Therapeutic Potential of Targeting Amino Acid Metabolism ......Therapeutic Potential of Targeting...

1
Therapeutic Potential of Targeting Amino Acid Metabolism in Pancreatic Cancer CORRESPONDING AUTHOR: (e): [email protected] (ph): +1-917-634-6165 Martin Fernandez-Zapico 1 Dae Won Kim 2 Philip Philip 3 Alexander Vandell 4 Jonathan Eckard 4 Ron Korn 5 Giuseppe Del Priore 4 Diane Simeone 6 1 Mayo Clinic 2 Moffitt Cancer Center 3 Karmanos Cancer Institute 4 TYME Inc. 5 Imaging Endpoints 6 NYU Langone Health RESULTS AND CASE STUDIES DOMINANT LIVER LESION PATIENT A BASELINE POST THERAPY PANCREAS HEAD LESION HEPATIC LESIONS PANCREATIC TAIL MASS 2.80 cm x 3.48cm 4.87 cm x 4.22cm 61-year-old female with stage IV PDAC. Previously treated with GA with Indoximod, FOLFIRINOX, and Pembrolizumab. SM-88 + MPS: Monotherapy in the fourth line setting was ongoing for 8.7 months as of 4/25/19 cutoff. Shown RECIST response observed at 80 days after initiating therapy. CA-19.9 was >27,000 at baseline and decreased >90% after three months of therapy. Â 16/24 (67%) patients from the Phase II study in pancreatic cancer demonstrated a reduction from baseline with a median percentage decrease of 63% in CTCs. Â LAT-1 has been reported to be important in EMT transition and mesenchymal cell survival (Holldorsson S et al.) POSSIBLY RELATED PROBABLY RELATED RELATED Phase II Trial (n=49) Grade 3 - Severe 6 1 2 Grade 4 - Life-Threatening 1 0 2 Grade 5 - Fatal 0 0 0 FHS and Compassionate Use (n=10) Grade 3 - Severe 0 0 0 Grade 4 - Life-Threatening 0 0 0 Grade 5 - Fatal 0 0 0 Patients * Indicates patients with OS of at least 180 days * * * * * * * * * * * * 200 – 100 – 0 – -100 – Grade 3/4 Adverse Events deemed related were observed in only two patients. Overall the Grade 3/4 adverse events at least possibly related included Depression, Arthralgia, Anemia, Aspartate aminotransferase increase, Hypokalemia, Diarrhea, Abdominal Pain, Portal Vein Thrombosis, Hypotension. Â Numerous cancers display an increased cellular demand for amino acids in order to sustain their increased proliferation rate. (Hosios A et al.) Â L-amino acid transporter (LAT-1) is responsible for cellular uptake of large non-polar amino acids including tyrosine. (Hayashi K et al.) Â LAT-1 overexpression in pancreatic cancer has been shown to be an important prognostic factor for development of chemoresistance and survival in patients with pancreatic ductal adenocarcinoma (PDAC). (Karia et al., and Yanagisawa et al.) Â SM-88 (racemetyrosine) is the racemic mixture of a tyrosine derivative designed to leverage LAT-1 overexpression for selective tumor uptake, and subsequent disruption of protein synthesis resulting in cancer cell death. Â In two clinical trials of SM-88 in patients with PDAC, no prominent safety signals were observed, and overall survival was longer than expected in this population. Â Several SM-88 treated PDAC patients experienced objective tumor responses, and a large proportion of patients displayed a reduction in circulating tumor cells (CTCs). LAT-1 is overexpressed in a wide variety of cancers including pancreas, prostate, and breast. Drugs that inhibit LAT-1 activity are under investigation for the treatment of cancer. (Hafliger et al.) Tumor imaging agents, including 3-18F-l-α-methyl-tyrosine ([18F] FAMT), utilize LAT-1 as a cellular uptake mechanism to selectively target cancer cells. (Wei L et al.) SM-88 is a novel anti-cancer agent (racemic D/L α-methyl-para- tyrosine) hypothesized to be transported into cells via LAT-1. This selective cancer uptake may result in increased efficacy and reduced frequency of adverse events. Once inside the cancer cell, SM-88 is believed to disrupt protein synthesis machinery, induce oxidative stress, and decrease defense mechanisms. Several D-amino acids have been shown to cause: Translational arrest (Fleisher R et al., Zheng G et al.) Increases in oxidative stress and apoptosis. (Bardaweel et al., 2013) Cancer cells' increased protein synthesis is accompanied by an upregulation of tRNAs (Pavon-Eternod et al., 2009). Mutations in ribosomes and other translational machinery components may decrease translational fidelity (Pelletier et al.), increasing the susceptibility of cancer cells to D-amino acid incorporation. Â Evaluation of the anti-cancer effects of SM-88 using in vitro (cell lines), and in vivo (xenograft) models of PDAC. Â Evaluation of SM-88 efficacy in models of additional cancer types. Â Determination of the contribution of the L and D isomers to the effects of SM-88. Â Detailed examination of potential systemic and tumor immunomodulatory effects of SM-88. Â Use of pre-clinical models to evaluate SM-88 effects on migration, invasion, and metastatic potential. Â Combinations of SM-88 and standard therapies. Â Altered amino acid (LAT-1) metabolism has shown to be a negative prognosticator in pancreatic cancer outcomes and progression. Â Results from clinical studies of SM-88 suggest the therapy is well tolerated and early signs of anti- tumor effects have been observed. Â Ongoing and planned pre-clinical studies with SM-88 could further clarify the mechanism and potential combinations where SM-88 could be studied. Â Future studies may clarify the potential utility of this approach in the treatment of pancreatic cancer. 1) Kairia AB et al. Relationship between LAT1 expression and resistance to chemotherapy in pancreatic ductal adenocarcinoma. Cancer Chemotherapy Pharmacol, 2018; 81(1):141-153 2) Yanagisawa N et.al. High Expression of L-type amino acid transporter 1 (LAT1) predicts poor prognosis in pancreatic ductal adenocarcinoma. J Clin Pathol. 2012; 65(11);1019-23 3) Noel et al. SM-88 Therapy in High-Risk Poor Prognosis Pancreatic Cancer, ESMO GI 2019 Poster 4) Resa B et al. b2 Adrenergic-Neurotrophin Feedforward Loop Promotes Pancreatic Cancer. Cancer Cell, 2018; 33: 75-90 5) Hayashi K et.al. Novel therapeutic approaches targeting L-type amino acid transporters for cancer treatment; World J Gastrointest Oncol 2017 January 15; 9(1): 21-29 6) Hosios A et.al. Amino Acids Rather than Glucose Account for the Majority of Cell Mass in Proliferating Mammalian Cells; Developmental Cell 36, 540–549 7) Hafliger et.al. The L-Type Amino Acid Transporter LAT1-An Emerging Target in Cancer; Int J Mol Sci. 2019 May 16;20(10) 8) Wei L et.al. Specific transport of 3-fluoro-l-α-methyl-tyrosine by LAT1 explains its specificity to malignant tumors in imaging; Cancer 2016: 107 (3) 347 9) Fleisher R et.al. D-amino acid-mediated translation arrest is modulated by the identity of the incoming aminoacyl-tRNA. 2018; Biochemistry, 57 (29) 4241-46 10) Zheng G et.al. Human D-Tyr-tRNATyr deacylase contributes to the resistance of the cell toD-amino acids. Biochem. J. (2009) 417, 85–94 11) Hermano E et.al. Macrophage Polarization in Pancreatic carcinoma: role of Heparanase enzyme. 2014; JCNI, 106 (12) 12) Noel et al. Annals of Oncology 2019, Volume 30, Issue Supplement_4, July 2019 13) Hollderson S et.al. Metabolic re-wiring of isogenic breast epithelial cell lines following epithelial to mesenchymal transition. 2017 Cancer Letters; 396, 117-129 14) Korner J et.al. A role for foregut tyrosine metabolism in glucose tolerance Molecular Metabolism. 2019; 23, 37-50 15) Renz B et.al. b2 Adrenergic-Neurotrophin Feedforward Loop Promotes Pancreatic Cancer. Cancer Cell, 2018; 33, 75–90 16) LamkinD et.al. β-adrenergic-stimulated macrophages: Comprehensive localization in the M1–M2 spectrum. Brain Behav Immun. 2016 October ; 57: 338–346 17) Baraweel S et.al. An in vitro based investigation into the cytotoxic effects of D-amino acids. 2013; Acta Pharm. 63: 467–478 18) Pavon-Eternod M et.al. tRNA over-expression in breast cancer and functional consequence. 2009; Nucleic Acids Research, 37 (21): 7268–80 19) Partecke L et.al. Chronic stress increases experimental pancreatic cancer growth, reduces survival and can be antagonised by beta-adrenergic receptor blockade. 2016; Pancreatology. 16 (3), 423-33 20) Pelletier J et.al. Ribosome biogenesis in cancer: new players and therapeutic avenues. 2018; Nature Reviews, 18, 51-63 CLINICAL TRIALS Â 10 patients with PDAC were enrolled in the First Human Study and subsequent compassionate use program between 2012 and 2017. These patients received 230mg of SM-88 five days per week. All patients also received M2PS (melanin 50 μg, melanotan II 10 μg, phenytoin 17 mg and sirolimus 0.25 mg per day). (Noel et al.) Â 49 patients with PDAC were enrolled in a randomized phase II of 460mg vs 920mg total daily doses of SM-88 in patients with radiographic PD, at least 1 prior line, and ECOG PS ≤2. All patients also received MPS (methox- salen 10 mg, phenytoin 50 mg, and sirolimus 0.5 mg per day). (Noel et al.) PROPOSED MECHANISM OF ACTION BASELINE POST THERAPY BASELINE POST THERAPY BASELINE POST THERAPY 58-year-old Caucasian male diagnosed with stage IV adenocarcinoma previously received FOLFOX with no tumor response and progressive disease. Baseline ECOG PS at time of SM-88 initiation was “2+” with CEA count of 291.4 U/mL. SM-88 + M2PS alone for 4 Months: Best RECIST Response observed at ~3 and maintained throughout treatment PATIENT B REFERENCES CONCLUSIONS/DISCUSSION ONGOING AND PLANNED WORK INTRODUCTION ADDITIONAL POTENTIAL EFFECTS CATECHOLAMINE AS A CANCER TARGET The L-isomer of SM-88 is known to inhibit tyrosine hydroxylase (Korner et al.), the rate limiting enzyme of catecholamine biosynthesis. Catecholamines have been shown to directly promote the growth of pancreatic cancer. (Renz et al., 2018; Partecke L et al.) Catecholamines also promote the development of an immune suppressive microenvironment within tumors by driving macrophages towards the M2 phenotype. (Lamkin et al., 2016) Tumor-associated macrophages (TAMs) in PDAC appear an important factor of the immune environment. (Hermano E, 2014) SM-88 induced reductions of catecholamines may inhibit cancer growth and promote the development of an anti-cancer M1 macrophage phenotype. CTC reduction following treatment with SM-88 in both the pancreatic and prostate cancer settings suggest that SM-88 has an effect on cell migration, invasion, and metastatic potential. Best Overall CTC % Change from Baseline Grade 3 and 4 Adverse Events in PDAC Patients

Transcript of Therapeutic Potential of Targeting Amino Acid Metabolism ......Therapeutic Potential of Targeting...

Therapeutic Potential of Targeting Amino Acid Metabolism in Pancreatic Cancer

CORRESPONDING AUTHOR:(e): [email protected]

(ph): +1-917-634-6165

Martin Fernandez-Zapico1 Dae Won Kim2 Philip Philip3 Alexander Vandell4 Jonathan Eckard4 Ron Korn5 Giuseppe Del Priore4 Diane Simeone6

1Mayo Clinic 2Moffitt Cancer Center 3Karmanos Cancer Institute 4TYME Inc. 5Imaging Endpoints 6NYU Langone HealthRESULTS AND CASE STUDIES

DOMINANT LIVER LESION

PAT

IEN

T A

BASELINE POST THERAPY

PANCREAS HEAD LESION

HEPATIC LESIONS PANCREATIC TAIL MASS

2.80 cmx 3.48cm4.87 cm x 4.22cm

61-year-old female with stage IV PDAC. Previously treated with GA with Indoximod, FOLFIRINOX, and Pembrolizumab.

SM-88 + MPS:Monotherapy in the fourth line setting was ongoing for 8.7 months as of 4/25/19 cutoff. Shown RECIST response observed at 80 days after initiating therapy. CA-19.9 was>27,000 at baseline and decreased >90% after three months of therapy.

 16/24 (67%) patients from the Phase II study in pancreatic cancer demonstrated a reduction from baseline with a median percentage decrease of 63% in CTCs.

 LAT-1 has been reported to be important in EMT transition and mesenchymal cellsurvival (Holldorsson S et al.)

POSSIBLY RELATED PROBABLY RELATED RELATED

Phase II Trial (n=49)

Grade 3 - Severe 6 1 2

Grade 4 - Life-Threatening 1 0 2

Grade 5 - Fatal 0 0 0

FHS and Compassionate Use (n=10)

Grade 3 - Severe 0 0 0

Grade 4 - Life-Threatening 0 0 0

Grade 5 - Fatal 0 0 0

Patients * Indicates patients with OS of at least 180 days

*

* * * *

*

* * * * * *

200 –

100 –

0 –

-100 –

Grade 3/4 Adverse Events deemed related were observed in only two patients.Overall the Grade 3/4 adverse events at least possibly related included Depression, Arthralgia, Anemia, Aspartate aminotransferase increase, Hypokalemia, Diarrhea, Abdominal Pain, Portal Vein Thrombosis, Hypotension.

 Numerous cancers display an increased cellular demand for amino acids in order to sustain their increased proliferation rate. (Hosios A et al.)

 L-amino acid transporter (LAT-1) is responsible for cellular uptake of large non-polar amino acids including tyrosine. (Hayashi K et al.)

 LAT-1 overexpression in pancreatic cancer has been shown to be an important prognostic factor for development of chemoresistance and survival in patients with pancreatic ductal adenocarcinoma (PDAC). (Karia et al., and Yanagisawa et al.)

 SM-88 (racemetyrosine) is the racemic mixture of a tyrosine derivative designed to leverage LAT-1 overexpression for selective tumor uptake, and subsequent disruption of protein synthesis resulting in cancer cell death.

 In two clinical trials of SM-88 in patients with PDAC, no prominent safety signals were observed, and overall survival was longer than expected in this population.

 Several SM-88 treated PDAC patients experienced objective tumor responses, and a large proportion of patients displayed a reduction in circulating tumor cells (CTCs).

⬣ LAT-1 is overexpressed in a wide variety of cancers including pancreas, prostate, and breast. Drugs that inhibit LAT-1 activity are under investigation for the treatment of cancer. (Hafliger et al.)

⬣ Tumor imaging agents, including 3-18F-l-α-methyl-tyrosine ([18F] FAMT), utilize LAT-1 as a cellular uptake mechanism to selectively target cancer cells. (Wei L et al.)

⬣ SM-88 is a novel anti-cancer agent (racemic D/L α-methyl-para-tyrosine) hypothesized to be transported into cells via LAT-1. This selective cancer uptake may result in increased efficacy and reduced frequency of adverse events.

⬣ Once inside the cancer cell, SM-88 is believed to disrupt protein synthesis machinery, induce oxidative stress, and decrease defense mechanisms.

⬣ Several D-amino acids have been shown to cause:

⬣ Translational arrest (Fleisher R et al., Zheng G et al.)

⬣ Increases in oxidative stress and apoptosis.(Bardaweel et al., 2013)

⬣ Cancer cells' increased protein synthesis is accompanied by an upregulation of tRNAs (Pavon-Eternod et al., 2009). Mutations in ribosomes and other translational machinery components may decrease translational fidelity (Pelletier et al.), increasing the susceptibility of cancer cells to D-amino acid incorporation.

 Evaluation of the anti-cancer effects of SM-88 using in vitro (cell lines), and in vivo (xenograft) models of PDAC.

 Evaluation of SM-88 efficacy in models of additional cancer types.

 Determination of the contribution of the L and D isomers to the effects of SM-88.

 Detailed examination of potential systemic and tumor immunomodulatory effects of SM-88.

 Use of pre-clinical models to evaluate SM-88 effects on migration, invasion, and metastatic potential.

 Combinations of SM-88 and standard therapies.

 Altered amino acid (LAT-1) metabolism has shown to be a negative prognosticator in pancreatic cancer outcomes and progression.

 Results from clinical studies of SM-88 suggest the therapy is well tolerated and early signs of anti-tumor effects have been observed.

 Ongoing and planned pre-clinical studies with SM-88 could further clarify the mechanism and potential combinations where SM-88 could be studied.

 Future studies may clarify the potential utility of this approach in the treatment of pancreatic cancer.

1) Kairia AB et al. Relationship between LAT1 expression and resistance to chemotherapy in pancreatic ductal adenocarcinoma. CancerChemotherapy Pharmacol, 2018; 81(1):141-153

2) Yanagisawa N et.al. High Expression of L-type amino acid transporter 1 (LAT1) predicts poor prognosis in pancreatic ductal adenocarcinoma. J Clin Pathol. 2012; 65(11);1019-23

3) Noel et al. SM-88 Therapy in High-Risk Poor Prognosis Pancreatic Cancer, ESMO GI 2019 Poster

4) Resa B et al. b2 Adrenergic-Neurotrophin Feedforward Loop Promotes Pancreatic Cancer. Cancer Cell, 2018; 33: 75-90

5) Hayashi K et.al. Novel therapeutic approaches targeting L-type amino acid transporters for cancer treatment; World J Gastrointest Oncol 2017 January 15; 9(1): 21-29

6) Hosios A et.al. Amino Acids Rather than Glucose Account for the Majority of Cell Mass in Proliferating Mammalian Cells; Developmental Cell 36, 540–549

7) Hafliger et.al. The L-Type Amino Acid Transporter LAT1-An Emerging Target in Cancer; Int J Mol Sci. 2019 May 16;20(10)

8) Wei L et.al. Specific transport of 3-fluoro-l-α-methyl-tyrosine by LAT1 explains its specificity to malignant tumors in imaging; Cancer 2016: 107 (3) 347

9) Fleisher R et.al. D-amino acid-mediated translation arrest is modulated by the identity of the incoming aminoacyl-tRNA. 2018; Biochemistry, 57 (29) 4241-46

10) Zheng G et.al. Human D-Tyr-tRNATyr deacylase contributes to the resistance of the cell toD-amino acids. Biochem. J. (2009) 417, 85–94

11) Hermano E et.al. Macrophage Polarization in Pancreatic carcinoma: role of Heparanase enzyme. 2014; JCNI, 106 (12)

12) Noel et al. Annals of Oncology 2019, Volume 30, Issue Supplement_4, July 2019

13) Hollderson S et.al. Metabolic re-wiring of isogenic breast epithelial cell lines following epithelial to mesenchymal transition. 2017 Cancer Letters; 396,117-129

14) Korner J et.al. A role for foregut tyrosine metabolism in glucose tolerance Molecular Metabolism. 2019; 23, 37-50

15) Renz B et.al. b2 Adrenergic-Neurotrophin Feedforward Loop Promotes Pancreatic Cancer. Cancer Cell, 2018; 33, 75–90

16) LamkinD et.al. β-adrenergic-stimulated macrophages: Comprehensivelocalization in the M1–M2 spectrum. Brain Behav Immun. 2016 October ; 57: 338–346

17) Baraweel S et.al. An in vitro based investigation into the cytotoxic effects of D-amino acids. 2013; Acta Pharm. 63: 467–478

18) Pavon-Eternod M et.al. tRNA over-expression in breast cancer and functional consequence. 2009; Nucleic Acids Research, 37 (21): 7268–80

19) Partecke L et.al. Chronic stress increases experimental pancreatic cancer growth, reduces survival and can be antagonised by beta-adrenergic receptor blockade. 2016; Pancreatology. 16 (3), 423-33

20) Pelletier J et.al. Ribosome biogenesis in cancer: new players and therapeutic avenues. 2018; Nature Reviews, 18, 51-63

CLINICAL TRIALSÂ 10 patients with PDAC were enrolled

in the First Human Study and subsequent compassionate use program between 2012 and 2017. These patients received 230mg of SM-88 five days per week. All patients also received M2PS (melanin 50 μg, melanotan II 10 μg, phenytoin 17 mg and sirolimus 0.25 mg per day). (Noel et al.)

 49 patients with PDAC were enrolled in a randomized phase II of 460mg vs 920mg total daily doses of SM-88 in patients with radiographic PD, at least 1 prior line, and ECOG PS ≤2. All patients also received MPS (methox-salen 10 mg, phenytoin 50 mg, and sirolimus 0.5 mg per day). (Noel et al.)

PROPOSED MECHANISM OF ACTION

BASELINE POST THERAPY

BASELINE POST THERAPY BASELINE POST THERAPY

58-year-old Caucasian male diagnosed with stage IV adenocarcinoma previously received FOLFOX with no tumor response and progressive disease. Baseline ECOG PS at time of SM-88 initiation was “2+” with CEA count of 291.4 U/mL.

SM-88 + M2PS alone for 4 Months: Best RECIST Response observed at ~3and maintained throughout treatment

PAT

IEN

T B

REFERENCESCONCLUSIONS/DISCUSSIONONGOING AND PLANNED WORK

INTRODUCTION

ADDITIONAL POTENTIAL EFFECTS

CATECHOLAMINE AS A CANCER TARGET

⬣ The L-isomer of SM-88 is known to inhibit tyrosine hydroxylase (Korner et al.), the rate limiting enzyme of catecholamine biosynthesis.

⬣ Catecholamines have been shown to directly promote the growth of pancreatic cancer. (Renz et al., 2018; Partecke L et al.)

⬣ Catecholamines also promote the development of an immune suppressive microenvironment within tumors by driving macrophages towards the M2 phenotype. (Lamkin et al., 2016)

⬣ Tumor-associated macrophages (TAMs) in PDAC appear an important factor of the immune environment. (Hermano E, 2014)

⬣ SM-88 induced reductions of catecholamines may inhibit cancer growth and promote the development of an anti-cancer M1 macrophage phenotype.

⬣ CTC reduction following treatment with SM-88 in both the pancreatic and prostate cancer settings suggest that SM-88 has an effect on cell migration, invasion, and metastatic potential.

Best Overall CTC % Change from Baseline

Grade 3 and 4 Adverse Events in PDAC Patients