlined by the manufacturer (AbDSerotec). New media containing 1/10 volume of Alamar Blue reagent was added to the wells and cells were incubated at 37 °C for 1 h. Fluorescence (560 nm excitation, 590 nm emission wavelengths) was measured using a SpectraMax-Plus384 fluorometer (Sunnyvale). Percent viability was determined by comparing DMSO treatment to inhibitor treatment. For organoids, ATP measurements (CellTiter-Glo Luminescent Cell Viability Assays, Promega) were used to assess proliferation and viability. Medium was discarded and CellTiter-Glo 3D reagent was added to each well. After incubating 30 min at room temperature on a rotary shaker, bioluminescence activity was assessed using a plate luminometer. Reverse transcription and real-time PCR RNA was reverse transcribed to cDNA using Superscript III (Invitrogen) and then used as a template for real-time PCR. Gene amplification was carried out on a StepOnePlus system (Applied Biosystems) using TaqMan Gene Expression Assays (Applied Biosystems). Assay IDs were: ACTB-Hs01060665_g1, HEY1-Hs00232618_m1. HEY2-Hs01012057_m1, NRARP-Hs04183811_s1, MYC-Hs00153408_m1, and HES5-Hs01387463_g1. All reactions were performed in triplicate and relative RNA quantity was calculated after normalizing to ACTB expression by the 2^−ΔΔCT method. Xenograft models Early passage PDX tissues were obtained through the Adenoid Cystic Carcinoma Research Foundation. All animal procedures were performed at XenoSTART (South Texas Accelerated Research Therapeutics, San Antonio) following Institutional Animal Care and Use Committee protocols. Fragments of tumor (~70 mm^3) were implanted subcutaneously into the flanks of 6–12-week-old female nu/nu athymic nude mice (The Jackson Laboratories or Charles River Laboratories). Upon reaching 150–300 mm^3 tumor volume, mice were randomized to either treatment (n = 5) or vehicle (n = 5–10) groups using blinded block randomization and therapeutic dosing was implemented. Tumor dimensions were measured using digital calipers blinded to the treatment group and tumor volume was calculated using the formula: TV = width^2 × length × 0.52. Tumors were harvested at termination, weighted, and used for histology, immunohistochemical staining, and RNA sequencing (RNA-seq) analysis (n = 3–5 per arm). Percent mean tumor growth inhibition (%TGI) induced by AL101 was calculated relative to the untreated control group using the formula: %TGI = 1 – (AL101 final – AL101 baseline) / (Control final – Control baseline). NOTCH luciferase reporter assay Full-length (FL) human NOTCH1 (FL-NOTCH1), ΔECD-NOTCH1, ACCx9-I1680N, and ACCx11-S1723ins28 transcripts were synthesized by GenScript and ligated into the pcDNA3.1 (+FLAG) expression vector. Constructs were transiently transfected into U2OS cells using Lipofectamine 2000 reagent (Invitrogen, #11668019) and assessed for their ability to activate a NOTCH-sensitive luciferase reporter gene [[110]38, [111]39]. U2OS cells were used because of their transfectability and low basal NOTCH activity. Briefly, cells were co-transfected (in five biological replicates) with 10 ng of pcDNA3.1 expression construct, a NOTCH-sensitive firefly luciferase reporter gene (TP1-CSLx12-FF), and an internal control Renilla luciferase plasmid (pRL-TK, Promega). Normalized firefly luciferase activities were measured in whole cell extracts prepared 48 h after transfection using the Dual-Luciferase kit (Promega #E1960) and an Infinite M200 luminometer (Tecan). RNA sequencing RNA-seq was performed on an Illumina NovaSeq-6000 instrument. Adapters were trimmed with Cutadapt [[112]40]. For PDXs, mouse reads were filtered out using an approach described by Callari et al. [[113]41]. Human reads were aligned to human reference genome GRCh37/hg19 using STAR and STAR-fusion [[114]42, [115]43]. Gene expression levels were calculated using featureCounts [[116]44] and gene expression levels were normalized using DESeq2 [[117]45]. Differentially expressed (DE) genes were detected using DESeq2 according to the following parameters: (i) average gene expression >50 normalized reads, (ii) log2(fold-change) >1 or log2(fold-change) <−1, (iii) false discovery rate (FDR) < 0.05. Pathway enrichment analysis Pathway enrichment analysis was performed using minimal hyper-geometric statistics [[118]46], based on an approach similar to that described by Eden et al. [[119]47]. MSigDB c2 (curated gene sets: Kyoto Encyclopedia of Genes and Genomes (KEGG)/Reactome pathways) [[120]48] were used as pathway references. Multiple hypothesis correction of the p values was