Abstract
Introduction.
Uterine leiomyosarcoma (uLMS) is a rare, highly aggressive malignancy. Recent data suggest 50% of uLMS may harbor alterations in the ATRX gene and such mutations may confer sensitivity to ataxia-telangiectasia-and-Rad3-related (ATR) kinase inhibitors. We sought to investigate the in vivo activity of Elimusertib (BAY1895344), a novel ATR-inhibitor, against ATRX-mutated uLMS patient-derived xenografts (PDXs).
Methods.
Two fully characterized uLMS (i.e., LEY-11 and LEY-16) were grafted into female CB-17/SCID mice. Treatments with control vehicle or BAY1895344 (20 mg/kg dosed twice daily 3 days on 4 days off) were given via oral gavage and tumor measurements as well as weights obtained twice weekly. Tumor volume differences were calculated with a two-way ANOVA. Mechanistic studies were performed ex vivo using BAY1895344 treated uLMS tumor samples by western blot analysis.
Results.
Both PDX LEY-11 and PDX LEY-16 harboring ATRX gene mutations demonstrated an aggressive behavior in vivo (i.e., control mice were euthanized on average at day 12.5 for PDX LEY-11 and at day 33 for PDX LEY-16). In both tumor models BAY1895344 20mg/kg dosed with an intermittent oral schedule was able to induce significant growth inhibition compared to vehicle control treatment (p<0.001 for both LEY-11 and LEY-16) and prolong median overall survival [PDX LEY-11 (12.5 vs. 42 days, p <0.001) and PDX LEY-16 (33 vs. 60 days, p < 0.001)]. There were not significant changes in weight between treatment and controls. By western blot assays BAY1895344 exposure decreased phosphorylated-ATR and increased expression of apoptotic molecules in LMS PDXs.
Conclusions.
BAY1895344 demonstrates promising in vivo activity against biologically aggressive PDX models of uLMS harboring ATRX mutations, with no significant toxicity. Clinical trials of BAY1895344 in uLMS patients are warranted.
Keywords: BAY1895344, ATR inhibitors, uterine leiomyosarcoma, ATRX, DAXX
Introduction
Uterine leiomyosarcoma (uLMS) is a rare but highly aggressive gynecologic malignancy that has an estimated 5-year overall survival of 50% for even early-stage disease. Though they are rare and account for only 3–7% of uterine malignancies, uLMS is the most common gynecologic sarcoma, accounting for 80% of cases [1]. Management consists of extirpative surgery followed by observation for early-stage disease (stage I-II) and adjuvant therapy for advanced-stage disease [1]. However, given that uLMS is a rare disease, evidence guiding recommendations for systemic therapy for recurrent or metastatic disease is poor., Several prospective studies demonstrate doxorubicin, gemcitabine, ifosfamide, and docetaxel to be active in advanced-stage disease, but in widespread metastatic disease outcomes are universally poor [2–5].
Given the lack of effective adjuvant therapies and poor overall survival, recent studies have examined the genetic landscape of uLMS to determine if there are common and potentially actionable genetic mutations. However, these studies had samples sizes of <30 and/or included leiomyosarcomas from extra-gynecologic anatomical sites [6–8]. In 2021, our research group published the largest genomic analysis to date of uLMS [9]. Using whole-genome-sequencing (WGS), whole exome sequencing (WES) and RNA-Sequencing (RNA-Seq), 56 uLMS along with 27 matched uLMS samples obtained through The Cancer Genome Atlas (TCGA) were evaluated and results demonstrated frequently mutated genes including TP53, ATRX, PTEN, and MEN1 genes [9]. Somatic Copy Number Variant (CNV) analysis revealed evidence of C-MYC copy-number gains and homologous recombination deficiency (HRD) signatures as well in a subset of uLMS. Lastly, in vivo monotherapy with olaparib, GS-626510, and copanlisibdemonstrated tumor growth inhibition and prolonged survival in SCID mice implanted with uLMS xenografts [9].
The ATRX gene is associated with a specific route of telomere lengthening in cancer cells, termed alternative lengthening of telomeres (ALT), which allows 10–15% of human tumors to achieve “immortalization” and unlimited tumor division through this mechanism [10]. Tumors of mesenchymal origin (such as uLMS) have been found to more commonly utilize ALT; inactivating mutations of ATRX and its associated death-domain associated protein (DAXX) are more common in tumor cells that utilize ALT [10,11]. The proteins encoded by the ATRX and DAXX genes facilitate binding of the histone variant H3.3 into telomeric chromatin and stalling of cell division in the presence of replicative stress, thus functioning as a tumor suppressor [10]. A similar and related protein, ataxia telangiectasia and Rad3-related protein (ATR), is a key component of DNA Damage Repair (DDR) for replicative stress, though not a tumor-suppressor [11]. ATR, along with ataxia telangiectasia mutated (ATM) and DNA dependent protein kinase (DNA-PK) form the cell’s main response to DDR; ATM and DDK are primarily involved in double-strand break repair and, as aforementioned, ATR is involved in repair of replicative stress [10–12]. As such, ATRX/DAXX and ATM mutations may lead to increased susceptibility/dependency to ATR for a cancer cell’s rescue from mitotic catastrophe, making it an appealing target for inhibition in the fight against cancer. Consistent with this view, ATRX mutations have been found to confer increased sensitivity to ATRi [13]. Currently, there are several ATR inhibitors (ATRi) in Phase I-II trials that have demonstrated promise, including BAY1895344, a selective inhibitor of ATR kinase activity [12]. In preclinical models of various human cancers with varying DDR mutations (i.e. ATM, ATR, ATRX, BRIP1, MLH1, etc.) BAY1895344 demonstrated significant activity both as monotherapy and in combination with varying agents including olaparib and rucaparib; however no sarcomas were studied [12]. Given that the prevalence of ATRX mutations in leiomyosarcoma is estimated to be over 30% [14] and the potential of ATRX as a novel therapeutic target, we sought to determine the in vivo activity of BAY1895344 against two recently established and fully sequenced ATRX-mutated uLMS PDXs.
Materials and Methods
2.1. Patient and Specimen Acquisition
The study protocol was approved by the Yale Human Investigation Committee and was conducted in accordance with the Declaration of Helsinki. DNA and RNA was extracted from two PDXs, which were established from two patients after informed consent from the participant and/or their legally authorized representative was obtained prior to initiating any research activities. Both patients underwent staging at Yale New Haven Hospital. Benign myometrial tissue was similarly collected from patients after informed consent from the participant and/or their legally authorized representative was obtained.
2.2. BAY1895344
BAY1895344 was obtained from Bayer AG and prepared as previously described [12]. Briefly, it was prepared into a stock weekly with a vehicle of PEG 400, water, and 100% ethanol and kept at 4°C shielded from light. Based on data published on the bioavailability of the compound in preclinical models, dosages of 10mg/kg twice daily and 20mg/kg twice daily were chosen for evaluation [12].
2.3. Whole-exome DNA sequencing and CNV analysis
Whole-exome sequencing and CNV analysis on uLMS tumors were undertaken as previously described [9].
2.4. Real-time (RT) reverse transcription PCR
RNA isolation was performed on two uLMS samples (LEY-11 and LEY-16) and 11 randomly selected uterine myometrium control fresh-frozen samples was performed using AllPrep DNA/RNA/Protein Mini Kit (Qiagen), according to the manufacturer’s instructions. Quantitative PCR was carried out with a 7500 Real-Time PCR System using the manufacturer’s recommended protocol (Applied Biosystems) to evaluate the expression of ATRX, ATR, and DAXX. The primers and probes were obtained from Applied Biosystems (i.e., ATRX, Assay ID: Hs00997529_m1; ATR Assay ID: Hs00992123_m1; DAXX Assay ID: Hs00154692_m1). The comparative threshold cycle method was used to determine gene expression in each sample, relative to the value of glyceraldehyde-3-phosphate dehydrogenase (assay ID Hs99999905_m1) RNA as an internal control.
2.5. Leiomyosarcoma PDX model and Treatment
Fresh frozen samples of the two fully characterized uLMS (LEY-11 and LEY-16) were xenografted into female CB17/lcrHsd-Prkdc/SCID mice into the subcutaneous tissue of the lower abdomen. Mice were triaged into treatment groups when the tumor was established at ≥ 0.2 cm3, which included a control group,BAY1895344 10mg/kg, and BAY1895344 20mg/kg. Animals were treated with BAY1895344 or normal saline via oral gavage twice daily for 3 days on/4 days off. Lower abdominal tumors were measured in length and width using calipers twice weekly. Animals were humanely euthanized when tumor volume reached 1.0 cm3 using the formula length × (width)2/2. Once all control group animals were euthanized, treatment group animals were followed for survival. Toxicity was assessed via mouse weight, which was recorded twice weekly; treatment was held if an animal’s weight dropped below 10% of their starting weight. All mice were housed and treated in accordance with the policies set forth by the Institutional Animal Care and Use Committee (IACUC) at Yale University.
2.6. Ex vivo tumor tissue culture and treatment
Tumor tissues were dissected from mice and cut into 3×3×3mm pieces. Tumor pieces were then incubated in RPMI 1640 medium (Life Technologies, Grand Island, NY) containing 10% fetal bovine serum (Seradigm, Radnor, PA) and penicillin/streptomycin (Life Technologies) at 37°C and 5% CO2. After 1 hour of incubation, tumor pieces were treated with medium containing BAY1895344 (0.3 μM or 3 μM) or the DMSO solvent (0.03%) as untreated control. At two hours, tumor pieces were homogenized and used for preparing protein lysates.
2.7. Western Blot
Tumor tissues were homogenized using a BeadBug Homogenizer (Benchmark Scientific, Sayreville, NJ). Protein lysates were prepared with lysis buffer (1% Triton X-100, 0.05% SDS, 100 mM Na2HPO4, and 150 mM NaCl). Protein lysate was electrophoresed on a 4–20% pre-cast SDS-polyacrylamide gel (Bio-Rad, Hercules, CA) and transferred onto Amersham Hybond 0.45 PVDF membranes (GE Healthcare, Chicago, IL). After blocking with 5% non-fat milk in PBS-0.05% Tween 20, the membranes were incubated with primary antibodies at 4°C overnight, and then secondary antibodies for 1 h at room temperature. Antibodies include anti-p-ATR antibody (#2853, Cell Signaling Technology), anti-ATR antibody (#13934, Cell Signaling Technology), p-CHK1 antibody (#2348, Cell Signaling Technology, Danvers, MA), cleaved-caspase-3 antibody (#9664, Cell Signaling Technology), HRP-conjugated anti-β-actin antibody (#HRP-6008, ProteinTech), anti-rabbit secondary antibody (#7074, Cell Signaling Technology). The blots were developed using Clarity or Clarity Max Western ECL Blotting Substrates (Bio-Rad).
2.8. Statistical Analysis
Statistical analysis was performed using GraphPad Prism version 7 (GraphPad Software, Inc. San Diego, CA). ANOVA was used to evaluate significant differences in tumor volumes at specific time points for the in vivo experiments. Overall survival (OS) data (OS defined as time of enrollment to either death or tumor volume of 1.0 cm3) were analyzed and plotted using Kaplan-Meier survival curves, which were compared using the log-rank test. Differences in all comparisons were considered statistically significant at p-values <0.05.
Results
LEY-11 and LEY-16 genetic landscape
Analyses of the genetic characteristics of the two uLMS prior to treatment or implantation as PDX (LEY-11 and LEY-16), including somatic single nucleotide variant (SNV) and CNV mutations, were highly consistent with the genetic landscape results recently reported by our group using a large set of fresh and formalin fixed uLMS (ie, 83 samples) [10]. As depicted in Table 1.A, we found SNV alterations in multiple driver genes including TP53, PTEN, ATRX, RB1 and KTM2B as well as CNV in C-MYC and MAP2K4 in LEY-11. Similarly, LEY-16 demonstrated SNV alterations in driver genes including TP53, SMARCA4 and NAV3, CNV in PTEN, MAP2K4 and ATRX genes, as well as multiple fusions/translocations (Table 1.B).
Table 1.
Genetic landscape of sequenced tumors. A) Genetic landscape of LEY-11. B) Genetic landscape of LEY-16.
| Table 1A. LEY-11 Genetic Landscape | ||||||||||||||
| i. Somatic SNVs in Cancer190 genes or HR48 gene | ||||||||||||||
| Chr | Pos | Ref | Alt | Gene | Type | AA | dbSNP | ExAC | MetaSVM | CADD | Ref:Alt | |||
| 17 | 7574003 | G | A | TP53 | stopgain | p.R303X | . | . | . | 37.0 | 24,302 | |||
| 10 | 89711950 | C | - | PTEN | frameshift deletion | p.P363Qfs*9 | . | . | . | . | 9,84 | |||
| X | 76855019 | T | - | ATRX | frameshift deletion | p.D1940lfs*15 | . | . | . | . | 99,57 | |||
| 13 | 49039469 | T | - | RB1 | frameshift deletion | p.L819Cfs*7 | . | . | . | . | 30,232 | |||
| 19 | 36220087 | C | T | KMT2B | stopgain | p.Q1603X | . | . | . | 44.0 | 11,96 | |||
| ii. Somatic CNVs covering Cancer190 or HR48 genes | ||||||||||||||
| Chr | Start | End | CNV type | Copy number | #Genes | Cancer190 genes | ||||||||
| 1 | 149901457 | 166839177 | Duplication | 3 | 365 | B4GALT3 (S100A7, S100A8, S100A9) | ||||||||
| 4 | 1219049 | 8952176 | Duplication | 4 | 70 | CRIPAK, FGFR3 | ||||||||
| 7 | 23730942 | 35013303 | Duplication | 3 | 64 | NFE2L3 | ||||||||
| 8 | 48114004 | 146279624 | Duplication | 3 | 402 | EPPK1, RAD21, SOX17 (MYC, NBN, RAD54B) | ||||||||
| 10 | 5919996 | 38407815 | Deletion | 1 | 130 | GATA3 | ||||||||
| 11 | 100665721 | 109295003 | Duplication | 3 | 51 | ATM | ||||||||
| 16 | 97334 | 33965671 | Duplication | 3 | 434 | AXIN1, CREBBP, SOCS1, TRAF7 (PALB2) | ||||||||
| 16 | 47117063 | 52060975 | Duplication | 3 | 22 | CYLD | ||||||||
| 16 | 68771193 | 76592630 | Duplication | 3 | 76 | CDH1 | ||||||||
| 17 | 909249 | 10233910 | Duplication | 3 | 213 | TP53 (RPA1) | ||||||||
| 17 | 10608193 | 16137459 | Duplication | 3 | 28 | MAP2K4, NCOR1 | ||||||||
| 18 | 18531196 | 78005326 | Duplication | 3 | 198 | BCL2, SETBP1, SMAD2, SMAD4 (RBBP8) | ||||||||
| 20 | 68253 | 26189020 | Duplication | 3 | 176 | FOXA2 | ||||||||
| 20 | 35788394 | 62905003 | Duplication | 3 | 253 | TSHZ2 | ||||||||
| 21 | 14982479 | 45514442 | Duplication | 3 | 162 | RUNX1, U2AF1 | ||||||||
| X | 5827024 | 57936904 | Duplication | 3 | 295 | BCOR, GATA1, KDM5C, KDM6A, SMC1A, USP9X | ||||||||
| X | 85631746 | 155240124 | Deletion | 1 | 365 | PHF6, STAG2 (BRCC3, RPA4) | ||||||||
| 11 | 89402478 | 96125438 | Duplication | 3 | 41 | (MRE11A) | ||||||||
| 17 | 16593655 | 18291667 | Duplication | 3 | 27 | (TOP3A) | ||||||||
|
iii. Fusions/Translocations in Kinase518/Cancer190/Cosmic567/HR48 genes None found | ||||||||||||||
| Table 1B. LEY-16 Genetic Landscape | ||||||||||||||
| i. Somatic SNVs in Cancer190 genes or HR48 gene | ||||||||||||||
| Chr | Pos | Ref | Alt | Gene | Type | AA | dbSNP | ExAC | MetaSVM | CADD | Ref:Alt | |||
| 17 | 7578406 | C | T | TP53 | Nonsynonymo us SNV | p.R136H | 8.24E-06 | 8.24E-06 | D | 31.0 | 16,46 | |||
| 19 | 11101886 | G | T | SMARCA4 | Nonsynonymo us SNV | p.A436S | . | . | T | 15.4 | 70,41 | |||
| 12 | 78443857 | T | C | NAV3 | Nonsynonymo us SNV | p.L703P | . | . | T | 25.4 | 37,4 | |||
| ii. Somatic CNVs covering Cancer190 or HR48 genes | ||||||||||||||
| Chr | Start | End | CNV type | Copy number | # Genes | Cancer190 genes | ||||||||
| 1 | 156643133 | 164762038 | Duplication | 4 | 135 | B4GALT3 (S100A7, S100A8, S100A9) | ||||||||
| 1 | 145368425 | 147608093 | Duplication | 3 | 32 | (BCL9) | ||||||||
| 6 | 121544283 | 157100664 | Duplication | 3 | 154 | ARID1B, TNFAIP3 | ||||||||
| 9 | 107456607 | 113734510 | Duplication | 3 | 31 | KLF4 | ||||||||
| 10 | 88635664 | 93871025 | Deletion | 1 | 44 | PTEN | ||||||||
| 14 | 58598133 | 82000184 | Deletion | 1 | 178 | TSHR (RAD51B) | ||||||||
| 17 | 8296018 | 12921314 | Duplication | 3 | 33 | MAP2K4 | ||||||||
| X | 76845218 | 77131148 | Deletion | 1 | 2 | ATRX | ||||||||
| 15 | 40993171 | 41672467 | Duplication | 3 | 18 | RAD51 | ||||||||
| iii. Fusions/Translocations in Kinase518/Cancer190/Cosmic567/HR48 genes | ||||||||||||||
| Gene1 | Gene2 | Genelclass | Gene2class | Breakpoint1 | Breakpoint2 | Type | Splitreads1 | Splitreads2 | Discordantmates | Coverage1 | Coverage2 | Confidence | Reading-frame | Calledby |
| ZCCHC4 | TEC | NA | Kinase | 4:25363923 | 4:48152959 | Inversion | 2 | 2 | 1 | 47 | 36 | High | Inframe | Arriba, Starfusion |
| HEG1 | CAMTA1 | NA | Cosmic567 | 3:124724109 | 1:7796404 | Translocation | 0 | 1 | 3 | 247 | 15 | High | Inframe | Arriba |
| NUSAP1 | RAD51 | NA | HR | 15:41669502 | 15:40990955 | Duplication | 2 | 4 | 11 | 1495 | 232 | High | Inframe | Amba, Starfusion |
| POLD2 | EEPD1 | HR | NA | 7:44154631 | 7:36334034 | Deletion/5’-5’ | 0 | 2 | 1 | 1 | 1 | medium | - | Arriba |
GISTIC significant duplications or deletions
Low ATRX expression by RT-PCR in PDX LEY-11 and PDX LEY-16
As both LEY-11 and LEY-16 demonstrated alterations in the ATRX gene, we used real-time PCR to evaluate ATRX RNA expression as well as the expression of other genes of this pathway such as ATR and DAXX in the two available PDX samples, along with 11 fresh frozen myometrium control samples obtained from benign hysterectomy specimens.. As demonstrated in Table 2, similar to our previous report studying 37 fresh-frozen uLMS (which included both LEY-11 and LEY-16) [9], we found both PDXs to express lower levels of ATRX transcripts when compared to benign uterine myometrium, while no differences in expression were noted in ATR and DAXX genes (Table 2).
Table 2.
RT-PCR expression of ATRX, ATR and DAXX in PDX LEY-16 and PDX LEY-11 and controls (benign myometrium).
| Sample | ATR Ct | ATR ΔCt | ATRX Ct | ATRX ΔCt | DAXX Ct | DAXX ΔCt |
|---|---|---|---|---|---|---|
| LEY-11 | 29.18 | 12.41 | 26.89 | 10.091 | 25.77 | 8.97 |
| LEY-16 | 28.53 | 8.41 | 35.77 | 15.978 | 26.39 | 6.59 |
| Leiomyoma controls | 32.80 | 11.08 | 30.22 | 6.54 | 30.69 | 6.46 |
In vivo Treatment of ATRX-mutated uLMS PDXs in SCID mice with BAY1895344 Produces a Cytostatic Effect and Prolongs Overall Survival
We next evaluated the impact of BAY1895344 in vivo in xenograft models generated by subcutaneously grafting fresh-frozen LEY-11 and LEY-16 samples into female CB17/lcrHsd-Prkdc/SCID mice. In both models, PDX LEY-11 and PDX LEY-16, BAY1895344 dosed intermittently demonstrated a significant cytostatic effect with a smaller tumor volume (Figure 1.A, 2.1.A, and 2.2.A). In PDX LEY-16, this reached statistical significance on day 22 of treatment (p<0.001); in PDX LEY-11, two separate experiments were undertaken given the more pronounced effect in this model compared to PDX LEY-16 and to also test BAY1895344 at a lower dose (10 mg/kg) (Figure 2). At a dose of 20mg/kg given twice daily (BID), the tumor volume inhibition reached significance at day 11 (p<0.001) in the first experiment (Figure 2.1.A) and day 8 (p<0.001) in the second experiment. However, at a lower dose of 10mg/kg BID, tumor volume inhibition was not significantly different than control (p=0.54). Additionally, the tumor growth rate for PDX LEY-11 was significantly slower in the 20mg/kg treatment groups: 0.093 cm3/day vs. 0.028 cm3/day in the control group and 20 mg/kg group, respectively in experiment 1 (p=0.002) and 0.112 cm3/day vs. 0.017 cm3/day in experiment 2 (p=0.009). The tumor growth rate for PDX LEY-11 treated with 10mg/kg of drug was not significantly different than control in experiment 2 (p=0.94). Overall survival curves demonstrated a significant median OS difference between BAY1895344 20mg/kg BID and control in PDX LEY-16 (Figure 1.B) and between 20mg/kg BID and control in both PDX LEY-11 experiments (Figure 2.1.B and 2.2.B). For PDX LEY-16, median OS for control vs. 20mg/kg BID was 33 vs. 60 days (p=0.0013) (Figure 1.B). For PDX LEY-11, median OS for control vs. 20mg/kg BID in experiment 1 was 12.5 vs. 42 days (p<0.001) (Figure 2.1.B); In experiment 2, median OS for control vs. 10mg/kg vs. 20mg/kg BID was 8 vs. 11 vs. 42 days; the difference in survival between control and 10mg/kg was not statistically significant (p=0.0016 in control vs. 20mg/kg; p=0.06 in control vs. 10mg/kg) (Figure 2.2.B). Twice daily oral doses for three days weekly (10 mg/kg or 20 mg/kg) were well tolerated with no impact on body weight compared with vehicle (Figure 1.C, 2.1.C, and 2.2.C).
Figure 1.

Antitumor activity and overall survival in mice inoculated with uLMS xenograft tumor model (PDX LEY-16) after treatment with BAY1895344 (20mg/kg) compared to vehicle control. A) BAY1895344 (20mg/kg) demonstrates significant tumor growth inhibition compared to vehicle control (p<0.001). B) Overall survival was significantly prolonged among mice treated with BAY1895344 (20mg/kg) (p=0.0013). C) Xenograft mice weight changes by treatment group did not differ.
Figure 2.

Antitumor activity and overall survival in mice inoculated with uLMS xenograft tumor model (PDX LEY-11) after treatment with BAY1895344 (20mg/kg or 10mg/kg) compared to vehicle control over two experiments. Experiment 1) Vehicle control vs. BAY1895344 (20mg/kg) 2.1.A) BAY1895344 (20mg/kg) demonstrates significant tumor growth inhibition compared to vehicle control (p<0.001) 2.1.B) Overall survival was significantly prolonged among mice treated with BAY1895344 (20mg/kg) (p<0.001). 2.1.C) Xenograft mice weight changes by treatment group. Experiment 2) Vehicle control vs. BAY1895344 (20mg/kg) vs. BAY1895344 (10mg/kg) 2.2.A) BAY1895344 (20mg/kg) demonstrated significant tumor growth inhibition compared to vehicle control (p<0.001); 10mg/kg dosage did not have significant inhibition compared to control (p=0.54) 2.2.B) Overall survival was significantly prolonged among mice treated with BAY1895344 (20mg/kg) (p=0.0016); Mice treated with BAY1985344 (10mg/kg) did not have significantly prolonged overall survival (p=0.06). 2.2.C) Xenograft mice weight changes by treatment group did not differ.
PDX LEY-11 BAY1895344 exposure Reduces Phosphorylated-ATR and increases cleaved caspase-3
As representatively demonstrated in Figure 3, in ex-vivo culture model of PDX LEY-11 tumors, treatment with ATR inhibitor BAY1895344 for 2 hours significantly reduced the level of phosphorylated ATR. The checkpoint kinase CHK1 undergoes ATR-mediated phosphorylation and activation. Accordingly, the inhibition of ATR by BAY1895344 treatment also led to the reduction of phosphorylated CHK1. As a marker of cell death induction, the activated caspase-3 (17 or 19 kDa) resulting from cleavage adjacent to Asp175 was progressively detected in the BAY1895344-treated tumors demonstrating increased tumor cell apoptosis.
Figure 3.

Representative western blot of ex-vivo tumor culture model of PDX LEY-11 treated with BAY1895344 for 2 hours. Treatment with BAY1895344 significantly reduced the level of phosphorylated ATR and phosphorylated CHK1; Levels of cleaved-caspase3 increase with prolonged treatment demonstrating increased cell apoptosis.
Discussion
There are several major challenges in the treatment of uLMS, including its relative resistance to traditional cytotoxic therapies, its rarity, and the relative lack of available targeted therapeutics. In our in vivo studies, BAY1895344 demonstrated significant cytostatic effect in both available PDX models. The aggressive biologic nature of these tumor models is highlighted by the low median overall survival in the vehicle group: 8–12.5 days in PDX LEY-11 and 33 days in PDX LEY-16. Additionally, the treatment was well tolerated with no differences in body weight between control-group and treatment-group mice. This is mirrored in a recent Phase I trial of BAY1895344, wherein the majority of dose-limiting toxicities were hematologic, which recovered with treatment holds [15]. Experiments were performed at two dosages (20mg/kg and 10mg/kg BID, 3 days on 4 off schedule): only the higher dosage was efficacious for tumor size reduction and survival benefit.
A recent publication from our laboratory with the largest WGS, WES, and RNA-Seq data to date demonstrated that 51% of samples harbored ATRX gene alterations, which result in decreased ATRX RNA expression when compared to wild type ATRX uLMS [9]. In this comprehensive genetic evaluation, we also found that ATRX gene derangement was significantly associated with decreased survival in uLMS patients [9]. Given the link between ATRX mutations and sensitivity to ATR inhibitors, we sought to investigate this novel targeted treatment in uLMS where an unmet clinical need exists [15].
ATR and its cascade demonstrate a promising target for novel clinical therapeutics. Preclinical data demonstrate many uLMS carry these alterations as well as additional alterations in DAXX and ATM genes, increasing the number of patients potentially responsive to novel treatment modalities targeting ALT. The recent Phase I trial and first-in-human trial of BAY 1895344 in patients with advanced solid tumors confirmed that the drug had promising antitumor activity, especially among those with ATM gene mutations [15]. In this regard, recent work from our lab demonstrated that tumors harboring ATRX alterations express lower levels of gene expression, which is associated with the aforementioned ALT pathway activation. It is worth noting that patients with ATRX/DAXX mutations were characterized by a significantly shorter survival, supporting the view that alterations in this DNA repair pathway trigger a more aggressive uLMS phenotype [9]. These clinical findings are further supported by our current preclinical study as demonstrated by the rapid in vivo growth of both uLMS PDX models harboring ATRX alterations in SCID mice. More importantly, these data suggest that novel ATR inhibitors currently in clinical trials such as BAY1895344, may represent new, potentially effective treatment options for the large subset of patients with advanced/recurrent uLMS carrying ATRX/DAXX mutations [9,16,17] Importantly, detection of ATRX and DAXX alterations is now practical with commercially available genomic profiling tests that sample tumoral specimens, making detection of these prevalent alterations more feasible [18]. Additionally, recent studies demonstrate that SMARCA4/BRG1 mutations can also confer susceptibility to ATR inhibition [19–21]; it is notable that LEY-16 was found to have alternations in SMARCA4 driver gene as well as ATRX.
.. Mechanistically, in vivo RT-PCR of pre-treatment PDX tissue demonstrated lower expression of ATRX gene transcripts in the PDX LEY-11 and PDX LEY-16 models, as expected given their known ATRX mutations. Moreover, western blotting assays performed ex vivo after exposure of uLMS to BAY1895344 demonstrated dephosphorylation of both ATR as well as CHK1, with a progressive increase in the induction of markers of apoptosis (i.e., caspase-3 proteins), compared to controls. Taken together these results demonstrate the impressive activity of BAY1895344 in inhibiting ATR activity and inducing apoptosis in uLMS.
This paper has a few important weaknesses. Without an appropriate in vitro model, we were not able to confirm effectivity of BAY1895344 on uLMS tumor cell lines outside of our PDX experiments. Additionally, we were not able to evaluate hematologic toxicity or other end organ toxicity directly in mice, only weight-based toxicity. Given that drug studies in humans are underway, more relevant toxicity of BAY1895344 is being evaluated [15].
In conclusion, we report for the first time the remarkable in vivo preclinical anti-tumor activity and the acceptable toxicity profile of BAY1895344 (Elimusertib) against biologically aggressive ATRX-mutated uterine leiomyosarcoma. Given that upwards of 51% of uLMS may express ATRX alterations, and an additional 19% may harbor DAXX gene mutations [10], ALT might represent a common feature of uLMS. Accordingly, we recommend all patients be offered genomic tumor profiling and that women with uLMS be included in early phase trials of ATR inhibitors in solid organ malignancies [22,23].
Elimusertib (BAY1895344) is a novel ATR kinase inhibitor effective against uterine leiomyosarcoma
Elimusertib decreased tumor growth in uterine leiomyosarcoma PDX mouse models with ATRX mutations
Elimusertib prolonged overall survival in PDX mouse models of uterine leiomyosarcoma
Tumor cells exposed to Elimusertib showed decreased phosphorylated-ATR and increased apoptotic molecules on western blot
Financial support:
This work was supported in part by grants from NIH U01 CA176067-01A1, the Deborah Bunn Alley, the Domenic Cicchetti, the Discovery to Cure Foundations and the Guido Berlucchi Foundations to AS. This investigation was also supported by NIH Research Grant CA-16359 from NCI and Standup-to-cancer (SU2C) convergence grant 2.0 to AS.
Footnotes
Conflict of Interest Statement
A.D.S. reports grants from PUMA, grants from IMMUNOMEDICS, grants from GILEAD, grants from SYNTHON, grants and personal fees from MERCK, grants from BOEHINGER-INGELHEIM, grants from GENENTECH, grants and personal fees from TESARO and grants and personal fees from EISAI. The other authors declare no conflict of interest.
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
References
- [1].Amant F, Coosemans A, Debiec-Rychter M, Timmerman D, Vergote I, Clinical management of uterine sarcomas, The Lancet Oncology. 10 (2009) 1188–1198. 10.1016/S1470-2045(09)70226-8. [DOI] [PubMed] [Google Scholar]
- [2].Hensley ML, Blessing JA, Degeest K, Abulafia O, Rose PG, Homesley HD, Fixed-dose rate gemcitabine plus docetaxel as second-line therapy for metastatic uterine leiomyosarcoma: a Gynecologic Oncology Group phase II study, Gynecol Oncol. 109 (2008) 323–328. 10.1016/j.ygyno.2008.02.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [3].Hensley ML, Maki R, Venkatraman E, Geller G, Lovegren M, Aghajanian C, Sabbatini P, Tong W, Barakat R, Spriggs DR, Gemcitabine and docetaxel in patients with unresectable leiomyosarcoma: results of a phase II trial, J Clin Oncol. 20 (2002) 2824–2831. 10.1200/JCO.2002.11.050. [DOI] [PubMed] [Google Scholar]
- [4].Look KY, Sandler A, Blessing JA, Lucci JA, Rose PG, Gynecologic Oncology Group (GOG) Study, Phase II trial of gemcitabine as second-line chemotherapy of uterine leiomyosarcoma: a Gynecologic Oncology Group (GOG) Study, Gynecol Oncol. 92 (2004) 644–647. 10.1016/j.ygyno.2003.11.023. [DOI] [PubMed] [Google Scholar]
- [5].Omura GA, Blessing JA, Major F, Lifshitz S, Ehrlich CE, Mangan C, Beecham J, Park R, Silverberg S, A randomized clinical trial of adjuvant adriamycin in uterine sarcomas: a Gynecologic Oncology Group Study, J Clin Oncol. 3 (1985) 1240–1245. 10.1200/JCO.1985.3.9.1240. [DOI] [PubMed] [Google Scholar]
- [6].Cuppens T, Moisse M, Depreeuw J, Annibali D, Colas E, Gil-Moreno A, Huvila J, Carpén O, Zikán M, Matias-Guiu X, Moerman P, Croce S, Lambrechts D, Amant F, Integrated genome analysis of uterine leiomyosarcoma to identify novel driver genes and targetable pathways, Int J Cancer. 142 (2018) 1230–1243. 10.1002/ijc.31129. [DOI] [PubMed] [Google Scholar]
- [7].Abeshouse A, Adebamowo C, Adebamowo SN, Akbani R, Akeredolu T, Ally A, Anderson ML, Anur P, Appelbaum EL, Armenia J, Auman JT, Bailey MH, Baker L, Balasundaram M, Balu S, Barthel FP, Bartlett J, Baylin SB, Behera M, Belyaev D, Bennett J, Benz C, Beroukhim R, Birrer M, Bocklage T, Bodenheimer T, Boice L, Bootwalla MS, Bowen J, Bowlby R, Boyd J, Brohl AS, Brooks D, Byers L, Carlsen R, Castro P, Chen H-W, Cherniack AD, Chibon F, Chin L, Cho J, Chuah E, Chudamani S, Cibulskis C, Cooper LAD, Cope L, Cordes MG, Crain D, Curley E, Danilova L, Dao F, Davis IJ, Davis LE, Defreitas T, Delman K, Demchok JA, Demetri GD, Demicco EG, Dhalla N, Diao L, Ding L, DiSaia P, Dottino P, Doyle LA, Drill E, Dubina M, Eschbacher J, Fedosenko K, Felau I, Ferguson ML, Frazer S, Fronick CC, Fulidou V, Fulton LA, Fulton RS, Gabriel SB, Gao J, Gao Q, Gardner J, Gastier-Foster JM, Gay CM, Gehlenborg N, Gerken M, Getz G, Godwin AK, Godwin EM, Gordienko E, Grilley-Olson JE, Gutman DA, Gutmann DH, Hayes DN, Hegde AM, Heiman DI, Heins Z, Helsel C, Hepperla AJ, Higgins K, Hoadley KA, Hobensack S, Holt RA, Hoon DB, Hornick JL, Hoyle AP, Hu X, Huang M, Hutter CM, Iacocca M, Ingram DR, Ittmann M, Iype L, Jefferys SR, Jones KB, Jones CD, Jones SJM, Kalir T, Karlan BY, Karseladze A, Kasaian K, Kim J, Kundra R, Kuo H, Ladanyi M, Lai PH, Laird PW, Larsson E, Lawrence MS, Lazar AJ, Lee S, Lee D, Lehmann K-V, Leraas KM, Lester J, Levine DA, Li I, Lichtenberg TM, Lin P, Liu J, Liu W, Liu EM, Lolla L, Lu Y, Ma Y, Madan R, Maglinte DT, Magliocco A, Maki RG, Mallery D, Manikhas G, Mardis ER, Mariamidze A, Marra MA, Martignetti JA, Martinez C, Mayo M, McLellan MD, Meier S, Meng S, Meyerson M, Mieczkowski PA, Miller CA, Mills GB, Moore RA, Morris S, Mose LE, Mozgovoy E, Mungall AJ, Mungall K, Nalisnik M, Naresh R, Newton Y, Noble MS, Novak JE, Ochoa A, Olvera N, Owonikoko TK, Paklina O, Parfitt J, Parker JS, Pastore A, Paulauskis J, Penny R, Pereira E, Perou CM, Perou AH, Pihl T, Pollock RE, Potapova O, Radenbaugh AJ, Ramalingam SS, Ramirez NC, Rathmell WK, Raut CP, Riedel RF, Reilly C, Reynolds SM, Roach J, Robertson AG, Roszik J, Rubin BP, Sadeghi S, Saksena G, Salner A, Sanchez-Vega F, Sander C, Schein JE, Schmidt HK, Schultz N, Schumacher SE, Sekhon H, Senbabaoglu Y, Setdikova G, Shelton C, Shelton T, Shen R, Shi Y, Shih J, Shmulevich I, Sica GL, Simons JV, Singer S, Sipahimalani P, Skelly T, Socci N, Sofia HJ, Soloway MG, Spellman P, Sun Q, Swanson P, Tam A, Tan D, Tarnuzzer R, Thiessen N, Thompson E, Thorne LB, Tong P, Torres KE, van de Rijn M, Van Den Berg DJ, Van Tine BA, Veluvolu U, Verhaak R, Voet D, Voronina O, Wan Y, Wang Z, Wang J, Weinstein JN, Weisenberger DJ, Wilkerson MD, Wilson RK, Wise L, Wong T, Wong W, Wrangle J, Wu Y, Wyczalkowski M, Yang L, Yau C, Yellapantula V, Zenklusen JC, Zhang J. (Julia), Zhang H, Zhang H, Zmuda E, Comprehensive and Integrated Genomic Characterization of Adult Soft Tissue Sarcomas, Cell. 171 (2017) 950–965.e28. 10.1016/j.cell.2017.10.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [8].Mäkinen N, Aavikko M, Heikkinen T, Taipale M, Taipale J, Koivisto-Korander R, Bützow R, Vahteristo P, Exome Sequencing of Uterine Leiomyosarcomas Identifies Frequent Mutations in TP53, ATRX, and MED12, PLoS Genet. 12 (2016) e1005850. 10.1371/journal.pgen.1005850. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [9].Choi J, Manzano A, Dong W, Bellone S, Bonazzoli E, Zammataro L, Yao X, Deshpande A, Zaidi S, Guglielmi A, Gnutti B, Nagarkatti N, Tymon-Rosario JR, Harold J, Mauricio D, Zeybek B, Menderes G, Altwerger G, Jeong K, Zhao S, Buza N, Hui P, Ravaggi A, Bignotti E, Romani C, Todeschini P, Zanotti L, Odicino F, Pecorelli S, Ardighieri L, Bilguvar K, Quick CM, Silasi D-A, Huang GS, Andikyan V, Clark M, Ratner E, Azodi M, Imielinski M, Schwartz PE, Alexandrov LB, Lifton RP, Schlessinger J, Santin AD, Integrated mutational landscape analysis of uterine leiomyosarcomas, PNAS. 118 (2021). 10.1073/pnas.2025182118. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [10].Amorim JP, Santos G, Vinagre J, Soares P, The Role of ATRX in the Alternative Lengthening of Telomeres (ALT) Phenotype, Genes (Basel). 7 (2016). 10.3390/genes7090066. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [11].Lecona E, Fernandez-Capetillo O, Targeting ATR in cancer, Nat Rev Cancer. 18 (2018) 586–595. 10.1038/s41568-018-0034-3. [DOI] [PubMed] [Google Scholar]
- [12].Wengner AM, Siemeister G, Lücking U, Lefranc J, Wortmann L, Lienau P, Bader B, Bömer U, Moosmayer D, Eberspächer U, Golfier S, Schatz CA, Baumgart SJ, Haendler B, Lejeune P, Schlicker A, von Nussbaum F, Brands M, Ziegelbauer K, Mumberg D, The Novel ATR Inhibitor BAY 1895344 Is Efficacious as Monotherapy and Combined with DNA Damage–Inducing or Repair–Compromising Therapies in Preclinical Cancer Models, Mol Cancer Ther. 19 (2020) 26–38. 10.1158/1535-7163.MCT-19-0019. [DOI] [PubMed] [Google Scholar]
- [13].Dyer MA, Qadeer ZA, Valle-Garcia D, Bernstein E, ATRX and DAXX: Mechanisms and Mutations, Cold Spring Harb Perspect Med. 7 (2017). 10.1101/cshperspect.a026567. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [14].Yang C-Y, Liau J-Y, Huang W-J, Chang Y-T, Chang M-C, Lee J-C, Tsai J-H, Su Y-N, Hung C-C, Jeng Y-M, Targeted next-generation sequencing of cancer genes identified frequent TP53 and ATRX mutations in leiomyosarcoma, Am J Transl Res. 7 (2015) 2072–2081. [PMC free article] [PubMed] [Google Scholar]
- [15].Yap TA, Tan DSP, Terbuch A, Caldwell R, Guo C, Goh BC, Heong V, Haris NRM, Bashir S, Drew Y, Hong DS, Meric-Bernstam F, Wilkinson G, Hreiki J, Wengner AM, Bladt F, Schlicker A, Ludwig M, Zhou Y, Liu L, Bordia S, Plummer R, Lagkadinou E, de Bono JS, First-in-Human Trial of the Oral Ataxia Telangiectasia and RAD3-Related (ATR) Inhibitor BAY 1895344 in Patients with Advanced Solid Tumors, Cancer Discov. 11 (2021) 80–91. 10.1158/2159-8290.CD-20-0868. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [16].Flynn RL, Cox KE, Jeitany M, Wakimoto H, Bryll AR, Ganem NJ, Bersani F, Pineda JR, Suvà ML, Benes CH, Haber DA, Boussin FD, Zou L, Alternative Lengthening of Telomeres Renders Cancer Cells Hypersensitive to ATR Inhibitors, Science. 347 (2015) 273–277. 10.1126/science.1257216. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [17].Hu Y, Shi G, Zhang L, Li F, Jiang Y, Jiang S, Ma W, Zhao Y, Songyang Z, Huang J, Switch telomerase to ALT mechanism by inducing telomeric DNA damages and dysfunction of ATRX and DAXX, Scientific Reports. 6 (2016) 32280. 10.1038/srep32280. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [18].Mosele F, Remon J, Mateo J, Westphalen CB, Barlesi F, Lolkema MP, Normanno N, Scarpa A, Robson M, Meric-Bernstam F, Wagle N, Stenzinger A, Bonastre J, Bayle A, Michiels S, Bièche I, Rouleau E, Jezdic S, Douillard J-Y, Reis-Filho JS, Dienstmann R, André F, Recommendations for the use of next-generation sequencing (NGS) for patients with metastatic cancers: a report from the ESMO Precision Medicine Working Group, Ann Oncol. 31 (2020) 1491–1505. 10.1016/j.annonc.2020.07.014. [DOI] [PubMed] [Google Scholar]
- [19].Kurashima K, Kashiwagi H, Shimomura I, Suzuki A, Takeshita F, Mazevet M, Harata M, Yamashita T, Yamamoto Y, Kohno T, Shiotani B, SMARCA4 deficiency-associated heterochromatin induces intrinsic DNA replication stress and susceptibility to ATR inhibition in lung adenocarcinoma, NAR Cancer. 2 (2020) zcaa005. 10.1093/narcan/zcaa005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [20].Gupta M, Concepcion CP, Fahey CG, Keshishian H, Bhutkar A, Brainson CF, Sanchez-Rivera FJ, Pessina P, Kim JY, Simoneau A, Paschini M, Beytagh MC, Stanclift CR, Schenone M, Mani DR, Li C, Oh A, Li F, Hu H, Karatza A, Bronson RT, Shaw AT, Hata AN, Wong K-K, Zou L, Carr SA, Jacks T, Kim CF, BRG1 Loss Predisposes Lung Cancers to Replicative Stress and ATR Dependency, Cancer Res. 80 (2020) 3841–3854. 10.1158/0008-5472.CAN-20-1744. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [21].Mardinian K, Adashek JJ, Botta GP, Kato S, Kurzrock R, SMARCA4: Implications of an altered chromatin-remodeling gene for cancer development and therapy, Mol Cancer Ther. (2021) 10.1158/1535-7163.MCT-21-0433. 10.1158/1535-7163.MCT-21-0433. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [22].Bayer A Multicenter, Non-randomized, Open-label Phase 1b Study to Determine the Maximum Tolerated and Recommended Phase 2 Dose of the ATR Inhibitor Elimusertib in Combination With Pembrolizumab and to Characterize Its Safety, Tolerability, Pharmacokinetics and Preliminary Anti-tumor Activity in Participants With Advanced Solid Tumors, clinicaltrials.gov, 2022. https://clinicaltrials.gov/ct2/show/NCT04095273 (accessed March 31, 2022).
- [23].National Cancer Institute (NCI), BAY 1895344 Plus Topoisomerase-1 (Top1) Inhibitors in Patients With Advanced Solid Tumors, Phase I Studies With Expansion Cohorts in Small Cell Lung Carcinoma (SCLC), Poorly Differentiated Neuroendocrine Carcinoma (PD-NEC) and Pancreatic Adenocarcinoma (PDA), clinicaltrials.gov, 2022. https://clinicaltrials.gov/ct2/show/NCT04514497 (accessed March 31, 2022).
