Concurrent Inhibition of IGF1R and ERK Increases Pancreatic Cancer Sensitivity to Autophagy Inhibitors

Research output: Contribution to journalJournal articleResearchpeer-review

Standard

Concurrent Inhibition of IGF1R and ERK Increases Pancreatic Cancer Sensitivity to Autophagy Inhibitors. / Stalnecker, Clint A.; Grover, Kajal R.; Edwards, A. Cole; Coleman, Michael F.; Yang, Runying; DeLiberty, Jonathan M.; Papke, Bjorn; Goodwin, Craig M.; Pierobon, Mariaelena; Petricoin, Emanuel F.; Gautam, Prson; Wennerberg, Krister; Cox, Adrienne D.; Der, Channing J.; Hursting, Stephen D.; Bryant, Kirsten L.

In: Cancer Research, Vol. 82, No. 4, 2022, p. 586-598.

Research output: Contribution to journalJournal articleResearchpeer-review

Harvard

Stalnecker, CA, Grover, KR, Edwards, AC, Coleman, MF, Yang, R, DeLiberty, JM, Papke, B, Goodwin, CM, Pierobon, M, Petricoin, EF, Gautam, P, Wennerberg, K, Cox, AD, Der, CJ, Hursting, SD & Bryant, KL 2022, 'Concurrent Inhibition of IGF1R and ERK Increases Pancreatic Cancer Sensitivity to Autophagy Inhibitors', Cancer Research, vol. 82, no. 4, pp. 586-598. https://doi.org/10.1158/0008-5472.CAN-21-1443

APA

Stalnecker, C. A., Grover, K. R., Edwards, A. C., Coleman, M. F., Yang, R., DeLiberty, J. M., Papke, B., Goodwin, C. M., Pierobon, M., Petricoin, E. F., Gautam, P., Wennerberg, K., Cox, A. D., Der, C. J., Hursting, S. D., & Bryant, K. L. (2022). Concurrent Inhibition of IGF1R and ERK Increases Pancreatic Cancer Sensitivity to Autophagy Inhibitors. Cancer Research, 82(4), 586-598. https://doi.org/10.1158/0008-5472.CAN-21-1443

Vancouver

Stalnecker CA, Grover KR, Edwards AC, Coleman MF, Yang R, DeLiberty JM et al. Concurrent Inhibition of IGF1R and ERK Increases Pancreatic Cancer Sensitivity to Autophagy Inhibitors. Cancer Research. 2022;82(4):586-598. https://doi.org/10.1158/0008-5472.CAN-21-1443

Author

Stalnecker, Clint A. ; Grover, Kajal R. ; Edwards, A. Cole ; Coleman, Michael F. ; Yang, Runying ; DeLiberty, Jonathan M. ; Papke, Bjorn ; Goodwin, Craig M. ; Pierobon, Mariaelena ; Petricoin, Emanuel F. ; Gautam, Prson ; Wennerberg, Krister ; Cox, Adrienne D. ; Der, Channing J. ; Hursting, Stephen D. ; Bryant, Kirsten L. / Concurrent Inhibition of IGF1R and ERK Increases Pancreatic Cancer Sensitivity to Autophagy Inhibitors. In: Cancer Research. 2022 ; Vol. 82, No. 4. pp. 586-598.

Bibtex

@article{a42d556a12194d02bb74c47695071bc8,
title = "Concurrent Inhibition of IGF1R and ERK Increases Pancreatic Cancer Sensitivity to Autophagy Inhibitors",
abstract = "The aggressive nature of pancreatic ductal adenocarcinoma (PDAC) mandates the development of improved therapies. As KRAS mutations are found in 95% of PDAC and are critical for tumor maintenance, one promising strategy involves exploiting KRAS-dependent metabolic perturbations. The macrometabolic process of autophagy is upregulated in KRAS-mutant PDAC, and PDAC growth is reliant on autophagy. However, inhibition of autophagy as monotherapy using the lysosomal inhibitor hydroxychloroquine (HCQ) has shown limited clinical efficacy. To identify strategies that can improve PDAC sensitivity to HCQ, we applied a CRISPR-Cas9 loss-of-function screen and found that a top sensitizer was the receptor tyrosine kinase (RTK) insulin-like growth factor 1 receptor (IGF1R). Additionally, reverse phase protein array pathway activation mapping profiled the signaling pathways altered by chloroquine (CQ) treatment. Activating phosphorylation of RTKs, including IGF1R, was a common compensatory increase in response to CQ. Inhibition of IGF1R increased autophagic flux and sensitivity to CQ-mediated growth suppression both in vitro and in vivo. Cotargeting both IGF1R and pathways that antagonize autophagy, such as ERK-MAPK axis, was strongly synergistic. IGF1R and ERK inhibition converged on suppression of glycolysis, leading to enhanced dependence on autophagy. Accordingly, concurrent inhibition of IGF1R, ERK, and autophagy induced cytotoxicity in PDAC cell lines and decreased viability in human PDAC organoids. In conclusion, targeting IGF1R together with ERK enhances the effectiveness of autophagy inhibitors in PDAC. Significance: Compensatory upregulation of IGF1R and ERK- MAPK signaling limits the efficacy of autophagy inhibitors chloroquine and hydroxychloroquine, and their concurrent inhibition synergistically increases autophagy dependence and chloroquine sensitivity in pancreatic ductal adenocarcinoma.",
author = "Stalnecker, {Clint A.} and Grover, {Kajal R.} and Edwards, {A. Cole} and Coleman, {Michael F.} and Runying Yang and DeLiberty, {Jonathan M.} and Bjorn Papke and Goodwin, {Craig M.} and Mariaelena Pierobon and Petricoin, {Emanuel F.} and Prson Gautam and Krister Wennerberg and Cox, {Adrienne D.} and Der, {Channing J.} and Hursting, {Stephen D.} and Bryant, {Kirsten L.}",
note = "Publisher Copyright: {\textcopyright} 2022 American Association for Cancer Research Inc.. All rights reserved.",
year = "2022",
doi = "10.1158/0008-5472.CAN-21-1443",
language = "English",
volume = "82",
pages = "586--598",
journal = "Cancer Research",
issn = "0008-5472",
publisher = "American Association for Cancer Research",
number = "4",

}

RIS

TY - JOUR

T1 - Concurrent Inhibition of IGF1R and ERK Increases Pancreatic Cancer Sensitivity to Autophagy Inhibitors

AU - Stalnecker, Clint A.

AU - Grover, Kajal R.

AU - Edwards, A. Cole

AU - Coleman, Michael F.

AU - Yang, Runying

AU - DeLiberty, Jonathan M.

AU - Papke, Bjorn

AU - Goodwin, Craig M.

AU - Pierobon, Mariaelena

AU - Petricoin, Emanuel F.

AU - Gautam, Prson

AU - Wennerberg, Krister

AU - Cox, Adrienne D.

AU - Der, Channing J.

AU - Hursting, Stephen D.

AU - Bryant, Kirsten L.

N1 - Publisher Copyright: © 2022 American Association for Cancer Research Inc.. All rights reserved.

PY - 2022

Y1 - 2022

N2 - The aggressive nature of pancreatic ductal adenocarcinoma (PDAC) mandates the development of improved therapies. As KRAS mutations are found in 95% of PDAC and are critical for tumor maintenance, one promising strategy involves exploiting KRAS-dependent metabolic perturbations. The macrometabolic process of autophagy is upregulated in KRAS-mutant PDAC, and PDAC growth is reliant on autophagy. However, inhibition of autophagy as monotherapy using the lysosomal inhibitor hydroxychloroquine (HCQ) has shown limited clinical efficacy. To identify strategies that can improve PDAC sensitivity to HCQ, we applied a CRISPR-Cas9 loss-of-function screen and found that a top sensitizer was the receptor tyrosine kinase (RTK) insulin-like growth factor 1 receptor (IGF1R). Additionally, reverse phase protein array pathway activation mapping profiled the signaling pathways altered by chloroquine (CQ) treatment. Activating phosphorylation of RTKs, including IGF1R, was a common compensatory increase in response to CQ. Inhibition of IGF1R increased autophagic flux and sensitivity to CQ-mediated growth suppression both in vitro and in vivo. Cotargeting both IGF1R and pathways that antagonize autophagy, such as ERK-MAPK axis, was strongly synergistic. IGF1R and ERK inhibition converged on suppression of glycolysis, leading to enhanced dependence on autophagy. Accordingly, concurrent inhibition of IGF1R, ERK, and autophagy induced cytotoxicity in PDAC cell lines and decreased viability in human PDAC organoids. In conclusion, targeting IGF1R together with ERK enhances the effectiveness of autophagy inhibitors in PDAC. Significance: Compensatory upregulation of IGF1R and ERK- MAPK signaling limits the efficacy of autophagy inhibitors chloroquine and hydroxychloroquine, and their concurrent inhibition synergistically increases autophagy dependence and chloroquine sensitivity in pancreatic ductal adenocarcinoma.

AB - The aggressive nature of pancreatic ductal adenocarcinoma (PDAC) mandates the development of improved therapies. As KRAS mutations are found in 95% of PDAC and are critical for tumor maintenance, one promising strategy involves exploiting KRAS-dependent metabolic perturbations. The macrometabolic process of autophagy is upregulated in KRAS-mutant PDAC, and PDAC growth is reliant on autophagy. However, inhibition of autophagy as monotherapy using the lysosomal inhibitor hydroxychloroquine (HCQ) has shown limited clinical efficacy. To identify strategies that can improve PDAC sensitivity to HCQ, we applied a CRISPR-Cas9 loss-of-function screen and found that a top sensitizer was the receptor tyrosine kinase (RTK) insulin-like growth factor 1 receptor (IGF1R). Additionally, reverse phase protein array pathway activation mapping profiled the signaling pathways altered by chloroquine (CQ) treatment. Activating phosphorylation of RTKs, including IGF1R, was a common compensatory increase in response to CQ. Inhibition of IGF1R increased autophagic flux and sensitivity to CQ-mediated growth suppression both in vitro and in vivo. Cotargeting both IGF1R and pathways that antagonize autophagy, such as ERK-MAPK axis, was strongly synergistic. IGF1R and ERK inhibition converged on suppression of glycolysis, leading to enhanced dependence on autophagy. Accordingly, concurrent inhibition of IGF1R, ERK, and autophagy induced cytotoxicity in PDAC cell lines and decreased viability in human PDAC organoids. In conclusion, targeting IGF1R together with ERK enhances the effectiveness of autophagy inhibitors in PDAC. Significance: Compensatory upregulation of IGF1R and ERK- MAPK signaling limits the efficacy of autophagy inhibitors chloroquine and hydroxychloroquine, and their concurrent inhibition synergistically increases autophagy dependence and chloroquine sensitivity in pancreatic ductal adenocarcinoma.

U2 - 10.1158/0008-5472.CAN-21-1443

DO - 10.1158/0008-5472.CAN-21-1443

M3 - Journal article

C2 - 34921013

AN - SCOPUS:85124888529

VL - 82

SP - 586

EP - 598

JO - Cancer Research

JF - Cancer Research

SN - 0008-5472

IS - 4

ER -

ID: 343129735