3090 the mat2a inhibitor ag-270 combines with both taxanes

1
Acknowledgments We would like to thank Champions Oncology, ChemPartner, CrownBio, and Horizon Discovery Ltd. for support with experimental work. Disclosures This work was funded by Agios Pharmaceuticals, Inc. Some of these data were previously presented in Kalev P et al. AACR-NCI-EORTC International Conference on Molecular Targets and Cancer Therapeutics, October 26–30, 2019, Boston, MA, USA: Poster B115. MLH, PK, MF, C-CC, EA-F, EM, SN, ML, RN, YT, JM, SAB, KMM, and KM: Agios – employment and stockholder. Editorial assistance was provided by Christine Ingleby, PhD, CMPP, Excel Medical Affairs, Horsham, UK, and supported by Agios. References 1. Beroukhim R et al. Nature 2010;463:899–905. 2. Zhang H et al. Cancer Genet Cytogenet 1996;86:22–8. 3. Cerami E et al. Cancer Discov. 2012;2:401–4. 4. Marjon K et al. Cell Rep 2016;15:544–87. 5. Braun CJ et al. Cancer Cell 2017;32:411–26. 6. Boutz PL et al. Genes Dev 2015;29:63–80. 7. Foucquier J, Guedj M. Pharmacol Res Perspect 2015;3:e00149. 8. Huck JJ et al. Mol Cancer Ther 2014;13:2170–83. BACKGROUND AG-270 is a first-in-class, oral, potent, reversible inhibitor of methionine adenosyltransferase 2A (MAT2A), currently being evaluated in a phase 1 trial in patients with advanced solid tumors and lymphomas with MTAP (S-methyl-5’-thioadenosine phosphorylase) deletion (ClinicalTrials.gov NCT03435250) MAT2A is a key enzyme in the methionine salvage pathway, responsible for generating the universal methyl donor, S-adenosylmethionine (SAM) 1,2 MTAP is deleted in ~15–25% of non–small cell lung cancers (NSCLC), ~25% of pancreatic, and ~30% of esophageal cancers 3 Biallelic loss of the MTAP gene sensitizes cancer cells to genetic depletion of protein arginine methyltransferase 5 (PRMT5) and the upstream metabolic enzyme, MAT2A (Figure 1) 4 To prioritize candidate combination partners for AG-270, a cell-based in vitro screening approach was employed using MTAP-null cell lines, in which AG-270 was combined with standard-of-care (SOC) agents as well as agents targeting pathways with hypothesized mechanistic links to MAT2A 5 To identify combination partners that potentiate the cell growth inhibition effects of MAT2A blockade To assess the tolerability and antitumor activity of AG-270 combined with SOC agents in clinically relevant in vivo patient-derived xenograft (PDX) and cell-derived xenograft (CDX) models To investigate the mechanism of action associated with AG-270 combined with taxane therapy METHODS An in vitro synergy screen tested 20 candidate combination partners in 37 MTAP-null cell lines, with full-dose curve matrices performed at Horizon using a 96-hr CellTiter-Glo readout Cell lines included kidney (n = 1), colorectal (n = 4), esophageal and gastric (n = 7), pancreatic (n = 8), and lung (n = 17) The strength of potential synergistic interactions between AG-270 and enhancer compounds in vitro in excess of Loewe additivity was measured using the Horizon- developed Synergy Score Splicing changes were determined from RNA sequencing data using rMATS 3.2.5; changes in usage of detained introns 6 were determined using DEXSeq The splicing changes were selected using the criterion false discovery rate–adjusted p-value < 0.05 In vivo xenograft studies were performed using institutional animal care and use committee guidelines; the combination analysis used belongs to the ‘response additivity’ class of synergy analysis, 7,8 with area under the tumor growth curves (AUC) compared between single-agent and combination treatments Hematoxylin and eosin (H&E) staining on formalin-fixed paraffin-embedded tumor tissues was performed by Mosaic Laboratories in accordance with validated procedures, with the percentage of multinucleated tumor cells and cytomegaly assessed by a pathologist Six mice were evaluated per treatment arm; all tissues were collected 12–24 hr after the last dose of AG-270 RESULTS A large-scale synergy screen revealed potential synergistic combinations (Figure 2) MAT2A inhibition led to substantial dysregulation of splicing in MTAP-null cells, including an increased number of transcripts containing detained introns (Figure 3A) Detained intron–containing transcripts fail to export into the cytosol and thus are not translated 6 MAT2A inhibitor–induced detained introns included genes involved in the DNA damage response and cell-cycle regulation, such as Aurora kinase B (Figure 3B) Quantitative reverse transcriptase (RT)-PCR analysis demonstrated that treatment with a MAT2A inhibitor led to increased expression of detained intron–containing transcripts and decreased levels of total (exon-exon) Aurora B expression (Figure 3C) OBJECTIVES CONCLUSIONS A cell-based screen identified taxanes and gemcitabine as therapeutic agents that could yield combination benefits with AG-270 MAT2A inhibition led to substantial dysregulation of splicing, including an increased number of transcripts containing detained introns, in MTAP-null cells, including in pathways that regulate the DNA damage response and cell cycle, which we suggest are mechanistically relevant to efficacy These data form the basis for combining AG-270 with taxanes (antimitotic agents) and gemcitabine (DNA-damaging agent) Combining AG-270 with taxanes and gemcitabine yielded additive-to- synergistic antitumor activity, with the docetaxel combination yielding 50% complete tumor regressions in select models; combination benefits were observed in PDX models derived from esophageal, NSCLC, and pancreatic cancers The combination of AG-270 with taxanes induced mitotic defects in tumor cells, as demonstrated by increased cytomegaly and multinucleation These preclinical findings have inspired an ongoing phase 1 study exploring AG-270 combined with docetaxel or gemcitabine/nab- paclitaxel (ClinicalTrials.gov NCT03435250) The MAT2A inhibitor AG-270 combines with both taxanes and gemcitabine to yield enhanced antitumor activity in patient-derived xenograft models Marc L Hyer, Peter Kalev, Mark Fletcher, Chi-Chao Chen, Elia Aguado-Fraile, Everton Mandley, Sheila Newhouse, Max Lein, Raj Nagaraja, Yesim Tuncay, Josh Murtie, Scott A Biller, Kevin M Marks, Katya Marjon Agios Pharmaceuticals, Inc., Cambridge, MA, USA Presented at the American Association for Cancer Research Virtual Annual Meeting II, June 22–24, 2020 3090 Figure 2. AG-270 synergized with antimitotic taxanes and gemcitabine in a cell-based assay A MAT2A inhibitor tool compound (AGI-24512) was used for all experiments shown in Figure 3 A. Splicing changes associated with MAT2A inhibition in HCT116 MTAP-null and MTAP-wt cells, determined by DEXSeq (detained introns) and rMATS (others) B. The Venn diagram shows the following three sets: genes with detained introns upregulated on MAT2A inhibition in HCT116 MTAP-null cells; genes in pathway GO:6281 DNA repair; and genes in pathway GO:0278 cell cycle C. Quantitative RT-PCR analysis demonstrated that treatment with a MAT2A inhibitor affected expression of detained intron–containing transcripts and total (exon-exon) Aurora B expression. Data shown are from a representative experiment performed in triplicate DMSO = dimethyl sulfoxide; wt = wild type Figure 3. MAT2A inhibition disrupted splicing and altered gene expression in MTAP-null cells Table 1. Efficacy data summary of AG-270 and taxanes, alone and combined Tumor model (indication, subtype) Taxane partner AG-270 tumor growth inhibition, % Taxane tumor growth inhibition, % Combination tumor growth inhibition, % Tolerability: Maximum mean BWL, % Combination analysis 7,8 KP4 (pancreatic) Docetaxel 66 70 94 < 5 Synergy ESX030 (esophageal, SCC) Docetaxel 68 44 90 < 2.5 Synergy LUX001 (NSCLC, SCC) Docetaxel 70 45 91 6 Synergy PA0372 (pancreatic) Paclitaxel 59 29 84 < 2.5 Additive ESX2263 a (esophageal, SCC) Docetaxel 27 12 57 < 5.0 Additive LUX034 (NSCLC, SCC) Docetaxel 41 27 60 < 6 Additive LU6412 b (NSCLC, Ad) Docetaxel 52 57 92 13.3 Additive PAX001 (pancreatic) Paclitaxel 94 22 94 7 Weakly additive CTG-1194 (NSCLC, Ad) Docetaxel 15 42 52 0 Additive PAX041 (pancreatic) Paclitaxel 55 12 50 < 2.5 Weakly additive Tumor growth inhibition % = [1–(TV i –TV 0 )/(TV vi –TV v0 )] ×100%; where TV i is the average tumor volume of the test group on a specific day and TV 0 is the average tumor volume of the same group on day 0, and TV vi is the average tumor volume of the vehicle group on a specific day and TV v0 is the average tumor volume of the vehicle group on day 0 Paclitaxel doses used in PAX001 and PAX041 were 7.5 mg/kg; in PA0372, 15 mg/kg. Docetaxel doses used in KP4, ESX030, ESX2263, CTG-1194, LU6412, LUX034 were 5 mg/kg; in LUX001, 2.5 mg/kg. Number of animals used per arm per model was 8–12 a Dosing holidays were provided b 1 of 12 animals was found dead in the combination group Ad = adenocarcinoma; BWL = body weight loss; SCC = squamous cell carcinoma Figure 5. AG-270 enhanced docetaxel therapy in an esophageal (SCC) MTAP-null PDX mouse model 0 50 100 150 0 500 1000 1500 2000 On Day 120, three tumors > 1000 mm 3 were removed. Last dose delivered on Day 135. On Day 136, three animals with tumors were removed from the study; the remaining six animals presented with no detectable tumor (complete response) and remained tumor free through end of the study, Day 155. Vehicle AG-270 AG-270 + docetaxel Docetaxel Treatment day Turnover volume (mm 3 ) Figure 6. Treatment with AG-270 combined with taxanes enhanced mitotic defects in xenograft tumors Tumor tissues were harvested and analyzed at the end of an efficacy study after multiple weeks of dosing; n = 6 animals per condition A. Quantification of the percentage of multinucleated cells and cells with cytomegaly by H&E staining and pathologist review in pancreatic xenograft models (KP4 and PA0372) treated with AG-270 as a single agent or in combination with taxanes. A nonparametric Kruskal-Wallis test with multiple comparison correction was used; * p < 0.05, ** p < 0.01, *** p < 0.001 B. Representative images of abnormal tumor cell morphology in KP4 xenografts treated with AG-270 in combination with docetaxel or vehicle control. Yellow arrows indicate multinucleation examples and red arrows indicate cells with cytomegaly Vehicle AG-270 Docetaxel AG-270 + docetaxel 0 1 2 3 4 KP4 model Multinucleated tumor cells (%) *** Vehicle AG-270 Paclitaxel AG-270 + paclitaxel 0 2 4 6 PA0372 model Multinucleated tumor cells (%) * * Vehicle AG-270 Docetaxel AG-270 + docetaxel 0 1 2 3 4 5 Tumor cells with cytomegaly (%) *** Vehicle AG-270 Paclitaxel AG-270 + paclitaxel 0 5 10 15 Tumor cells with cytomegaly (%) ** * A. Quantification of multinucleated cells and cytomegaly in pancreatic xenograft tumors B. Tumor cell morphology in KP4 xenograft tumors Multinucleated cells Cells with cytomegaly Vehicle AG-270 + docetaxel Table 2. Efficacy data summary of AG-270 and gemcitabine, alone and combined Tumor model (indication, subtype) AG-270 tumor growth inhibition, % Gemcitabine tumor growth inhibition, % Combination tumor growth inhibition, % Tolerability: Maximum mean BWL, % Combination analysis 7,8 KP4 (pancreatic) 66 43 82 < 5 Additive PAX041 (pancreatic) 55 16 69 < 5 Additive LU6431 (NSCLC, SCC) 45 43 69 < 5 Additive PAX001 (pancreatic) 83 56 89 < 5 Weakly additive LU1513 a (NSCLC, SCC) 74 90 101 < 11 Weakly additive Tumor growth inhibition calculated as described for Table 1 Number of animals used per arm per model was 8–12 a In the LU1513 study, one of 12 animals in the combination arm lost > 20% BWL on Day 11 and was removed from the study; maximum mean BWL in this group was 10% Figure 1. Targeting MAT2A in cancers with MTAP deletion Me = methylation; MTA = 5’-methylthioadenosine; MTR-1P = 5-methylthioribose-1-phosphate MTAP enzyme is lost MTA AG-270 accumulates MTR-1P MTAP PRMT5 MAT2A Me Me Me Me Splicing event modulation in DNA damage and cell cycle pathways SAM Methionine Chromosome 9p21 deleted in 15% of cancers Splicing Complex Treatment with vehicle, AG-270 (oral 100 mg/kg once daily), docetaxel (intravenous 5 mg/kg on Days 1, 15, 29, and weekly thereafter), or AG-270 + docetaxel in an esophageal (ESX030) PDX model. One animal in the AG-270 + docetaxel group experienced BWL > 20% and received an AG-270 dosing holiday from Days 55 to 60; BWL recovered. Maximum mean BWL for the combination group was < 3%. Y-axis shows mean + SE; n = 12 animals per group Treatment with vehicle, AG-270 (oral 100 mg/kg once daily), gemcitabine (intraperitoneal 20 mg/kg every 3 days), or AG-270 + gemcitabine in a pancreatic CDX model (KP4). Maximum mean BWL in the combination group was < 5%. Y-axis shows mean + SE; n = 12 animals per group Figure 7. AG-270 enhanced gemcitabine therapy in an MTAP-null pancreatic CDX (KP4) mouse model 0 10 20 30 40 0 500 1000 1500 2000 Treatment day Turnover volume (mm 3 ) Vehicle AG-270 AG-270 + gemcitabine Gemcitabine 0 5 10 15 20 25 Combination partners Synergy score Gemcitabine Ranked by median extent of synergy Aurora kinase inhibitor AZD1152 Aurora kinase inhibitor ABT-348 Paclitaxel A summary of the efficacy of AG-270 and taxanes as single agents and in combination in xenograft models is shown in Table 1 AG-270 demonstrated increased antitumor activity when combined with docetaxel in a lung PDX mouse model (Figure 4) AG-270 demonstrated increased antitumor activity when combined with docetaxel in an esophageal PDX mouse model (Figure 5) The combination of AG-270 with taxanes induced cytomegaly (abnormal cell enlargement) and multinucleation in xenograft tumor cells (Figure 6) A summary of the efficacy of AG-270 and gemcitabine as single agents and in combination in xenograft models is shown in Table 2 AG-270 demonstrated increased antitumor activity when combined with gemcitabine in a pancreatic CDX mouse model (Figure 7) Figure 4. AG-270 enhanced docetaxel therapy in an NSCLC (SCC) MTAP-null PDX mouse model 0 50 100 150 0 500 1000 1500 2000 Vehicle AG-270 AG-270 + docetaxel Docetaxel Treatment day Turnover volume (mm 3 ) Treatment with vehicle, AG-270 (oral 100 mg/kg once daily), docetaxel (intravenous 2.5 mg/kg every 7 days × 18 doses), or AG-270 + docetaxel in a lung (LUX001) PDX model. BWL was observed in two animals in the AG-270 group (20% and 16%) and all groups were given AG-270 dosing holidays on Days 16–21. In the AG-270 + docetaxel group, one animal reached 20% BWL and was given AG-270 dosing holidays on Days 54–59, 65–73, and 77–83; one animal reached > 20% BWL and was given an AG-270 dosing holiday on Days 38–46 and a docetaxel dosing holiday on Day 42. Body weight recovered in all animals and maximum mean BWL for the combination group was 6%. Y-axis shows mean + SE; n = 8 animals per group DMSO MAT2A inhibitor 0 1 2 3 4 5 DMSO MAT2A inhibitor 0.0 0.5 1.0 1.5 A B C 88 48 12 131 HCT116 MTAP –/– HCT116 MTAP –/– Aurora kinase B gene expression (fold change) Cell cycle 1012 Detained introns up 1216 DNA repair 526 Aurora kinase B detained introns (fold change) 0 500 1000 1500 2000 Number of splicing events altered upon MAT2A inhibition Alternative 5' splicing Alternative 3' splicing Mutually exclusive exons Retained introns Skipped exons Detained introns HCT116 MTAP wt HCT116 MTAP –/– Last dose was delivered on Day 114. On Day 120, four animals with tumors were removed from the group; the remaining four animals presented with no detectable tumor (complete response) and remained tumor free through end of the study, Day 141 [email protected]

Upload: others

Post on 24-May-2022

2 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: 3090 The MAT2A inhibitor AG-270 combines with both taxanes

Acknowledgments We would like to thank Champions Oncology, ChemPartner, CrownBio, and Horizon Discovery Ltd. for support with experimental work.

Disclosures This work was funded by Agios Pharmaceuticals, Inc. Some of these data were previously presented in Kalev P et al. AACR-NCI-EORTC International Conference on Molecular Targets and Cancer Therapeutics, October 26–30, 2019, Boston, MA, USA: Poster B115. MLH, PK, MF, C-CC, EA-F, EM, SN, ML, RN, YT, JM, SAB, KMM, and KM: Agios – employment and stockholder. Editorial assistance was provided by Christine Ingleby, PhD, CMPP, Excel Medical Affairs, Horsham, UK, and supported by Agios.

References 1. Beroukhim R et al. Nature 2010;463:899–905.2. Zhang H et al. Cancer Genet Cytogenet 1996;86:22–8.3. Cerami E et al. Cancer Discov. 2012;2:401–4.4. Marjon K et al. Cell Rep 2016;15:544–87. 5. Braun CJ et al. Cancer Cell 2017;32:411–26. 6. Boutz PL et al. Genes Dev 2015;29:63–80. 7. Foucquier J, Guedj M. Pharmacol Res Perspect 2015;3:e00149.8. Huck JJ et al. Mol Cancer Ther 2014;13:2170–83.

BACKGROUND• AG-270 is a first-in-class, oral, potent, reversible inhibitor of methionine

adenosyltransferase 2A (MAT2A), currently being evaluated in a phase 1 trial in patientswith advanced solid tumors and lymphomas with MTAP (S-methyl-5’-thioadenosinephosphorylase) deletion (ClinicalTrials.gov NCT03435250)

• MAT2A is a key enzyme in the methionine salvage pathway, responsible for generatingthe universal methyl donor, S-adenosylmethionine (SAM)1,2

• MTAP is deleted in ~15–25% of non–small cell lung cancers (NSCLC), ~25% ofpancreatic, and ~30% of esophageal cancers3

• Biallelic loss of the MTAP gene sensitizes cancer cells to genetic depletion of proteinarginine methyltransferase 5 (PRMT5) and the upstream metabolic enzyme, MAT2A (Figure 1)4

• To prioritize candidate combination partners for AG-270, a cell-based in vitro screeningapproach was employed using MTAP-null cell lines, in which AG-270 was combined withstandard-of-care (SOC) agents as well as agents targeting pathways with hypothesizedmechanistic links to MAT2A5

• To identify combination partners that potentiate the cell growth inhibition effects ofMAT2A blockade

• To assess the tolerability and antitumor activity of AG-270 combined with SOC agentsin clinically relevant in vivo patient-derived xenograft (PDX) and cell-derived xenograft(CDX) models

• To investigate the mechanism of action associated with AG-270 combined with taxane therapy

METHODS• An in vitro synergy screen tested 20 candidate combination partners in 37 MTAP-null cell

lines, with full-dose curve matrices performed at Horizon using a 96-hr CellTiter-Glo readout− Cell lines included kidney (n = 1), colorectal (n = 4), esophageal and gastric (n = 7),

pancreatic (n = 8), and lung (n = 17)− The strength of potential synergistic interactions between AG-270 and enhancer

compounds in vitro in excess of Loewe additivity was measured using the Horizon-developed Synergy Score

• Splicing changes were determined from RNA sequencing data using rMATS 3.2.5;changes in usage of detained introns6 were determined using DEXSeq− The splicing changes were selected using the criterion false discovery rate–adjusted

p-value < 0.05• In vivo xenograft studies were performed using institutional animal care and use

committee guidelines; the combination analysis used belongs to the ‘response additivity’class of synergy analysis,7,8 with area under the tumor growth curves (AUC) comparedbetween single-agent and combination treatments

• Hematoxylin and eosin (H&E) staining on formalin-fixed paraffin-embedded tumor tissueswas performed by Mosaic Laboratories in accordance with validated procedures, with thepercentage of multinucleated tumor cells and cytomegaly assessed by a pathologist− Six mice were evaluated per treatment arm; all tissues were collected 12–24 hr after

the last dose of AG-270

RESULTS• A large-scale synergy screen revealed potential synergistic combinations (Figure 2)• MAT2A inhibition led to substantial dysregulation of splicing in MTAP-null cells, including

an increased number of transcripts containing detained introns (Figure 3A)• Detained intron–containing transcripts fail to export into the cytosol and thus are not translated6

• MAT2A inhibitor–induced detained introns included genes involved in the DNA damageresponse and cell-cycle regulation, such as Aurora kinase B (Figure 3B)

• Quantitative reverse transcriptase (RT)-PCR analysis demonstrated that treatment witha MAT2A inhibitor led to increased expression of detained intron–containing transcriptsand decreased levels of total (exon-exon) Aurora B expression (Figure 3C)

OBJECTIVES

CONCLUSIONS

• A cell-based screen identified taxanes and gemcitabine as therapeuticagents that could yield combination benefits with AG-270

• MAT2A inhibition led to substantial dysregulation of splicing, includingan increased number of transcripts containing detained introns, in MTAP-null cells, including in pathways that regulate the DNA damage response and cell cycle, which we suggest are mechanistically relevant to efficacy

• These data form the basis for combining AG-270 with taxanes (antimitotic agents) and gemcitabine (DNA-damaging agent)

• Combining AG-270 with taxanes and gemcitabine yielded additive-to-synergistic antitumor activity, with the docetaxel combination yielding 50% complete tumor regressions in select models; combination benefits were observed in PDX models derived from esophageal, NSCLC, and pancreatic cancers

• The combination of AG-270 with taxanes induced mitotic defects in tumor cells, as demonstrated by increased cytomegaly and multinucleation

• These preclinical findings have inspired an ongoing phase 1 study exploring AG-270 combined with docetaxel or gemcitabine/nab-paclitaxel (ClinicalTrials.gov NCT03435250)

The MAT2A inhibitor AG-270 combines with both taxanes and gemcitabine to yield enhanced antitumor activity in patient-derived xenograft models

Marc L Hyer, Peter Kalev, Mark Fletcher, Chi-Chao Chen, Elia Aguado-Fraile, Everton Mandley, Sheila Newhouse, Max Lein, Raj Nagaraja, Yesim Tuncay, Josh Murtie, Scott A Biller, Kevin M Marks, Katya Marjon

Agios Pharmaceuticals, Inc., Cambridge, MA, USA

Presented at the American Association for Cancer Research Virtual Annual Meeting II, June 22–24, 2020

3090

Figure 2. AG-270 synergized with antimitotic taxanes and gemcitabine in a cell-based assay

A MAT2A inhibitor tool compound (AGI-24512) was used for all experiments shown in Figure 3 A. Splicing changes associated with MAT2A inhibition in HCT116 MTAP-null and MTAP-wt cells, determined by DEXSeq (detained introns)and rMATS (others) B. The Venn diagram shows the following three sets: genes with detained introns upregulated on MAT2A inhibition in HCT116 MTAP-null cells; genes in pathway GO:6281 DNA repair; and genes in pathway GO:0278 cell cycleC. Quantitative RT-PCR analysis demonstrated that treatment with a MAT2A inhibitor affected expression of detained intron–containing transcripts andtotal (exon-exon) Aurora B expression. Data shown are from a representative experiment performed in triplicateDMSO = dimethyl sulfoxide; wt = wild type

Figure 3. MAT2A inhibition disrupted splicing and altered gene expression in MTAP-null cells

Table 1. Efficacy data summary of AG-270 and taxanes, alone and combined

Tumor model (indication, subtype) Taxane partner AG-270 tumor growth inhibition, %

Taxane tumor growth inhibition, %

Combination tumor growth inhibition, %

Tolerability: Maximum mean BWL, %

Combination analysis7,8

KP4 (pancreatic) Docetaxel 66 70 94 < 5 SynergyESX030 (esophageal, SCC) Docetaxel 68 44 90 < 2.5 SynergyLUX001 (NSCLC, SCC) Docetaxel 70 45 91 6 SynergyPA0372 (pancreatic) Paclitaxel 59 29 84 < 2.5 AdditiveESX2263a (esophageal, SCC) Docetaxel 27 12 57 < 5.0 AdditiveLUX034 (NSCLC, SCC) Docetaxel 41 27 60 < 6 AdditiveLU6412b (NSCLC, Ad) Docetaxel 52 57 92 13.3 AdditivePAX001 (pancreatic) Paclitaxel 94 22 94 7 Weakly additiveCTG-1194 (NSCLC, Ad) Docetaxel 15 42 52 0 AdditivePAX041 (pancreatic) Paclitaxel 55 12 50 < 2.5 Weakly additive

Tumor growth inhibition % = [1–(TVi–TV0)/(TVvi–TVv0)] ×100%; where TVi is the average tumor volume of the test group on a specific day and TV0 is the average tumor volume of the same group on day 0, and TVvi is the average tumor volume of the vehicle group on a specific day and TVv0 is the average tumor volume of the vehicle group on day 0 Paclitaxel doses used in PAX001 and PAX041 were 7.5 mg/kg; in PA0372, 15 mg/kg. Docetaxel doses used in KP4, ESX030, ESX2263, CTG-1194, LU6412, LUX034 were 5 mg/kg; in LUX001, 2.5 mg/kg. Number of animals used per arm per model was 8–12 aDosing holidays were provided b1 of 12 animals was found dead in the combination group Ad = adenocarcinoma; BWL = body weight loss; SCC = squamous cell carcinoma

Figure 5. AG-270 enhanced docetaxel therapy in an esophageal (SCC) MTAP-null PDX mouse model

0 50 100 1500

500

1000

1500

2000

On Day 120, three tumors > 1000 mm3 were removed.

Last dose delivered on Day 135. On Day 136, three animals with tumors wereremoved from the study; the remaining six animals presented with no detectable tumor (complete response) and remained tumor free through end of the study, Day 155.

Vehicle

AG-270AG-270 + docetaxel

Docetaxel

Treatment day

Turn

over

vol

ume

(mm

3 )

Figure 6. Treatment with AG-270 combined with taxanes enhanced mitotic defects in xenograft tumors

Tumor tissues were harvested and analyzed at the end of an efficacy study after multiple weeks of dosing; n = 6 animals per condition A. Quantification of the percentage of multinucleated cells and cells with cytomegaly by H&E staining and pathologist review in pancreaticxenograft models (KP4 and PA0372) treated with AG-270 as a single agent or in combination with taxanes. A nonparametric Kruskal-Wallis test with multiple comparison correction was used; * p < 0.05, ** p < 0.01, *** p < 0.001 B. Representative images of abnormal tumor cell morphology in KP4 xenografts treated with AG-270 in combination with docetaxel or vehiclecontrol. Yellow arrows indicate multinucleation examples and red arrows indicate cells with cytomegaly

Vehicl

e

AG-270

Docetax

el

AG-270 +

docetax

el0

1

2

3

4

KP4 model

Mul

tinuc

leat

ed tu

mor

ce

lls (%

)

***

Vehicl

e

AG-270

Paclita

xel

AG-270 +

paclita

xel

0

2

4

6

PA0372 model

Mul

tinuc

leat

ed tu

mor

ce

lls (%

)

**

Vehicl

e

AG-270

Docetax

el

AG-270 +

docetax

el0

1

2

3

4

5

Tum

or c

ells

with

cy

tom

egal

y (%

)

***

Vehicl

e

AG-270

Paclita

xel

AG-270 +

paclita

xel

0

5

10

15

Tum

or c

ells

with

cyt

omeg

aly

(%)

***

A. Quantification of multinucleated cells and cytomegaly in pancreatic xenograft tumors

B. Tumor cell morphology in KP4 xenograft tumors

Multinucleated cellsCells with cytomegaly

Vehicle

AG-270 + docetaxel

Table 2. Efficacy data summary of AG-270 and gemcitabine, alone and combined

Tumor model (indication,subtype)

AG-270 tumor growth inhibition, %

Gemcitabine tumor growth inhibition, %

Combination tumor growth inhibition, %

Tolerability: Maximum mean

BWL, %

Combination analysis7,8

KP4 (pancreatic) 66 43 82 < 5 AdditivePAX041 (pancreatic) 55 16 69 < 5 Additive

LU6431 (NSCLC, SCC) 45 43 69 < 5 Additive

PAX001 (pancreatic) 83 56 89 < 5 Weakly additive

LU1513a (NSCLC, SCC) 74 90 101 < 11 Weakly additive

Tumor growth inhibition calculated as described for Table 1 Number of animals used per arm per model was 8–12aIn the LU1513 study, one of 12 animals in the combination arm lost > 20% BWL on Day 11 and was removed from the study; maximum mean BWL in this group was 10%

Figure 1. Targeting MAT2A in cancers with MTAP deletion

Me = methylation; MTA = 5’-methylthioadenosine; MTR-1P = 5-methylthioribose-1-phosphate

MTAP enzyme is lost

MTA

AG-270

accumulates

MTR-1P

MTAP

PRMT5 MAT2A

Me

Me MeMe

Splicing event modulation in DNA damage and cell cycle pathways

SAM Methionine

Chromosome 9p21deleted in 15% of cancers

SplicingComplex

Treatment with vehicle, AG-270 (oral 100 mg/kg once daily), docetaxel (intravenous 5 mg/kg on Days 1, 15, 29, and weekly thereafter), or AG-270 + docetaxel in an esophageal (ESX030) PDX model. One animal in the AG-270 + docetaxel group experienced BWL > 20% and received an AG-270 dosing holiday from Days 55 to 60; BWL recovered. Maximum mean BWL for the combination group was < 3%. Y-axis shows mean + SE; n = 12 animals per group

Treatment with vehicle, AG-270 (oral 100 mg/kg once daily), gemcitabine (intraperitoneal 20 mg/kg every 3 days), or AG-270 + gemcitabine in a pancreatic CDX model (KP4). Maximum mean BWL in the combination group was < 5%. Y-axis shows mean + SE; n = 12 animals per group

Figure 7. AG-270 enhanced gemcitabine therapy in an MTAP-null pancreatic CDX (KP4) mouse model

0 10 20 30 400

500

1000

1500

2000

Treatment day

Turn

over

vol

ume

(mm

3 )

Vehicle

AG-270AG-270 + gemcitabine

Gemcitabine

0

5

10

15

20

25

Combination partners

Syne

rgy

scor

e

Gem

cita

bine

Ranked by median extent of synergyA

uror

a ki

nase

inhi

bito

r AZD

1152

Aur

ora

kina

se in

hibi

tor

AB

T-34

8

Pac

litax

el

• A summary of the efficacy of AG-270 and taxanes as single agents and in combination inxenograft models is shown in Table 1

• AG-270 demonstrated increased antitumor activity when combined with docetaxelin a lung PDX mouse model (Figure 4)

• AG-270 demonstrated increased antitumor activity when combined with docetaxelin an esophageal PDX mouse model (Figure 5)

• The combination of AG-270 with taxanes induced cytomegaly (abnormal cell enlargement)and multinucleation in xenograft tumor cells (Figure 6)

• A summary of the efficacy of AG-270 and gemcitabine as single agents and in combinationin xenograft models is shown in Table 2

• AG-270 demonstrated increased antitumor activity when combined with gemcitabinein a pancreatic CDX mouse model (Figure 7)

Figure 4. AG-270 enhanced docetaxel therapy in an NSCLC (SCC) MTAP-null PDX mouse model

0 50 100 1500

500

1000

1500

2000 Vehicle

AG-270AG-270 + docetaxel

Docetaxel

Treatment day

Turn

over

vol

ume

(mm

3 )

Treatment with vehicle, AG-270 (oral 100 mg/kg once daily), docetaxel (intravenous 2.5 mg/kg every 7 days × 18 doses), or AG-270 + docetaxel in a lung (LUX001) PDX model. BWL was observed in two animals in the AG-270 group (20% and 16%) and all groups were given AG-270 dosing holidays on Days 16–21. In the AG-270 + docetaxel group, one animal reached 20% BWL and was given AG-270 dosing holidays on Days 54–59, 65–73, and 77–83; one animal reached > 20% BWL and was given an AG-270 dosing holiday on Days 38–46 and a docetaxel dosing holiday on Day 42. Body weight recovered in all animals and maximum mean BWL for the combination group was 6%. Y-axis shows mean + SE; n = 8 animals per group

DMSO MAT2A inhibitor0

1

2

3

4

5

DMSO MAT2A inhibitor0.0

0.5

1.0

1.5

A B

C

88

4812 131

HCT116 MTAP–/– HCT116 MTAP–/–

Aur

ora

kina

se B

gen

eex

pres

sion

(fol

d ch

ange

)

Cell cycle1012

Detained introns up

1216

DNArepair526

Aur

ora

kina

se B

det

aine

din

tron

s (fo

ld c

hang

e)

0 500 1000 1500 2000Number of splicing events altered

upon MAT2A inhibition

Alternative 5' splicingAlternative 3' splicingMutually exclusive exonsRetained intronsSkipped exonsDetained introns

HCT116 MTAP wt

HCT116 MTAP –/–

Last dose was delivered on Day 114. On Day 120, four animals with tumors were removed from the group; the remaining four animals presented with no detectable tumor (complete response) and remained tumor free through end of the study, Day 141

[email protected]