Gastrointestinal/liver-targeted TLR7 agonist for treatment ...

1
v Drug Design: using medchem principles for liver-specific drugs (e.g. statins) v In vitro: selective on TLR7 (pDC) over TLR8 (monocyte); active across species v PK: pulsatile kinetics, high exposure in GI and liver, low peripheral tissue distribution v PD: robust IP-10 and ISG response (desired), with minimal TNFa, IL-6, IL-10 (detrimental) v Efficacy: efficacious in orthotopic models of colorectal and liver cancer (CT26, Hepa1.6, 4T1) as single agent and/or in combination with anti-PD1/L1 v MOA: increased activated CD103 + DC frequency in DLN and tumor-specific CD8 + TILs v Tox: no major toxicity identified in relevant species v Clinical Applications: GI malignancies with liver metastases and potentially other cancers with liver metastases v Product Differentiation: potential wider therapeutic window compared to other oral TLR7 agonists or TLR7 agonist administered systemically v References: 1) Science 2018, 362, 6411 ; 2) Cancer Discov. 2018, 8, 1250-1257; 3) Curr Top Med Chem 2013, 13, 857-866; 4) Cell Reports 2018, 25, 3074-3085; 5) Trends Immunol. 2016, 37, 855-865; 6) PNAS 1996, 93, 9730-9735 For more information please email: [email protected] 0 1 2 3 4 5 6 7 8 DMSO 0.4 3.7 33 [Compound] (µM) %IFNa-positive pDCs APR003 R848 Apros Therapeutics, San Diego, CA Andrew T. Miller, Evelyn Rodrigo, Manny Corpuz, David Plouffe, Tom Y.-H. Wu Gastrointestinal/liver-targeted TLR7 agonist for treatment of colorectal and liver cancers 1. Introduction Checkpoint inhibitors (PD-1/PD-L1 antibodies) have become the backbone of IO therapies, however, only a fraction of patients respond Current data suggest response rates are influenced by a myriad of mechanisms affecting the tumor microenvironment, including mutational burden and level of immune infiltrate (ref 1) Innate immune agonists have the capacity to convert “cold” immune quiescent tumors into “hot” infiltrated tumors, thus providing a complementary mechanism to checkpoint blockade 2. Toll-Like Receptor 7 Expression: Human TLR7 expressed mainly in plasmacytoid dendritic cells (professional APC) Function: TLR7 activation elicits Type I interferon, upregulates costimulatory molecule (CD86), increases antigen processing/presentation (MHC), and drives greater T cell stimulation Druggability: One of the few innate immune receptors that can be activated by small molecule, which allows for fine tuning of ADME/PK properties using proven medchem design principles Examples: Intratumoral TLR7 agonists have demonstrated pre-clinical and early clinical efficacy in solid tumors (NKTR- 262, MEDI-9197, LHC165) 3. APR003 is a GI/Liver-Targeted Oral TLR7 agonist Systemic administration leads to poor tolerability and potential immune tolerance induction Intratumoral administration affords local immune priming of tumor antigens leading to systemic (abscopal) immunity, but application limited to cutaneous accessible tumors Oral GI/liver targeted TLR7 agonist can circumvent the limitations of both systemic and intratumoral approaches to treat colorectal and liver cancers, and potentially other cancers with liver metastases Inflammation Systemic Intratumoral Oral GI/liver- targeted Dowling, ImmunoHorizons 2018, 2, 185-197 INNATE ADAPTIVE 4. GI/Liver Targeting Through OATP 0.5μM APR003 5μM APR003 5μM APR003 + BSP 0.5μM APR003 5μM APR003 5μM APR003 + Rifampicin OATP1A2 (enterocytes) OATP1B1 (hepatocytes) 5. In vitro Activity HEK-TLR7 assay 0.001 0.01 0.1 1 10 100 0 20 40 60 80 100 Conc. (μM) % MaxAct R848 APR003 GS-9620 0.001 0.01 0.1 1 10 100 0 20 40 60 80 100 Conc. (μM) % MaxAct APR003 GS-9620 R848 HEK-TLR8 assay pDC IFNa assay hPBMC IP-10 assay 0.01 0.1 1 0 50 100 150 uM % of A001 Max APR003 R848 GS-9620 Effective concentration at 50% of maximal response (EC 50 ) are shown for sigmoidal dose response curves. Lowest effective concentration (LEC), defined as the lowest concentration of test compound that elicited at least 3-fold signal over vehicle control, are shown for bell-shaped dose response curves. 6. Pharmacokinetics 7. Pharmacodynamics 0 20 40 60 80 100 120 1 10 Fold over vehicle Time (min) Primary Response (30mpk; PO): IFNb expression (<2hrs) Small Intestine Mesenteric LN Liver Large Intestine Secondary Response (30mpk; PO): 0 20 40 60 80 100 120 1 10 100 1000 Fold over vehicle Time (min) IP-10 (<2hrs) Small Intestine Mesenteric LN Liver Large Intestine 0.1 1 10 100 1000 1 10 100 100 1000 10000 ISG15 (fold over vehicle) ISG15 (liver) IP-10 (serum) [IP-10] pg/mL Dose-dependency (5hrs) Secondary Response: [APR003] mg/kg Tissue Distribution in mice dose PO (1hr) Plasma Heart Liver Spleen Lung Kidney Brain Stomach Small Intestine Large Intestine Testicle Ovary Fat Skin Skeletal 1 10 100 1000 10000 100000 Conc (ng/mL) Pulsatile kinetics (ideal for in situ vaccination): High clearance, short elimination half-life, and low volume of distribution Tissue-restricted distribution: concentrates in the GI, liver, and kidney (consistent with transporter profile) Primary response (transient): IFNb expression elicited in GI, liver, mesenteric lymph node Secondary response (amplified): interferon stimulated genes (ISG) in the target tissues and interferon-inducible protein 10 (IP-10) in serum Dose dependency: no bell-shape response observed over 1000-fold range 8. IP-10 Induction Over TNFa 0.1 1 10 100 10 100 1000 10000 0 100 200 300 400 [compound] mpk [IP10] pg/mL [TNFa] pg/mL PD dose response Body weight change after a single high dose 0 1 2 3 4 5 6 7 -15 -10 -5 0 5 10 % change in body weight Vehicle GS-9620 (150mpk) Days APR003 (300mpk) GS-9620 (IP-10) APR003 (TNFa) GS-9620 (TNFa) APR003 (IP-10) APR003 induced stronger IP-10 response over TNFa compared to GS-9620 (oral TLR7 agonist, Phase 2 for HBV) GS-9620 caused more body weight loss at doses that produced similar IP-10 levels as APR003 9. Orthotopic Syngeneic Liver Tumor Model 10. CT26 Liver Tumor Model 11. CT26 Liver Tumor Model (Dose Response) 12. CT26 Colorectal Tumor Model 13. Local Immune Priming (CD103 dendritic cells) 14. Increased Tumor-Infiltrating CD8 T Cells 15. Tissue-Specific vs Systemic Administration 16. Summary Bioluminescence imaging allows non-invasive monitoring of tumor growth on internal organs Cross-validation by gross examination, liver weights, and survival 0 10 20 30 4 6 8 10 Bioluminesence Log10 (Flux p/s) 0 10 20 30 4 6 8 10 0 10 20 30 4 6 8 10 0 10 20 30 4 6 8 10 Vehicle Anti-PDL1 APR003 0 10 20 30 40 50 60 0 20 40 60 80 100 Day % survival 0 5 10 15 20 25 30 35 6 8 10 Mean bioluminesence Log10 (ph/s) ± SEM * Days post-implant *** randomization and start of treatment treatments Tumor Burden Survival SC tumor rechallenge of survivors APR003 + Anti-PDL1 Vehicle aPDL1 APR003 APR003 + aPDL1 Median Survival Time 41 days 43 days 52.5 days 58.5 days ** *** Tumor Burden 0 5 10 15 20 25 0 500 1000 Tumor volume (mm3) Age-matched Control aPDL1 APR003 APR003 + anti-PDL1 Day post-subQ rechallenge P-value * = <0.05 ** = <0.01 *** = <0.001 In an orthotopic CT26 liver cancer model, APR003 (30 mg/kg) decreased tumor burden and increased survival as a single agent and effects were augmented in combo with anti-PDL1 Survivors resistant to subcutaneous tumor re-challenge, suggesting adaptive systemic immunity Re-challenge survivors subQ 0 7 14 21 35 68 Day Dose 12 18 25 33 Vehicle APR003 APR003 + Anti-PDL1 Day Liver weights 28 Anti-PDL1 4 Image In an orthotopic CT26 colon cancer model, APR003 (30 mg/kg) decreased tumor burden and increased survival as single agent and effects were augmented in combo with anti-PDL1 APR003 induced a greater frequency of activated CD103+ dendritic cells in liver draining lymph nodes, but not in distal non-draining lymph nodes CD8 B220 CD3 5.6 9.7 8 13 CD8 + CD3 + B220 - AH1-dextramer Vehicle Anti-PDL1 APR003 APR003 + anti-PDL1 Combination treatment of APR003 on top of anti-PD-L1 further increased tumor-specific (AH1+) CD8+ T cells in the liver tumor * n.s. Tumor Burden 0 10 20 30 40 6 8 10 Day post implant Mean bioluminesence Log10 (Flux p/s) treatments randomization and start of treatment Vehicle control Anti-PDL1 APR003 (SubQ) + aPDL1 APR003 (oral) + aPDL1 Negative control Median Survival (d) 42 45 >98 * n.s. 80 Vehicle control Anti-PDL1 APR003 (oral) + aPDL1 APR003 (subQ) + aPDL1 100 1000 10000 [IP10] pg/mL IP-10 Vehicle control Anti-PDL1 APR003 (oral) + aPDL1 APR003 (subQ) + aPDL1 100 1000 [IL-6] pg/mL IL-6 Vehicle control Anti-PDL1 APR003 (oral) + aPDL1 APR003 (subQ) + aPDL1 10 100 [TNFa] pg/mL TNFa Cytokine profile (5hr post 5 th dose) Systemic administration (via subcutaneous injection) of APR003 is less efficacious compared to tissue-specific oral administration at comparable liver exposure However, despite similar levels of IP-10 induction, systemic administration (higher serum exposure) induced more serum IL-6 and TNFa, which were not essential for anti-tumor efficacy and can lead to poor tolerability Therefore, GI/liver-targeted oral administration is a more optimal way to deliver TLR7 agonist to colorectal and liver cancers over systemic administration 10 20 30 40 5 6 7 8 9 Day post implant Bioluminesence Log10 (Flux p/s) 10 20 30 40 5 6 7 8 9 Day post implant Bioluminesence Log10 (Flux p/s) 10 20 30 40 5 6 7 8 9 Day post implant Bioluminesence Log10 (Flux p/s) 10 20 30 40 5 6 7 8 9 Day post implant Bioluminesence Log10 (Flux p/s) 0 10 20 30 40 6 7 8 Day post implant Bioluminesence Log10 (Flux p/s) + SEM enrollment Tumor Burden APR003 + Anti-PDL1 Vehicle Anti-PDL1 APR003 ** * **** 0 10 20 30 40 50 60 70 0 20 40 60 80 100 Day post implant % survival Survival Vehicle anti-PDL1 APR003 APR003 + aPDL1 Median Survival Time 26.5 days 41 days 36.5 days 59.5 days * **** *** ** P-value * = <0.05 ** = <0.01 *** = <0.001 **** = < 0.0001 * *** **** *** Efficacy in Dose Response (D29) 0 10 20 30 6 8 10 Mean bioluminesence Log10 (Flux p/s) +/- SEM Vehicle anti-PDL1 A221 (1mpk) + anti-PDL1 A221 (3mpk) + anti-PDL1 A221 (10mpk) + anti-PDL1 A221 (30mpk) + anti-PDL1 A221 (100mpk) + anti-PDL1 A221 (300mpk) + anti-PDL1 Negative control randomization and start of treatment Days post implant Tumor Burden Vehicle Anti - PDL1 1 3 10 30 100 300 A221 [mpk] + Anti-PDL1 - ctrl e 1 1 1 1 1 1 1 l 5 6 7 8 9 10 Mean bioluminesence Log10 (Flux p/s) +/- SEM P-value * = <0.05 ** = <0.01 *** = <0.001 **** = <0.0001 APR003 exhibited anti-tumor efficacy over a wide dose range (10 – 300 mg/kg) in combination with anti-PDL1 BSP and rifampicin are inhibitors of OATP1A2 and OATP1B1, respectively Cellular Assay R848 GS9620 APR003 TLR7 EC 50 (µM) 0.14 0.21 0.23 TLR8 EC 50 (µM) 2 >30 4.4 THP1 (TLR2/4/8) EC 50 (µM) 9.7 >30 >30 hu PBMC IP-10 LEC (µM) 0.05 0.05 0.14 mk PBMC IP-10 LEC (µM) 0.14 0.14 0.41 ms Spleno IP-10 LEC (µM) <0.02 0.14 <0.02 AACR 2020 Poster #684 Target selectivity: APR003 is >10-fold selective for TLR7 over TLR8 Cellular activity: Active in plasmacytoid dendritic cells, but not in THP-1 monocytes Species cross reactivity: Active across mouse, monkey, and human cells TLR7 activation Type I IFN (transient) IP-10, ISG (amplified) General Protocol : On Day 0, CT26-luc cells were surgically implanted into the right liver lobe of Balb/c mice. On Day 4, mice were imaged, randomized, and dosed twice-weekly for 4 weeks (APR003 and/or anti-PDL1). Tumor burden were monitored by bioluminescence imaging, and the statistical significance was calculated by 2-way ANOVA with multi-group comparison. Animal survival was tracked, and the statistical significance was calculated by log-rank test of Kaplan-Meier curves. On Day 0, CT26-luc were surgically implanted into the right liver lobe of Balb/c mice. On Day 11, mice were imaged, randomized, and dosed on Day 11, 15, and 18, and then sacrificed on Day 19 to assess the frequency of activated CD103+ dendritic cells (cDC1) in the liver draining LNs (celiac, portal, and first mesenteric) versus non-draining LNs (axillary/brachial) by staining for MHCII and CD86. Liver draining lymph nodes Axillary/Brachial lymph nodes CD45 MHC II Live/singlet gated CD103 CD86 Liver Tumor draining LNs Vehicle APR003 e 1 1 o e 1 1 o 0.0 0.5 1.0 1.5 2.0 %CD45+ MHCIIhi CD103+ CD86+ Frequency of activated cDC1 * * CD103 + dendritic cells (cDC1s) are known to transport antigens to the lymph nodes and cross-present to tumor-specific CD8 + T cells (ref 5) On Day 0, CT26-luc were surgically implanted into the right liver lobe of Balb/c mice. Following 3 therapeutic doses, liver tumors were removed, and single cell suspensions were stained and analyzed by flow cytometry to assess TILs. On Day 0, CT26-luc cells were surgically implanted into cecum of Balb/c mice. On Day 3, mice are imaged, randomized, and dosed twice- weekly for 4 weeks (APR003 and/or anti-PDL1). On Day 0, CT26-luc cells were surgically implanted into the right liver lobe of Balb/c mice. On Day 4, mice were imaged, randomized, and dosed twice-weekly for 4 weeks with APR003 (either PO or SC) and anti-PDL1. AH1 is a known antigen in the CT26 tumor cells (ref 6) As clinical proof-of-concept, intratumoral TLR9 agonists have shown promising anti-tumor responses in combination with checkpoint inhibitors; however, successful clinical applications have been limited to cutaneous-accessible tumor types (ref 2) APR003 was designed using medicinal chemistry principles of liver-targeting drugs, including transporter uptake properties (ref 3) Route PK Parameters Mouse Monkey IV Dose (mg/kg) 3 1 t 1/2 (h) 2.92 1.38 CL (mL/min/kg) 99.6 21.6 V dss (L/kg) 1.88 0.688 Oral Dose (mg/kg) 10 5 T max (h) 0.25 0.58 C max (ng/mL) 94.2 104 AUC 0–t (ng·h/mL) 76.5 178 F (%) 4.6 4.2 PK Liver Serum AUC (ng*h/mL) PO (30mpk) 5000 310 SC (100μg) 4400 1800 TNFa is not required for T cell mediated anti-tumor efficacy and is associated with poor tolerability at high levels (ref 4) LOCAL IMMUNE PRIMING LEADS TO SYSTEMIC ANTI-TUMOR IMMUNITY http://www.aprostx.com/

Transcript of Gastrointestinal/liver-targeted TLR7 agonist for treatment ...

RESEARCH POSTER PRESENTATION TEMPLATE © 2019

www.PosterPresentations.com

v Drug Design: using medchem principles for liver-specific drugs (e.g. statins)v In vitro: selective on TLR7 (pDC) over TLR8 (monocyte); active across speciesv PK: pulsatile kinetics, high exposure in GI and liver, low peripheral tissue distributionv PD: robust IP-10 and ISG response (desired), with minimal TNFa, IL-6, IL-10 (detrimental)v Efficacy: efficacious in orthotopic models of colorectal and liver cancer (CT26, Hepa1.6,

4T1) as single agent and/or in combination with anti-PD1/L1v MOA: increased activated CD103+ DC frequency in DLN and tumor-specific CD8+ TILsv Tox: no major toxicity identified in relevant speciesv Clinical Applications: GI malignancies with liver metastases and potentially other cancers

with liver metastasesv Product Differentiation: potential wider therapeutic window compared to other oral TLR7

agonists or TLR7 agonist administered systemicallyv References: 1) Science 2018, 362, 6411 ; 2) Cancer Discov. 2018, 8, 1250-1257; 3) Curr

Top Med Chem 2013, 13, 857-866; 4) Cell Reports 2018, 25, 3074-3085; 5) Trends Immunol. 2016, 37, 855-865; 6) PNAS 1996, 93, 9730-9735

For more information please email: [email protected]

012345678

DMSO 0.4 3.7 33

[Compound] (µM)

%IF

Na-

posi

tive

pDC

s APR003R848

Apros Therapeutics, San Diego, CA

Andrew T. Miller, Evelyn Rodrigo, Manny Corpuz, David Plouffe, Tom Y.-H. Wu

Gastrointestinal/liver-targeted TLR7 agonist for treatment of colorectal and liver cancers

1. Introduction• Checkpoint inhibitors (PD-1/PD-L1

antibodies) have become the backbone of IO therapies, however, only a fraction of patients respond

• Current data suggest response rates are influenced by a myriad of mechanisms affecting the tumor microenvironment, including mutational burden and level of immune infiltrate (ref 1)

• Innate immune agonists have the capacity to convert “cold” immune quiescent tumors into “hot” infiltrated tumors, thus providing a complementary mechanism to checkpoint blockade

2. Toll-Like Receptor 7

• Expression: Human TLR7 expressed mainly in plasmacytoid dendritic cells (professional APC)

• Function: TLR7 activation elicits Type I interferon, upregulates costimulatory molecule (CD86), increases antigen processing/presentation (MHC), and drives greater T cell stimulation

• Druggability: One of the few innate immune receptors that can be activated by small molecule, which allows for fine tuning of ADME/PK properties using proven medchem design principles

• Examples: Intratumoral TLR7 agonists have demonstrated pre-clinical and early clinical efficacy in solid tumors (NKTR-262, MEDI-9197, LHC165)

3. APR003 is a GI/Liver-Targeted Oral TLR7 agonist

• Systemic administration leads to poor tolerability and potential immune tolerance induction• Intratumoral administration affords local immune priming of tumor antigens leading to

systemic (abscopal) immunity, but application limited to cutaneous accessible tumors• Oral GI/liver targeted TLR7 agonist can circumvent the limitations of both systemic and

intratumoral approaches to treat colorectal and liver cancers, and potentially other cancers with liver metastases

InflammationSystemic Intratumoral Oral GI/liver-

targeted

Dowling, ImmunoHorizons 2018, 2, 185-197

INNATE ADAPTIVE

4. GI/Liver Targeting Through OATP

0.5µMAPR003

5µMAPR003

5µMAPR003 + BSP

0.5µMAPR003

5µMAPR003

5µMAPR003

+ Rifampicin

OATP1A2(enterocytes)

OATP1B1(hepatocytes)

5. In vitro ActivityHEK-TLR7 assay

0.001 0.01 0.1 1 10 1000

20

40

60

80

100

Conc. (µM)

% M

axA

ct

HEK-TLR7

R848

APR003

GS-9620

0.001 0.01 0.1 1 10 1000

20

40

60

80

100

Conc. (µM)

% M

axA

ct

HEK-TLR8

APR003

GS-9620

R848

HEK-TLR8 assay

pDC IFNa assay hPBMC IP-10 assay

0.01 0.1 10

50

100

150

uM

% o

f A00

1 M

ax

APR003 (hPBMC)

APR003R848

GS-9620

0.01 0.1 10

50

100

150

uM

% o

f A00

1 M

ax

APR003 (hPBMC)

APR003R848

GS-9620

• Effective concentration at 50% of maximal response (EC50)are shown for sigmoidal dose response curves.

• Lowest effective concentration (LEC), defined as the lowest concentration of test compound that elicited at least 3-fold signal over vehicle control, are shown for bell-shaped dose response curves.

6. Pharmacokinetics

7. Pharmacodynamics

0 20 40 60 80 100 1201

10

Fold

ove

r ve

hicl

e

IFNb expression

Small Intestine

Mesenteric LN

Liver

Large Intestine

Time (min)

Primary Response (30mpk; PO):IFNb expression (<2hrs)

0 20 40 60 80 100 1201

10

100

1000

Fold

ove

r ve

hicl

e

IP-10 expression

Small Intestine

Mesenteric LN

Liver

Large Intestine

Time (min)

Primary target activation in GI, liver, MLN

Secondary Response (30mpk; PO):

0 20 40 60 80 100 1201

10

100

1000

Fold

ove

r ve

hicl

e

IP-10 expression

Small Intestine

Mesenteric LN

Liver

Large Intestine

Time (min)

IP-10 (<2hrs)

0 20 40 60 80 100 1201

10

100

1000

Fold

ove

r ve

hicl

e

IP-10 expression

Small Intestine

Mesenteric LN

Liver

Large Intestine

Time (min)Amplified secondary ISG response in target tissues

0.1 1 10 100 10001

10

100

100

1000

10000

[compound] mpk

ISG

15 (f

old

over

veh

icle

)

A221 (liver PD vs serum PD)

ISG15 (liver)

IP-10 (serum)

[IP-10] pg/m

L

Dose-dependency (5hrs)Secondary Response:

[APR003] mg/kg

Dose-dependent increase in liver and serum

Tissue Distribution in mice dose PO (1hr)

Plasma

HeartLive

r

SpleenLung

KidneyBrai

n

Stomach

Small In

testin

e

Large I

ntestin

e

Testi

cleOva

ry FatSkin

Skelet

al1

10

100

1000

10000

100000

Con

c (n

g/m

L)

• Pulsatile kinetics (ideal for in situ vaccination): High clearance, short elimination half-life, and low volume of distribution

• Tissue-restricted distribution: concentrates in the GI, liver, and kidney (consistent with transporter profile)

• Primary response (transient): IFNb expression elicited in GI, liver, mesenteric lymph node• Secondary response (amplified): interferon stimulated genes (ISG) in the target tissues

and interferon-inducible protein 10 (IP-10) in serum• Dose dependency: no bell-shape response observed over 1000-fold range

8. IP-10 Induction Over TNFa

0.1 1 10 10010

100

1000

10000

0

100

200

300

400

[compound] mpk

[IP10

] pg/

mL

GS-9620 (IP-10)

APR003 (TNFa)GS-9620 (TNFa)

APR003 (IP-10)

[TNFa] pg/m

L

PD dose response Body weight change after a single high dose

0 1 2 3 4 5 6 7-15

-10

-5

0

5

10

% c

hang

e in

bod

y w

eigh

t

% change in body weight from Day 0

Vehicle

A221 (300mpk)

GS-9620 (150mpk)

Days

APR003 (300mpk)

0.1 1 10 10010

100

1000

10000

0

100

200

300

400

[compound] mpk

[IP10

] pg/

mL

GS-9620 (IP-10)

APR003 (TNFa)GS-9620 (TNFa)

APR003 (IP-10)

[TNFa] pg/m

L

• APR003 induced stronger IP-10 response over TNFa compared to GS-9620 (oral TLR7 agonist, Phase 2 for HBV)

• GS-9620 caused more body weight loss at doses that produced similar IP-10 levels as APR003

9. Orthotopic Syngeneic Liver Tumor Model

10. CT26 Liver Tumor Model

11. CT26 Liver Tumor Model (Dose Response)

12. CT26 Colorectal Tumor Model

13. Local Immune Priming (CD103 dendritic cells)

14. Increased Tumor-Infiltrating CD8 T Cells

15. Tissue-Specific vs Systemic Administration

16. Summary

• Bioluminescence imaging allows non-invasive monitoring of tumor growth on internal organs• Cross-validation by gross examination, liver weights, and survival

0 10 20 304

6

8

10

Bio

lum

ines

ence

Log

10 (F

lux

p/s)

Veh

negative control

0 10 20 304

6

8

10

Bio

lum

ines

ence

Log

10 (F

lux

p/s)

aPDL1

negative control0 10 20 30

4

6

8

10

Bio

lum

ines

ence

Log

10 (F

lux

p/s)

A221 (30mpk) + anti-PDL1negative control

0 10 20 304

6

8

10

Bio

lum

ines

ence

Log

10 (F

lux

p/s)

A221 (30mpk)

negative control

Vehicle Anti-PDL1 APR003

0 10 20 30 40 50 600

20

40

60

80

100

Day

% s

urvi

val

Survival

Vehicle

aPDL1

A221A221 + aPDL1

Tumor free mice: re-challenged subQ on D68

Median Survival Time

41 days

43 days

52.5 days58.5 days

**

***

0 5 10 15 20 25 30 35

6

8

10

Mea

n bi

olum

ines

ence

Log

10 (p

h/s)

± S

EM

VehaPDL1A221 (30mpk)A221 (30mpk) + anti-PDL1negative control

*

Days post-implant

***

randomization and start of treatment

treatments

Tumor Burden Survival

SC tumor rechallengeof survivors

APR003 + Anti-PDL1

0 10 20 30 40 50 600

20

40

60

80

100

Day

% s

urvi

val

Survival

Vehicle

aPDL1

APR003APR003 + aPDL1

Tumor free mice: re-challenged subQ on D68

Median Survival Time

41 days

43 days

52.5 days58.5 days

**

***

Tumor Burden

0 5 10 15 20 250

500

1000

Tum

or v

olum

e (m

m3)

Day 10, 22 post-subQ rechallengeAge-matched Control

aPDL1

APR003

APR003 + anti-PDL1

Day post-subQ rechallenge

P-value* = <0.05** = <0.01*** = <0.001

Tumor burden was monitored weekly by bioluminescent imaging. Data on left show mean bioluminescence and individual animal traces below.

• In an orthotopic CT26 liver cancer model, APR003 (30 mg/kg) decreased tumor burden and increased survival as a single agent and effects were augmented in combo with anti-PDL1

• Survivors resistant to subcutaneous tumor re-challenge, suggesting adaptive systemic immunity

On Day 0, CT26-luc cells are surgically implanted into the right liver lobe of Balb/c mice. On Day 4, mice are imaged, randomized, and dosed 2x/wk for 4 wks

Re-challenge survivors subQ

0 7 14 21 35 68Day

Dose

12

18

25

33

Vehicle APR003 APR003 + Anti-PDL1Day Liver weights

28

Anti-PDL1

4

Image

• In an orthotopic CT26 colon cancer model, APR003 (30 mg/kg) decreased tumor burden and increased survival as single agent and effects were augmented in combo with anti-PDL1

• APR003 induced a greater frequency of activated CD103+ dendritic cells in liver draining lymph nodes, but not in distal non-draining lymph nodes

CD8

B220

CD3

5.6 9.7 8 13

CD8+

CD3+B220-

AH1-dextramer

Following 3 therapeutic doses, liver tumors were removed, and single cell suspensions were stained and analyzed by flow cytometry to assess TILs.

Vehicle Anti-PDL1 APR003APR003

+ anti-PDL1

• Combination treatment of APR003 on top of anti-PD-L1 further increased tumor-specific (AH1+) CD8+ T cells in the liver tumor

*

n.s.

Tumor Burden

0 10 20 30 40

6

8

10

Day post implant

Mea

n bi

olum

ines

ence

Log

10 (F

lux

p/s)

Exp035: CT26 IH

Vehicle control

Anti-PDL1

APR003 (SubQ) + aPDL1

APR003 (oral) + aPDL1

Negative control

treatments

randomization and start of treatment

0 10 20 30 40

6

8

10

Day post implant

Mea

n bi

olum

ines

ence

Log

10 (F

lux

p/s)

Exp035: CT26 IH

Vehicle control

Anti-PDL1

APR003 (SubQ) + aPDL1

APR003 (oral) + aPDL1

Negative control

treatments

randomization and start of treatment

Median Survival (d)4245>98 * n.s.

80

On Day 0, CT26-luc are surgically implanted into the right liver lobe of Balb/c mice. On Day 4, mice are imaged, randomized, and dosed 2x/wk for 4 wks

Vehicl

e contro

l

Anti-PDL1

APR003 (

oral)

+ aPDL1

APR003 (

subQ) +

aPDL1

100

1000

10000

[IP10

] pg/

mL

Serum IP-10 (post 5th dose)IP-10

Vehicl

e contro

l

Anti-PDL1

APR003 (

oral)

+ aPDL1

APR003 (

subQ) +

aPDL1

100

1000

[IL-6

] pg/

mL

Serum IL-6 (post 5th dose)IL-6

Vehicl

e contro

l

Anti-PDL1

APR003 (

oral)

+ aPDL1

APR003 (

subQ) +

aPDL1

10

100

[TN

Fa] p

g/m

L

Serum TNFa (post 5th dose)TNFa

Cytokine profile (5hr post 5th dose)

• Systemic administration (via subcutaneous injection) of APR003 is less efficacious compared to tissue-specific oral administration at comparable liver exposure

• However, despite similar levels of IP-10 induction, systemic administration (higher serum exposure) induced more serum IL-6 and TNFa, which were not essential for anti-tumor efficacy and can lead to poor tolerability

• Therefore, GI/liver-targeted oral administration is a more optimal way to deliver TLR7 agonist to colorectal and liver cancers over systemic administration

10 20 30 405

6

7

8

9

Day post implant

Bio

lum

ines

ence

Log

10 (F

lux

p/s)

10 20 30 405

6

7

8

9

Day post implant

Bio

lum

ines

ence

Log

10 (F

lux

p/s)

10 20 30 405

6

7

8

9

Day post implant

Bio

lum

ines

ence

Log

10 (F

lux

p/s)

10 20 30 405

6

7

8

9

Day post implant

Bio

lum

ines

ence

Log

10 (F

lux

p/s)

0 10 20 30 406

7

8

Day post implant

Bio

lum

ines

ence

Log

10 (F

lux

p/s)

+ S

EM

Vehicle

aPDL1

A221

A221 + aPDL1

enrollmentTumor Burden

APR003 + Anti-PDL1Vehicle Anti-PDL1 APR003

On Day 0, CT26-luc are surgically implanted into the cecum of Balb/c mice. On Day 3 mice are imaged and randomized, followed by 2x/wk dosing on Day 4 for 4 weeks. Tumor burden was monitored weekly by bioluminescent imaging. Data on left show mean bioluminescence and individual animal traces below.

**

*

****

0 10 20 30 40 50 60 700

20

40

60

80

100

Day post implant

% s

urvi

val

Survival

Vehicle

anti-PDL1

APR003

APR003 + aPDL1

Survival

0 10 20 30 40 50 60 700

20

40

60

80

100

Day post implant

% s

urvi

val

Survival

Vehicle

anti-PDL1

APR003

APR003 + aPDL1

MedianSurvival Time26.5 days

41 days36.5 days59.5 days *

*********

P-value* = <0.05** = <0.01*** = <0.001**** = < 0.0001

***********

On Day 0, 1e5 CT26-luc were surgically implanted into the right liver lobe of Balb/c mice. On Day 5, mice were imaged, randomized, and dosed twice perweek starting on Day 5. Tumor burden was monitored weekly by bioluminescent imaging.

Efficacy in Dose Response (D29)

0 10 20 30

6

8

10

Mea

n bi

olum

ines

ence

Log

10 (F

lux

p/s)

+/-

SEM

CT26 IH model

Vehicle

anti-PDL1

A221 (1mpk) + anti-PDL1

A221 (3mpk) + anti-PDL1

A221 (10mpk) + anti-PDL1

A221 (30mpk) + anti-PDL1

A221 (100mpk) + anti-PDL1

A221 (300mpk) + anti-PDL1

Negative control

randomization and start of treatment

Days post implant

Tumor Burden

Vehicl

e

Anti-PDL1 1 3 10 30 100 300

A221 [mpk] + Anti-PDL1- ctrl

Vehicl

e

anti-

PDL1

A221 (

1mpk)

+ an

ti-PDL1

A221 (

3mpk)

+ an

ti-PDL1

A221 (

10m

pk) +

anti-

PDL1

A221 (

30m

pk) +

anti-

PDL1

A221 (

100m

pk) +

anti-

PDL1

A221 (

300m

pk) +

anti-

PDL1

Negat

ive co

ntrol

5

6

7

8

9

10

Mea

n bi

olum

ines

ence

Log

10 (F

lux

p/s)

+/-

SE

M

Mean - Day 29P-value

* = <0.05** = <0.01*** = <0.001**** = <0.0001

• APR003 exhibited anti-tumor efficacy over a wide dose range (10 – 300 mg/kg) in combination with anti-PDL1

BSP and rifampicin are inhibitors of OATP1A2 and OATP1B1, respectively

Cellular Assay R848 GS9620 APR003

TLR7 EC50 (µM) 0.14 0.21 0.23

TLR8 EC50 (µM) 2 >30 4.4

THP1 (TLR2/4/8) EC50 (µM) 9.7 >30 >30

hu PBMC IP-10 LEC (µM) 0.05 0.05 0.14

mk PBMC IP-10 LEC (µM) 0.14 0.14 0.41

ms Spleno IP-10 LEC (µM) <0.02 0.14 <0.02

AACR 2020Poster #684

• Target selectivity: APR003 is >10-fold selective for TLR7 over TLR8• Cellular activity: Active in plasmacytoid dendritic cells, but not in THP-1 monocytes• Species cross reactivity: Active across mouse, monkey, and human cells

TLR7 activation

Type I IFN(transient)

IP-10, ISG(amplified)

General Protocol: On Day 0, CT26-luc cells were surgically implanted into the right liver lobe of Balb/c mice. On Day 4, mice were imaged, randomized, and dosed twice-weekly for 4 weeks (APR003 and/or anti-PDL1). Tumor burden were monitored by bioluminescence imaging, and the statistical significance was calculated by 2-way ANOVA with multi-group comparison. Animal survival was tracked, and the statistical significance was calculated by log-rank test of Kaplan-Meier curves.

On Day 0, CT26-luc were surgically implanted into the right liver lobe of Balb/c mice. On Day 11, mice were imaged, randomized, and dosed on Day 11, 15, and 18, and then sacrificed on Day 19 to assess the frequency of activated CD103+ dendritic cells (cDC1) in the liver draining LNs (celiac, portal, and first mesenteric) versus non-draining LNs (axillary/brachial) by staining for MHCII and CD86.

Liver draining lymph nodes

Axillary/Brachial lymph nodes

CD45

MH

C II

Live/singlet gated

CD103

CD

86

Liver Tumor draining LNs

Vehicle

APR003

Vehicl

e

anti-P

DL1A22

1

Combo

Vehicl

e

anti-P

DL1A22

1

Combo0.0

0.5

1.0

1.5

2.0

%C

D45

+ M

HC

IIhi C

D10

3+ C

D86

+

Frequency of activated cDC1

Vehicle

anti-PDL1

A221

Combo

Draining LNs Non-draining LNs

**

CD103+ dendritic cells (cDC1s) are known to transport antigens to the lymph nodes and cross-present to tumor-specific CD8+T cells (ref 5)

On Day 0, CT26-luc were surgically implanted into the right liver lobe of Balb/c mice. Following 3 therapeutic doses, liver tumors were removed, and single cell suspensions were stained and analyzed by flow cytometry to assess TILs.

On Day 0, CT26-luc cells were surgically implanted into cecum of Balb/c mice. On Day 3, mice are imaged, randomized, and dosed twice-weekly for 4 weeks (APR003 and/or anti-PDL1).

On Day 0, CT26-luc cells were surgically implanted into the right liver lobe of Balb/c mice. On Day 4, mice were imaged, randomized, and dosed twice-weekly for 4 weeks with APR003 (either PO or SC) and anti-PDL1.

AH1 is a known antigen in the CT26 tumor cells (ref 6)

• As clinical proof-of-concept, intratumoral TLR9 agonists have shown promising anti-tumor responses in combination with checkpoint inhibitors; however, successful clinical applications have been limited to cutaneous-accessible tumor types (ref 2)

• APR003 was designed using medicinal chemistry principles of liver-targeting drugs, including transporter uptake properties (ref 3)

Route PK Parameters Mouse Monkey

IV

Dose (mg/kg) 3 1

t1/2 (h) 2.92 1.38

CL (mL/min/kg) 99.6 21.6

Vdss (L/kg) 1.88 0.688

Oral

Dose (mg/kg) 10 5

Tmax (h) 0.25 0.58

Cmax (ng/mL) 94.2 104

AUC0–t (ng·h/mL) 76.5 178

F (%) 4.6 4.2

PKLiver Serum

AUC (ng*h/mL)

PO (30mpk) 5000 310

SC (100μg) 4400 1800

TNFa is not required for T cell mediated anti-tumor efficacy and is associated with poor tolerability at high levels (ref 4)

LOCAL IMMUNE PRIMING LEADS TO SYSTEMIC ANTI-TUMOR IMMUNITY

http://www.aprostx.com/