blockade of the pparα metabolic checkpoint with tpst-1120 ... · receptor alpha (pparα) is the...

1
ABSTRACT Tumors evolve to modulate metabolism to promote their own survival and to suppress tumor- specific immunity. Hypoxic conditions in the tumor microenvironment (TME) induce fatty acid oxidation (FAO), and diverse malignancies are reliant on this metabolic pathway. Additionally, suppressive immune cell populations including M2 macrophages, myeloid-derived suppressor cells and regulatory T cells preferentially utilize FAO. Peroxisome proliferator-activated receptor alpha (PPARα) is the principal transcription factor that regulates the expression of FAO genes, and this metabolic checkpoint is critical for tumor proliferation. TPST-1120 is a first-in-class selective competitive antagonist of the human PPARα. To test the hypothesis that blocking FAO with TPST-1120 confers anti-tumor efficacy, we assessed TPST-1120 in multiple syngeneic and xenograft mouse models. Blockade of PPARα with TPST-1120 mediated potent anti-tumor immune responses and significant tumor regression in syngeneic models of breast, lung, colon, pancreatic and melanoma in addition to xenograft models of CLL, AML, pancreatic and melanoma cancers as a monotherapy or in combination with chemotherapy. In pancreatic and breast cancer models, TPST-1120 augmented regression of tumor growth in combination with chemotherapy. In combination with anti-PD1, TPST-1120 treatment resulted in significant reduction of tumor growth in ovarian orthotopic (ID8) and colon (MC38) models; cured mice were completely protected against autologous tumor challenge, strongly suggesting immunological T cell memory against the primary tumor. Studies in genetic knock-out mice indicated that macrophages and antigen cross-presenting dendritic cells are required for TPST- 1120 activity, mediated through thrombospondin-1(TSP-1) and stimulator of interferon genes (STING). Consistent with prior reports, inhibition of PPARa with TPST-1120 skewed macrophages in vivo toward an M1 effector phenotype. These results provide the rationale for evaluating TPST-1120 in patients with advanced malignancies. A Phase 1/1b open-label, dose- escalation and dose-expansion study of TPST-1120 as a single agent or in combination with systemic anti-cancer therapies is planned in early 2019. METHODS TPST-1120 in vitro killing was conducted on primary CLL tumor cells Efficacy as a monotherapy or in combination with chemotherapy or anti-PD1 was evaluated in multiple syngeneic mouse models To characterize its mechanism of anti-tumor immunity, TPST-1120 was evaluated in knock-out models of TSP-1, STING and BatF3. ELISA was used to measure plasma and tumor matrix protein thrombospondin-1 (TSP-1), and FGF21 Gene expression analysis was performed by quantitative RT-PCR Flow cytometry was used to evaluate immune cell infiltrate in the tumor REFERENCES 1. Camarda R, Zhou AY, Kohnz, RA, Balakrishnan, S, Mahieu, C, et al. (2016) Nat Med, 22(4): 427-432 2. Nath A, & Chan C. (2016) Sci Rep, 6: 1-13 3. Luo X, Cheng C, Tan Z, Li N, Tang M, et al. (2017) Mol Cancer, 16(1): 76 4. Carracedo A, Cantley LC, Pandolfi PP. (2013) Nat Rev Cancer 13(4): 227-32 5. Kaipainen A, Kieran MW, Huang S, Butterfield C, Bielenberg D, et al. (2007) PLOS ONE 2(2): e260 6. Senni N, Savall M, Cabrerizo Granados D, Alves-Guerra MC, Sartor C, et al.(2018). Gut, 0: 1-3 CONCLUSIONS TPST-1120 Blocks PPARα Metabolic Checkpoint First-in-class oral PPARα antagonist Blocks transcription of PPARα target genes Shifts metabolism from FAO to glycolysis Multi-pronged mechanism: 1. Direct killing of tumor cells dependent on FAO 2. Shifting FAO to glycolysis supports immune effector cells and inhibits suppressor cells 3. Restoration of homeostatic TSP-1 levels Significant anti-tumor efficacy as single agent and in combination therapies A Phase 1/1b open-label, dose-escalation and dose-expansion study of TPST-1120 as a single agent or in combination with systemic anti-cancer therapies in subjects with advanced solid tumors initiated in Q12019 (NCT03829436) RESULTS INTRODUCTION TPST-1120 is a first-in-class, orally administered, potent, small molecule selective antagonist of the human peroxisome proliferator-activated receptor alpha (PPARα) PPARα is a transcription factor which induces the expression of genes that regulate fatty acid oxidation (FAO) and inflammation (Figure 1). PPARα and FAO gene signatures are enriched in metastatic tumors (Figure2). 1-4 Tumor growth is suppressed in PPARα-deficient mice. 5-6 TPST-1120 has significant anti-tumor activity as a monotherapy and in combination with chemotherapy or anti-PD1 Abs. Anti-tumor activity of TPST-1120 is mediated through: 1) direct killing of tumor cells dependent on FAO; 2) Shift FAO to glycolysis supports immune effector cells and inhibits suppressor cells; and 3) restore TSP-1 to homeostatic levels. FIGURE 10: TPST-1120 Combination with Anti-PD1 Induces Memory Response in MC38 Colon Model Figure 1: PPARα regulates key genes involved in FAO, metabolism and inflammation. Upon ligand binding, PPARα heterodimerizes with RXR, stimulates gene transcription of target genes involved in FAO and lipid metabolism. PPARα also negatively modulate gene transcription by inhibiting DNA-binding of several other transcription factors, such as NF-κB. FIGURE 1: PPARα Induces Fatty Acid Oxidation (FAO) FAT/CD36 FACS Genes regulate FAO and metabolism (ie. FGF21, PDK4, MT1, FAT/CD36, FCS, CPT1/2, CAT) and inflammation (ie. NFkB targets) Fattyacyl CoA Fatty acids Inside Cell Fattyacyl CoA Acetyl CoA β Oxidation Nucleus PPARɑ RXR Outside Cell aa CPT1 CPT2 CAT Fattyacyl CoA Acylcarnitine Carnitine Acetyl CoA β Oxidation Mitochondrial Acylcarnitine Carnitine Blockade of the PPARα metabolic checkpoint with TPST-1120 suppresses tumor growth and stimulates anti-tumor immunity Chan C. Whiting 1 , Nick Stock 2 , Davorka Messmer 2 , Austin Chen 2 , Lisa Rahbaek 2 , Derek Metzger 1 , Amanda Enstrom 1 , Michael Sturdivant 5 , Nicholas DeVito 5 , David Spaner 3 , Peppi Prasit 2 , Brent Hanks 5 , Dipak Panigrahy 4 , Ginna Laport 1 1 Tempest Therapeutics, Inc., San Francisco, CA; 2 Inception Sciences, San Diego, CA; 3 Sunnybrook, Toronto, Canada; 4 Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA; 5 Duke Cancer Institute, Duke University Medical Center, Durham, NC Figure 2: Enrichment of FAO associated genes including PPARα was evaluated in 10,000 unique datasets representing 33 tumor types in TCGA (cancergenome.nih.gov). FIGURE 2: Upregulation of PPARα and FAO Genes in Diverse Tumors FAO Upregulated Tumors PPAR α /FAO Gene Signature Low Expression High Expression Increasing FAO usage TPST-1120: PPARα Target Validation and Target Engagement The anti-tumor effects of PPARα antagonism is dependent upon immune modulation Figure 5: TPST-1120 directly inhibits proliferation of human primary CLL tumor cells in a dose dependent manner after 48 hours of incubation (n=14). FIGURE 5: TPST-1120 Induces Direct Tumor Cell Cytotoxicity in Primary CLL Cells Figure 6: TPST-1120 (30 mg/kg PO BID) response against PancOH7 tumors was improved by combination with gemcitabine 50 mg/kg IP, q4d (N=5/group) in PancOH7. 4/5 mice in the combo group were alive at day 210 (treatment discontinued at day 48). FIGURE 6: TPST-1120 Has Monotherapy Effect and Significantly Enhances Benefit of Chemotherapy Figure 7: A) TPST-1120 at 30 mg/kg PO BID had no effect in STING or B) Batf3 knockout mice (n=10) compared to wild type control (n=5). FIGURE 7: TPST-1120 Anti-Tumor Activity Requires Innate and Adaptive Immunity A) STING deficient/Goldenticket Model B) BATF3 Knockout Model Figure 9: A) TPST-1120 has potent anti-tumor monotherapy in WT mice but this activity was lost when PancOH7 tumors implanted in TSP-1 knock-out mice. B.) Anti-tumor efficacy is associated with increase in TSP-1 levels in the TME FIGURE 9: TPST-1120 Anti-Tumor Efficacy is Dependent on TSP-1 A.) Deletion of TSP1 gene ablates therapeutic anti-tumor efficacy of TPST-1120 in PancOH7 model B.) TPST-1120 induces an increase in tumor TSP-1 Figure 10: A.)Established MC38 tumors (100-150mm3) treated with vehicle (n=10), TPST-1120 (n=7), anti-PD1 alone (n=10), or TPST-1120 + anti-PD1 (n=15) were followed for tumor growth. By day 24 only animals in the combination group remained alive. Three were tumor free at day 60 post removal of therapy and were tumor-free upon autologous tumor re-challenge while aggressive tumor growth occurred in naïve mice. B.) Adoptive transfer of splenocytes from naïve C57BL/6 mice or MC38 tumor bearing mice cured with TPST + αPD-1 into naïve recipient C57BL/6 mice, followed by challenge with 1 x 10 6 MC38 tumor cells. Mice were then monitored for tumor growth. Tumor growth was significantly delayed in mice receiving splenocytes from cured mice compared to those from naïve mice. Splenocytes from naïve mice 0 200 400 600 800 1000 1200 1400 1600 1800 2000 0 5 10 15 20 25 30 35 40 45 50 Tumor Volume (mm 3 ) Days Post-Splenocyte Transfer Splenocytes from cured mice B.) Transferrable anti-tumor immunity from cured mice treated with TPST-1120 and α-PD1 Source: Kaipainen et al., PLoS ONE 2007, 2(2): e260 and unpublished results Figure 3: Tumor growth is inhibited in PPARα knock-out (KO) mice. Tumors establish but spontaneously regress in KO mice. FIGURE 3: Spontaneous Tumor Regression in the Absence of PPARα Sarcoma (SV40 & H-ras transformed mouse embryo fibroblast tumor cell line) Tumor Establishment but Not Progression Deletion of PPARα in host and tumor cells prevents usage of fatty acid oxidation (FAO) Observation repeated in melanoma and lung tumor models Tumor from PPARα KO contains lipid droplets (white) prior to regression 0 200 400 600 0 15 25 35 45 55 65 75 0 2000 4000 6000 8000 10000 12000 14000 16000 0 15 25 35 45 55 65 75 400 PPARα Wild Type (n=9) Tumor Volume (mm 3) PPARα KO (n = 8) Days Post Implantation Tumor Volume (mm 3) Large-scale Y axis Small-scale Y axis Days Post Implantation Tumor section (H&E) A.) MC38 colon model TPST-1120 + anti-PD1 therapy induces cures in MC38 tumor-bearing mice Re-challenge cured mice Complete protection against autologous tumor challenge in cured mice 2 months off TPST-1120 Tx Figure 8: A) TPST-1120 has potent anti-tumor efficacy when combined with anti-PD1 in syngeneic BRAF V600E PTEN -/- model. B.) Combination TPST-1120 plus anti-PD1 Ab shows anti-tumor efficacy is associated with synergistic increase in T cell infiltration into the TME. FIGURE 8: TPST-1120 Combination with anti-PD1 mAb Induces Significant Anti-Tumor Response and Tumor CD8 T cell Infiltration A.) Significant anti-tumor efficacy of TPST-1120 when combined with anti-PD1 B.) Synergistic Tumor CD8+ T-cell Infiltrate of TPST-1120 with anti-PD1 TPST-1120 PPARα LIPID METABOLISM / FATTY ACID OXIDATION Supports effector immune cells Tumor cell death Transcription blocked Figure 4: A.) PPARα induced Fgf21 protein and target gene Pdk4 increases under fasting is inhibited with TPST-1120 at 1, 10, and 100mg/kg PO BID in C57BL6/N mice (non-tumor bearing). B.) Pdk4 gene expression is inhibited in B16-F10 melanoma tumors in mice treated with TPST-1120 30 mg/kg PO BID compared to vehicle treated. FIGURE 4: PPARα Antagonist TPST-1120 Inhibits PPARα-Dependent Targets B) Tumor Pdk4 in Tumor Bearing Mice A) TPST-1120 inhibits PPARα mediated plasma Fgf21 protein and blood Pdk4 gene

Upload: others

Post on 24-Jun-2020

4 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Blockade of the PPARα metabolic checkpoint with TPST-1120 ... · receptor alpha (PPARα) is the principal transcription factor that regulates the expression of FAO genes, and this

ABSTRACT

Tumors evolve to modulate metabolism to promote their own survival and to suppress tumor-specific immunity. Hypoxic conditions in the tumor microenvironment (TME) induce fatty acid oxidation (FAO), and diverse malignancies are reliant on this metabolic pathway. Additionally, suppressive immune cell populations including M2 macrophages, myeloid-derived suppressor cells and regulatory T cells preferentially utilize FAO. Peroxisome proliferator-activated receptor alpha (PPARα) is the principal transcription factor that regulates the expression of FAO genes, and this metabolic checkpoint is critical for tumor proliferation. TPST-1120 is a first-in-class selective competitive antagonist of the human PPARα. To test the hypothesis that blocking FAO with TPST-1120 confers anti-tumor efficacy, we assessed TPST-1120 in multiple syngeneic and xenograft mouse models. Blockade of PPARα with TPST-1120 mediated potent anti-tumor immune responses and significant tumor regression in syngeneic models of breast, lung, colon, pancreatic and melanoma in addition to xenograft models of CLL, AML, pancreatic and melanoma cancers as a monotherapy or in combination with chemotherapy. In pancreatic and breast cancer models, TPST-1120 augmented regression of tumor growth in combination with chemotherapy. In combination with anti-PD1, TPST-1120 treatment resulted in significant reduction of tumor growth in ovarian orthotopic (ID8) and colon (MC38) models; cured mice were completely protected against autologous tumor challenge, strongly suggesting immunological T cell memory against the primary tumor. Studies in genetic knock-out mice indicated that macrophages and antigen cross-presenting dendritic cells are required for TPST-1120 activity, mediated through thrombospondin-1(TSP-1) and stimulator of interferon genes (STING). Consistent with prior reports, inhibition of PPARa with TPST-1120 skewed macrophages in vivo toward an M1 effector phenotype. These results provide the rationale for evaluating TPST-1120 in patients with advanced malignancies. A Phase 1/1b open-label, dose-escalation and dose-expansion study of TPST-1120 as a single agent or in combination with systemic anti-cancer therapies is planned in early 2019.

METHODS

�� TPST-1120 in vitro killing was conducted on primary CLL tumor cells �� Efficacy as a monotherapy or in combination with chemotherapy or anti-PD1 was

evaluated in multiple syngeneic mouse models �� To characterize its mechanism of anti-tumor immunity, TPST-1120 was evaluated in

knock-out models of TSP-1, STING and BatF3. �� ELISA was used to measure plasma and tumor matrix protein thrombospondin-1 (TSP-1),

and FGF21�� Gene expression analysis was performed by quantitative RT-PCR�� Flow cytometry was used to evaluate immune cell infiltrate in the tumor

REFERENCES

1. Camarda R, Zhou AY, Kohnz, RA, Balakrishnan, S, Mahieu, C, et al. (2016) Nat Med, 22(4): 427-432

2. Nath A, & Chan C. (2016) Sci Rep, 6: 1-133. Luo X, Cheng C, Tan Z, Li N, Tang M, et al. (2017) Mol Cancer, 16(1): 764. Carracedo A, Cantley LC, Pandolfi PP. (2013) Nat Rev Cancer 13(4): 227-325. Kaipainen A, Kieran MW, Huang S, Butterfield C, Bielenberg D, et al. (2007) PLOS ONE 2(2): e2606. Senni N, Savall M, Cabrerizo Granados D, Alves-Guerra MC, Sartor C, et al.(2018). Gut, 0: 1-3

CONCLUSIONS

TPST-1120 Blocks PPARα Metabolic CheckpointFirst-in-class oral PPARα antagonist �� Blocks transcription of PPARα target genes�� Shifts metabolism from FAO to glycolysis �� Multi-pronged mechanism:

1. Direct killing of tumor cells dependent on FAO2. Shifting FAO to glycolysis supports immune

effector cells and inhibits suppressor cells3. Restoration of homeostatic TSP-1 levels

�� Significant anti-tumor efficacy as single agent and in combination therapies�� A Phase 1/1b open-label, dose-escalation and

dose-expansion study of TPST-1120 as a single agent or in combination with systemic anti-cancer therapies in subjects with advanced solid tumors initiated in Q12019 (NCT03829436)

RESULTS

INTRODUCTION

�� TPST-1120 is a first-in-class, orally administered, potent, small molecule selective antagonist of the human peroxisome proliferator-activated receptor alpha (PPARα) �� PPARα is a transcription factor which induces the expression of genes that regulate fatty

acid oxidation (FAO) and inflammation (Figure 1).�� PPARα and FAO gene signatures are enriched in metastatic tumors (Figure2).1-4

�� Tumor growth is suppressed in PPARα-deficient mice.5-6 �� TPST-1120 has significant anti-tumor activity as a monotherapy and in combination with

chemotherapy or anti-PD1 Abs.�� Anti-tumor activity of TPST-1120 is mediated through: 1) direct killing of tumor cells

dependent on FAO; 2) Shift FAO to glycolysis supports immune effector cells and inhibits suppressor cells; and 3) restore TSP-1 to homeostatic levels.

FIGURE 10: TPST-1120 Combination with Anti-PD1 Induces Memory Response in MC38 Colon Model

Figure 1: PPARα regulates key genes involved in FAO, metabolism and inflammation. Upon ligand binding, PPARα heterodimerizes with RXR, stimulates gene transcription of target genes involved in FAO and lipid metabolism. PPARα also negatively modulate gene transcription by inhibiting DNA-binding of several other transcription factors, such as NF-κB.

FIGURE 1: PPARα Induces Fatty Acid Oxidation (FAO)

FAT/CD36

FACS

Genes regulate FAO and metabolism (ie. FGF21, PDK4, MT1, FAT/CD36, FCS, CPT1/2, CAT) and inflammation (ie. NFkB targets)

Fattyacyl CoA

Fatty acids

Inside Cell

Fattyacyl CoA

Acetyl CoA

β Oxidation

Nucleus

PPARɑ RXR

Outside Cell

aa

CPT1

CPT2

CAT

Fattyacyl CoA Acylcarnitine Carnitine

Acetyl CoA

β Oxidation

Mitochondrial

Acylcarnitine Carnitine

Blockade of the PPARα metabolic checkpoint with TPST-1120 suppresses tumor growth and stimulates anti-tumor immunity Chan C. Whiting1, Nick Stock2, Davorka Messmer2, Austin Chen2, Lisa Rahbaek2, Derek Metzger1, Amanda Enstrom1, Michael Sturdivant5, Nicholas DeVito5, David Spaner3, Peppi Prasit2, Brent Hanks5, Dipak Panigrahy4, Ginna Laport1

1Tempest Therapeutics, Inc., San Francisco, CA; 2Inception Sciences, San Diego, CA; 3Sunnybrook, Toronto, Canada; 4Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA; 5Duke Cancer Institute, Duke University Medical Center, Durham, NC

Figure 2: Enrichment of FAO associated genes including PPARα was evaluated in 10,000 unique datasets representing 33 tumor types in TCGA (cancergenome.nih.gov).

FIGURE 2: Upregulation of PPARα and FAO Genes in Diverse Tumors

FAO Upregulated Tumors

PPARα/FAO Gene Signature

Low Expression

High Expression

Increasing FAO usage

TPST-1120: PPARα Target Validation and Target Engagement

The anti-tumor effects of PPARα antagonism is dependent upon immune modulation

Figure 5: TPST-1120 directly inhibits proliferation of human primary CLL tumor cells in a dose dependent manner after 48 hours of incubation (n=14).

FIGURE 5: TPST-1120 Induces Direct Tumor Cell Cytotoxicity in Primary CLL Cells

Figure 6: TPST-1120 (30 mg/kg PO BID) response against PancOH7 tumors was improved by combination with gemcitabine 50 mg/kg IP, q4d (N=5/group) in PancOH7. 4/5 mice in the combo group were alive at day 210 (treatment discontinued at day 48).

FIGURE 6: TPST-1120 Has Monotherapy Effect and Significantly Enhances Benefit of Chemotherapy

Figure 7: A) TPST-1120 at 30 mg/kg PO BID had no effect in STING or B) Batf3 knockout mice (n=10) compared to wild type control (n=5).

FIGURE 7: TPST-1120 Anti-Tumor Activity Requires Innate and Adaptive Immunity

A) STING deficient/Goldenticket Model B) BATF3 Knockout Model

Figure 9: A) TPST-1120 has potent anti-tumor monotherapy in WT mice but this activity was lost when PancOH7 tumors implanted in TSP-1 knock-out mice. B.) Anti-tumor efficacy is associated with increase in TSP-1 levels in the TME

FIGURE 9: TPST-1120 Anti-Tumor Efficacy is Dependent on TSP-1

PancOH7 model

A.) Deletion of TSP1 gene ablates therapeutic anti-tumor efficacy of TPST-1120 in PancOH7 model B.) TPST-1120 induces an increase in tumor TSP-1

Figure 10: A.)Established MC38 tumors (100-150mm3) treated with vehicle (n=10), TPST-1120 (n=7), anti-PD1 alone (n=10), or TPST-1120 + anti-PD1 (n=15) were followed for tumor growth. By day 24 only animals in the combination group remained alive. Three were tumor free at day 60 post removal of therapy and were tumor-free upon autologous tumor re-challenge while aggressive tumor growth occurred in naïve mice. B.) Adoptive transfer of splenocytes from naïve C57BL/6 mice or MC38 tumor bearing mice cured with TPST + αPD-1 into naïve recipient C57BL/6 mice, followed by challenge with 1 x 106 MC38 tumor cells. Mice were then monitored for tumor growth. Tumor growth was significantly delayed in mice receiving splenocytes from cured mice compared to those from naïve mice.

TPST-1120 + α-PD1 Induces Effective Immune Memory

Splenocytes from naïve mice

0

200

400

600

800

1000

1200

1400

1600

1800

2000

0 5 10 15 20 25 30 35 40 45 50

Tum

or V

olum

e (m

m3 )

Days Post-Splenocyte Transfer

Splenocytes from cured mice

B.) Transferrable anti-tumor immunity from cured mice treated with TPST-1120 and α-PD1

Source: Kaipainen et al., PLoS ONE 2007, 2(2): e260 and unpublished resultsFigure 3: Tumor growth is inhibited in PPARα knock-out (KO) mice. Tumors establish but spontaneously regress in KO mice.

FIGURE 3: Spontaneous Tumor Regression in the Absence of PPARα

Sarcoma (SV40 & H-ras transformed mouse embryo fibroblast tumor cell line)

Tumor Establishment but Not Progression

Deletion of PPARα in host and tumor cells prevents usage of fatty acid oxidation (FAO)

Observation repeated in melanoma and lung tumor models

Tumor from PPARα KO contains lipid droplets (white) prior to

regression 0

200

400

600

0 15 25 35 45 55 65 75 0

2000

4000

6000

8000

10000

12000

14000

16000

0 15 25 35 45 55 65 75 400

PPARα Wild Type (n=9) Tu

mor

Vol

ume

(mm

3)

PPARα KO (n = 8)

Days Post Implantation

Tum

or V

olum

e (m

m3)

Large-scale Y axis Small-scale Y axis

Days Post Implantation

Tumor section (H&E)

A.) MC38 colon model

TPST-1120 + anti-PD1 therapy induces cures in MC38 tumor-bearing mice

Re-challenge cured mice

Complete protection against autologous tumor challenge in cured mice

2 months off TPST-1120 Tx

Figure 8: A) TPST-1120 has potent anti-tumor efficacy when combined with anti-PD1 in syngeneic BRAFV600EPTEN-/- model. B.) Combination TPST-1120 plus anti-PD1 Ab shows anti-tumor efficacy is associated with synergistic increase in T cell infiltration into the TME.

FIGURE 8: TPST-1120 Combination with anti-PD1 mAb Induces Significant Anti-Tumor Response and Tumor CD8 T cell Infiltration

A.) Significant anti-tumor efficacy of TPST-1120 when combined with anti-PD1

B.) Synergistic Tumor CD8+ T-cell Infiltrate of TPST-1120 with anti-PD1

TPST-1120 PPARα

LIPID METABOLISM / FATTY ACID OXIDATION

Supports effector

immune cells

Tumor cell death

Transcription blocked

Figure 4: A.) PPARα induced Fgf21 protein and target gene Pdk4 increases under fasting is inhibited with TPST-1120 at 1, 10, and 100mg/kg PO BID in C57BL6/N mice (non-tumor bearing). B.) Pdk4 gene expression is inhibited in B16-F10 melanoma tumors in mice treated with TPST-1120 30 mg/kg PO BID compared to vehicle treated.

FIGURE 4: PPARα Antagonist TPST-1120 Inhibits PPARα-Dependent Targets

B) Tumor Pdk4 in Tumor Bearing Mice A) TPST-1120 inhibits PPARα mediated plasma Fgf21 protein and blood Pdk4 gene