ros - x-fold change in ros+ cells from baseline: alexander … › 265040820 › files ›...

1
In Vitro and In Vivo Anticancer Effects of D/L-alpha-metyrosine (SM-88), A Novel Metabolism-Based Therapy CORRESPONDING AUTHOR: (e): [email protected] (ph): +1 (212) 461-2314 Alexander Vandell 1 Jonathan Eckard 1 Steve Hoffman 1 Giuseppe Del Priore 1 Martin Fernandez-Zapico 2 1 TYME Inc.; New York, New York 2 Mayo Clinic; Rochester, Minnesota D/L-alpha-metyrosine (SM-88, racemetyrosine) has displayed broad anticancer activity in patients across 15 different tumor types. While amino acid metabolism has been leveraged in oncology imaging for decades, SM-88 offers a potential novel therapeutic approach to selectively disrupt cancer cells. A comprehensive in vitro and in vivo experimental program is underway to elucidate the mechanism of action and further characterize the anti-cancer effects of SM-88. In vitro effects on apoptosis, viability, reactive oxygen species induction, autophagy, migration and invasion, cell cycle changes, and protein synthesis are being investigated. In vivo xenograft models are being used to explore the anti-cancer effects of SM-88 and its potential as an immune modulator. This poster provides an update on in vitro and in vivo data as of May 29, 2020. METHODS INTRODUCTION 1. Yamamoto, Keisuke et al. “Autophagy promotes immune evasion of pancreatic cancer by degrading MHC-I.” Nature vol. 581,7806 (2020): 100-105. doi:10.1038/s41586-020-2229-5 2. Renz, Bernhard W et al. “β2 Adrenergic-Neurotrophin Feedforward Loop Promotes Pancreatic Cancer.” Cancer cell vol. 33,1 (2018): 75-90.e7. doi:10.1016/j.ccell.2017.11.007 3. Zheng, Gen et al. “Human D-Tyr-tRNA(Tyr) deacylase contributes to the resistance of the cell to D-amino acids.” The Biochemical journal vol. 417,1 (2009): 85-94. doi:10.1042/BJ20080617 4. Kim, Hye-Jung, and Harvey Cantor. “CD4 T-cell subsets and tumor immunity: the helpful and the not-so-helpful.” Cancer immunology research vol. 2,2 (2014): 91-8. doi:10.1158/2326- 6066.CIR-13-0216 5. Nagano, Taiki et al. “d-amino acid oxidase promotes cellular senescence via the production of reactive oxygen species.” Life science alliance vol. 2,1 e201800045. 18 Jan. 2019, doi:10.26508/lsa.201800045 REFERENCES DISCUSSION AND FUTURE DIRECTIONS CONCLUSIONS RESULTS Oral Administration of SM-88 Reduces Tumor Growth in an HCT-116 Colon Cancer Xenograft Model A pilot in vivo xenograft experiment was conducted to evaluate the effects of SM-88 in a colon cancer model. HCT-116 tumor growth in mice treated with orally administered 324 mg/kg/day SM-88 was significantly reduced compared to those treated with Vehicle alone by Day 19 after treatment initiation (p < 0.05). No differences in body weight changes were noted between treatment groups. Number of HCT-116 Tumor-bearing Mice Alive on Each Treatment Day Day 1 5 8 12 15 19 Vehicle 11 11 11 11 11 11 81 mg/kg 11 11 11 11 11 11 162 mg/kg 11 11 11 11 11 10 324 mg/kg 11 11 11 11 11 9 Figure 1 Figure 2a Figure 2b IP Administration of SM-88 Reduces Tumor Growth in a Pan02 Pancreatic Cancer Syngenic Xenograft Model Alterations to the Pan02 Tumor Microenvironment Following Exposure to SM-88 (Day 34) Exposure to SM-88 may decrease intra-tumoral Treg populations, which may reduce immunosuppressive signaling within the tumor. Small increases in intracellular B cell populations were also observed following treatment with SM-88. N = 5 for all treatment groups. Figure 3: Treg and B Cells Figure 3d: Treg Figure 3e: B Cells Figure 3: CD4, CD8 (Total Number) and CD4/CD8 Ratio Figure 3a: CD4 Figure 3b: CD8 Figure 3c: CD4/CD8 Ratio To evaluate potential effects of SM-88 on the tumor immune microenvironment, flow cytometry was used to characterize the immune populations present in five randomly selected Pan02 tumors collected at study termination (Day 34). Exposure to SM-88 appears to decrease intra-tumoral CD4+ T cell populations, while preserving CD8+ populations, leading to decreases in the CD4+/CD8+ ratio. CD4/CD8 ratio was p= 0.015 for 75 mg/kg/day vs. Vehicle (ANOVA with Tukey post hoc). N = 5 for all treatment groups. SM-88 Induces Reactive Oxygen Species (ROS) Production The potential of SM-88 to increase ROS production was investigated using four cell lines, two pancreatic cancer (Pan02 and PANC1) and two breast cancer (4T1 and MCF-7). Dose dependent increases were observed in ROS production following 24 hours of exposure to SM-88 ME. Figure 4a: Pan02 Figure 4b: PANC1 Figure 4c: MCF-7 Figure 4d: 4T1 Figure 4: ROS Positive Cells 0.00 0.25 0.50 1.00 2.50 0 5 10 15 20 SM-88 Concentration (mM) Fold Change in ROS Positive Cells from Baseline 0.00 0.25 0.50 1.00 2.50 0 2 4 6 8 SM-88 Concentration (mM) Fold Change in ROS Positive Cells from Baseline 0.00 0.25 0.50 1.00 2.50 0 10 20 30 40 SM-88 Concentration (mM) Fold Change in ROS Positive Cells from Baseline 0.00 0.25 0.50 1.00 2.50 0 1 2 3 SM-88 Concentration (mM) Fold Change in ROS Positive Cells from Baseline 0 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 0 500 1000 1500 2000 Days Tumor Volume (mm 3 ) SM-88 Affects Autophagy Figure 5a The potential of SM-88 to affect autophagy was investigated using three cell lines, two pancreatic cancer (Pan02 and PANC1) and one ovarian cancer (HeLa). Exposure to SM-88 ME appeared to promote a dose dependent increase in LC3B expression at 24 hours in PANC1 and HeLa cells (Figure 5a). Exposure to 1 mM SM-88 ME promoted an increase in LC3B expression beginning at least 6 hours after treatment in both Pan02 and PANC1 cells. SM-88 ME exposure either did not affect or slightly increased p62 expression. These data suggest that SM-88 affects autophagic mechanisms, and follow-up experiments are underway to determine whether the increases in LC3B expression are due to autophagy induction or blockade of autophagosome and lysosome fusion. Figure 5b HeLa PANC1 Pan02 PANC1 Pan02 LC3B p62 GAPDH LC3B p62 GAPDH Pan02 xenograft size (mean ± SEM) Figure 3: Macrophages (Total Number) and M1/M2 Ratio Figure 3f: M1 Figure 3g: M2 Figure 3h: M1/M2 Ratio Exposure to SM-88 may preserve M1 polarized macrophage populations while decreasing populations of M2 polarized macrophages. Within the tumor microenvironment, M1 macrophages have anti-tumor effects, while M2 macrophages often promote tumor growth. Thus, alterations in the M1/M2 ratio could contribute to suppression or reversal of tumor growth. N = 5 for all treatment groups. e y y 0 5000 10000 15000 20000 Total number e y y 0 5000 10000 15000 20000 e y y 0 2 4 6 M1/M2 ratio To further evaluate the anti-cancer effects of SM-88, a second in vivo experiment was conducted using a pancreatic cancer model. Pan02 tumor growth in mice treated with intraperitoneally administered 75 mg/kg/day SM-88 ME was significantly reduced compared to those treated with Vehicle alone on Day 34 after treatment initiation (Figure 2a). All mice (n=30) were alive at end of treatment (Day 34). No differences in body weight changes were noted between treatment groups. Excised Pan02 tumors (Day 34) from the subcutaneous xenograft model are shown in Figure 2b. A higher proportion of mice treated with SM-88 maintained tumors below a volume of 500mm 3 (~2.5% of total body weight), with an indication of dose-related effect. SM-88 has demonstrated encouraging efficacy in 15 different tumor types. This may be due to the effects of SM-88 on cancer through multiple mechanisms, including immune modulation, alterations in autophagy and ROS induction. Alterations in autophagy have been associated with increased MHC1 expression in pancreatic cancer (Yamamoto et al., 2020). We are deepening our examination of the impact of SM-88 on autophagy/mitophagy, as well as any phenotypical changes that might be related to these changes including MHC1 expression. We are continuing to explore the immune modulatory effects observed in these preliminary experiments. Based on these and other ongoing experiments, we are beginning to explore SM-88 combinations with chemotherapies, targeted agents, or immuno-oncology therapies. We are continuing to explore these and other effects of SM-88, as well as potential biomarkers of SM-88 sensitivity, through experiments on human organoids, and metabolomic and gene expression analyses. The importance of amino acid metabolism in cancer has gained greater awareness over the past decade; however, potential therapeutic approaches in this area remain limited. SM-88 has been dosed in over 180 cancer patients and has shown encouraging efficacy and safety findings to date. Early data indicate that SM-88 may promote increases in ROS generation and alterations in autophagy in certain cancer cell types (Pan02 pancreatic cancer). Potential immune modulatory effects of SM-88, including alterations in macrophage polarization and other key tumor immune cells (CD4+ and Treg) are being examined. In all in vitro cell line experiments, and the Pan02 xenograft model, the SM-88 methyl-ester (SM-88 ME) was used, which has the same active moiety as SM-88, but with improved solubility characteristics. The SM-88 free acid form was used in the HCT-116 xenograft experiment. Autophagy: Changes in LC3B and p62 expression were investigated using standard Western Blot techniques. ROS Induction: Following treatment with SM-88, ROS induction was assessed using the CellROX (Thermo- Fisher) flow cytometry assay kit. Gating for ROS+/live cell selection was based on data obtained from negative controls. HCT-116 Xenograft: Female athymic nude mice were implanted subcutaneously with HCT-116 cells. Once tumors reached 50-100 mm 3 , mice were treated with either Vehicle alone, 81 mg/kg/day SM-88, 162 mg/kg/day SM-88, or 324 mg/kg/day SM-88 administered orally (n = 11 per group). Pan02 Xenograft Study: C57BL/6 mice were treated with either Vehicle alone, 25 mg/kg/day of SM-88 ME via intraperitoneal (IP) injection, or 75 mg/kg/day of SM-88 ME IP (n = 10 per group). Treatments began on Day 0. On Day 4, Pan02 cells were implanted subcutaneously into each mouse. Tumor Immune Profiling: On Day 34, Pan02 tumors from five randomly selected mice per treatment arm were processed for immune analysis by flow cytometry using a 16 color myeloid and lymphocyte panel following standard protocols.

Upload: others

Post on 24-Jun-2020

3 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: ROS - X-fold change in ROS+ cells from baseline: Alexander … › 265040820 › files › doc_downloads › ... · 2020-06-17 · In Vitro and In Vivo Anticancer Effects of D/L-alpha-metyrosine

In Vitro and In Vivo Anticancer Effects of D/L-alpha-metyrosine (SM-88), A Novel Metabolism-Based Therapy

CORRESPONDING AUTHOR:

(e): [email protected]

(ph): +1 (212) 461-2314

Alexander Vandell1 Jonathan Eckard1 Steve Hoffman1 Giuseppe Del Priore1 Martin Fernandez-Zapico2

1TYME Inc.; New York, New York 2Mayo Clinic; Rochester, Minnesota

D/L-alpha-metyrosine (SM-88, racemetyrosine) has displayed broad anticancer activity in patients across 15 different tumor types. While amino acid metabolism has been leveraged in oncology imaging for decades, SM-88 offers a potential novel therapeutic approach to selectively disrupt cancer cells.

A comprehensive in vitro and in vivo experimental program is underway to elucidate the mechanism of action and further characterize the anti-cancer effects of SM-88.

In vitro effects on apoptosis, viability, reactive oxygen species induction, autophagy, migration and invasion, cell cycle changes, and protein synthesis are being investigated.

In vivo xenograft models are being used to explore the anti-cancer effects of SM-88 and its potential as an immune modulator.

This poster provides an update on in vitro and in vivo data as of May 29, 2020.

METHODS

INTRODUCTION

1. Yamamoto, Keisuke et al. “Autophagy promotes immune evasion of pancreatic cancer by degrading MHC-I.” Nature vol. 581,7806 (2020): 100-105. doi:10.1038/s41586-020-2229-5

2. Renz, Bernhard W et al. “β2 Adrenergic-Neurotrophin Feedforward Loop Promotes Pancreatic Cancer.” Cancer cell vol. 33,1 (2018): 75-90.e7. doi:10.1016/j.ccell.2017.11.007

3. Zheng, Gen et al. “Human D-Tyr-tRNA(Tyr) deacylase contributes to the resistance of the cell to D-amino acids.” The Biochemical journal vol. 417,1 (2009): 85-94. doi:10.1042/BJ20080617

4. Kim, Hye-Jung, and Harvey Cantor. “CD4 T-cell subsets and tumor immunity: the helpful and the not-so-helpful.” Cancer immunology research vol. 2,2 (2014): 91-8. doi:10.1158/2326-6066.CIR-13-0216

5. Nagano, Taiki et al. “d-amino acid oxidase promotes cellular senescence via the production of reactive oxygen species.” Life science alliance vol. 2,1 e201800045. 18 Jan. 2019, doi:10.26508/lsa.201800045

REFERENCESDISCUSSION AND FUTURE DIRECTIONSCONCLUSIONS

RESULTS

Oral Administration of SM-88 Reduces Tumor Growth

in an HCT-116 Colon Cancer Xenograft Model

A pilot in vivo xenograft experiment was conducted to evaluate the effects of SM-88 in a colon cancer model.

HCT-116 tumor growth in mice treated with orally administered 324 mg/kg/day SM-88 was significantly reduced compared to those treated with Vehicle alone by Day 19 after treatment initiation (p < 0.05).

No differences in body weight changes were noted between treatment groups.

Number of HCT-116 Tumor-bearing Mice Alive on Each Treatment Day

Day 1 5 8 12 15 19

Vehicle 11 11 11 11 11 11

81 mg/kg 11 11 11 11 11 11

162 mg/kg 11 11 11 11 11 10

324 mg/kg 11 11 11 11 11 9

Figure 1 Figure 2a

Figure 2b

IP Administration of SM-88 Reduces Tumor Growth in a

Pan02 Pancreatic Cancer Syngenic Xenograft Model

Alterations to the Pan02 Tumor Microenvironment

Following Exposure to SM-88 (Day 34)

Exposure to SM-88 may decrease intra-tumoral Treg populations, which may reduce immunosuppressive signaling within the tumor.

Small increases in intracellular B cell populations were also observed following treatment with SM-88. N = 5 for all treatment groups.

Figure 3: Treg and B Cells

Figure 3d: Treg Figure 3e: B Cells

Figure 3: CD4, CD8 (Total Number) and CD4/CD8 Ratio

Figure 3a: CD4 Figure 3b: CD8 Figure 3c: CD4/CD8 Ratio

To evaluate potential effects of SM-88 on the tumor immune microenvironment, flow cytometry was used to characterize the immune populations present in five randomly selected Pan02 tumors collected at study termination (Day 34).

Exposure to SM-88 appears to decrease intra-tumoral CD4+ T cell populations, while preserving CD8+ populations, leading to decreases in the CD4+/CD8+ ratio.

CD4/CD8 ratio was p= 0.015 for 75 mg/kg/day vs. Vehicle (ANOVA with Tukey post hoc). N = 5 for all treatment groups.

SM-88 Induces Reactive Oxygen Species (ROS) Production

The potential of SM-88 to increase ROS production was investigated using four cell lines, two pancreatic cancer (Pan02 and PANC1) and two breast cancer (4T1 and MCF-7).

Dose dependent increases were observed in ROS production following 24 hours of exposure to SM-88 ME.

Figure 4a: Pan02 Figure 4b: PANC1

Figure 4c: MCF-7 Figure 4d: 4T1

Figure 4: ROS Positive Cells

0.00 0.25 0.50 1.00 2.500

5

10

15

20

SM-88 Concentration (mM)

Fo

ld C

han

ge in

RO

S

Po

sit

ive C

ells f

rom

Baselin

e

ROS - X-fold change in ROS+ cells from baseline:

Pan02

Pan02

0.00 0.25 0.50 1.00 2.500

2

4

6

8

SM-88 Concentration (mM)

Fo

ld C

han

ge in

RO

S

Po

sit

ive C

ells f

rom

Baselin

e

ROS - X-fold change in ROS+ cells from baseline:

MCF-7

MCF-7

0.00 0.25 0.50 1.00 2.500

10

20

30

40

SM-88 Concentration (mM)

Fo

ld C

han

ge in

RO

S

Po

sit

ive C

ells f

rom

Baselin

e

ROS - X-fold change in ROS+ cells from baseline:

4T1

4T1

0.00 0.25 0.50 1.00 2.500

1

2

3

SM-88 Concentration (mM)

Fo

ld C

han

ge in

RO

S

Po

sit

ive C

ells f

rom

Baselin

e

ROS - X-fold change in ROS+ cells from baseline:

PANC1

PANC1

0 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 190

500

1000

1500

2000

Days

Tu

mo

r V

olu

me (

mm

3)

HCT-116 xenograft size (mean ± SEM)

Vehicle

81 mg/kg

162 mg/kg

324 mg/kg

SM-88 Affects Autophagy

Figure 5a The potential of SM-88 to affect autophagy was investigated using three cell lines, two pancreatic cancer (Pan02 and PANC1) and one ovarian cancer (HeLa).

Exposure to SM-88 ME appeared to promote a dose dependent increase in LC3B expression at 24 hours in PANC1 and HeLa cells (Figure 5a).

Exposure to 1 mM SM-88 ME promoted an increase in LC3B expression beginning at least 6 hours after treatment in both Pan02 and PANC1 cells.

SM-88 ME exposure either did not affect or slightly increased p62 expression.

These data suggest that SM-88 affects autophagic mechanisms, and follow-up experiments are underway to determine whether the increases in LC3B expression are due to autophagy induction or blockade of autophagosome and lysosome fusion.

Figure 5b

HeLaPANC1Pan02

PANC1Pan02

LC3B

p62

GAPDH

LC3B

p62

GAPDH

0 7 14 21 28 35

0

100

200

300

400

500

600

700

Days of treatment

Tu

mo

r vo

lum

e (

mm

3)

Pan02 Subcutaneous Xenograft Model

Pan02 xenograft size (mean ± SEM)

Vehicle

25 mg/kg/day

75 mg/kg/day

Exploratory StatsANOVA withTukey

25 mg vs Vehicle: NS75 mg vs Vehicle: p = 0.038

Figure 3: Macrophages (Total Number) and M1/M2 Ratio

Figure 3f: M1 Figure 3g: M2 Figure 3h: M1/M2 Ratio

Exposure to SM-88 may preserve M1 polarized macrophage populations while decreasing populations of M2 polarized macrophages.

Within the tumor microenvironment, M1 macrophages have anti-tumor effects, while M2 macrophages often promote tumor growth.

Thus, alterations in the M1/M2 ratio could contribute to suppression or reversal of tumor growth. N = 5 for all treatment groups.

Vehic

le

25 m

g/kg/d

ay

75 m

g/kg/d

ay

0

5000

10000

15000

20000

To

tal n

um

ber

M1 Macrophages (total number)

Vehicle

25 mg/kg/day

75 mg/kg/day

Vehic

le

25 m

g/kg/d

ay

75 m

g/kg/d

ay

0

5000

10000

15000

20000

To

tal n

um

ber

M2 Macrophages (total number)

Vehicle

25 mg/kg/day

75 mg/kg/day

Vehic

le

25 m

g/kg/d

ay

75 m

g/kg/d

ay

0

2

4

6

M1/M

2 r

ati

o

M1/M2 Ratio

Vehicle

25 mg/kg/day

75 mg/kg/day

To further evaluate the anti-cancer effects of SM-88, a second in vivo experiment was conducted using a pancreatic cancer model.

Pan02 tumor growth in mice treated with intraperitoneally administered 75 mg/kg/day SM-88 ME was significantly reduced compared to those treated with Vehicle alone on Day 34 after treatment initiation (Figure 2a).

All mice (n=30) were alive at end of treatment (Day 34). No differences in body weight changes were noted between treatment groups. Excised Pan02 tumors (Day 34) from the subcutaneous xenograft model are shown in Figure 2b. A higher proportion of mice treated with SM-88 maintained tumors below a volume of 500mm3

(~2.5% of total body weight), with an indication of dose-related effect.

SM-88 has demonstrated encouraging efficacy in 15 different tumor types. This may be due to the effects of SM-88 on cancer through multiple mechanisms, including immune modulation, alterations in autophagy and ROS induction.

Alterations in autophagy have been associated with increased MHC1 expression in pancreatic cancer (Yamamoto et al., 2020). We are deepening our examination of the impact of SM-88 on autophagy/mitophagy, as well as any phenotypical changes that might be related to these changes including MHC1 expression.

We are continuing to explore the immune modulatory effects observed in these preliminary experiments.

Based on these and other ongoing experiments, we are beginning to explore SM-88 combinations with chemotherapies, targeted agents, or immuno-oncology therapies.

We are continuing to explore these and other effects of SM-88, as well as potential biomarkers of SM-88 sensitivity, through experiments on human organoids, and metabolomic and gene expression analyses.

The importance of amino acid metabolism in cancer has gained greater awareness over the past decade; however, potential therapeutic approaches in this area remain limited.

SM-88 has been dosed in over 180 cancer patients and has shown encouraging efficacy and safety findings to date.

Early data indicate that SM-88 may promote increases in ROS generation and alterations in autophagy in certain cancer cell types (Pan02 pancreatic cancer).

Potential immune modulatory effects of SM-88, including alterations in macrophage polarization and other key tumor immune cells (CD4+ and Treg) are being examined.

In all in vitro cell line experiments, and the Pan02 xenograft model, the SM-88 methyl-ester (SM-88 ME) was used, which has the same active moiety as SM-88, but with improved solubility characteristics. The SM-88 free acid form was used in the HCT-116 xenograft experiment.

Autophagy: Changes in LC3B and p62 expression were investigated using standard Western Blot techniques.

ROS Induction: Following treatment with SM-88, ROS induction was assessed using the CellROX (Thermo-Fisher) flow cytometry assay kit. Gating for ROS+/live cell selection was based on data obtained from negative controls.

HCT-116 Xenograft: Female athymic nude mice were implanted subcutaneously with HCT-116 cells. Once tumors reached 50-100 mm3, mice were treated with either Vehicle alone, 81 mg/kg/day SM-88, 162 mg/kg/day SM-88, or 324 mg/kg/day SM-88 administered orally (n = 11 per group).

Pan02 Xenograft Study: C57BL/6 mice were treated with either Vehicle alone, 25 mg/kg/day of SM-88 ME via intraperitoneal (IP) injection, or 75 mg/kg/day of SM-88 ME IP (n = 10 per group). Treatments began on Day 0. On Day 4, Pan02 cells were implanted subcutaneously into each mouse.

Tumor Immune Profiling: On Day 34, Pan02 tumors from five randomly selected mice per treatment arm were processed for immune analysis by flow cytometry using a 16 color myeloid and lymphocyte panel following standard protocols.