Protective autophagy elicited by RAF→MEK→ERK inhibition suggests a treatment strategy for RAS-driven cancers

Conan G Kinsey, Soledad A Camolotto, Amelie M Boespflug, Katrin P Guillen, Mona Foth, Amanda Truong, Sophia S Schuman, Jill E Shea, Michael T Seipp, Jeffrey T Yap, Lance D Burrell, David H Lum, Jonathan R Whisenant, G Weldon Gilcrease 3rd, Courtney C Cavalieri, Kaitrin M Rehbein, Stephanie L Cutler, Kajsa E Affolter, Alana L Welm, Bryan E Welm, Courtney L Scaife, Eric L Snyder, Martin McMahon, Conan G Kinsey, Soledad A Camolotto, Amelie M Boespflug, Katrin P Guillen, Mona Foth, Amanda Truong, Sophia S Schuman, Jill E Shea, Michael T Seipp, Jeffrey T Yap, Lance D Burrell, David H Lum, Jonathan R Whisenant, G Weldon Gilcrease 3rd, Courtney C Cavalieri, Kaitrin M Rehbein, Stephanie L Cutler, Kajsa E Affolter, Alana L Welm, Bryan E Welm, Courtney L Scaife, Eric L Snyder, Martin McMahon

Abstract

Pancreatic ductal adenocarcinoma (PDA) was responsible for ~ 44,000 deaths in the United States in 2018 and is the epitome of a recalcitrant cancer driven by a pharmacologically intractable oncoprotein, KRAS1-4. Downstream of KRAS, the RAF→MEK→ERK signaling pathway plays a central role in pancreatic carcinogenesis5. However, paradoxically, inhibition of this pathway has provided no clinical benefit to patients with PDA6. Here we show that inhibition of KRAS→RAF→MEK→ERK signaling elicits autophagy, a process of cellular recycling that protects PDA cells from the cytotoxic effects of KRAS pathway inhibition. Mechanistically, inhibition of MEK1/2 leads to activation of the LKB1→AMPK→ULK1 signaling axis, a key regulator of autophagy. Furthermore, combined inhibition of MEK1/2 plus autophagy displays synergistic anti-proliferative effects against PDA cell lines in vitro and promotes regression of xenografted patient-derived PDA tumors in mice. The observed effect of combination trametinib plus chloroquine was not restricted to PDA as other tumors, including patient-derived xenografts (PDX) of NRAS-mutated melanoma and BRAF-mutated colorectal cancer displayed similar responses. Finally, treatment of a patient with PDA with the combination of trametinib plus hydroxychloroquine resulted in a partial, but nonetheless striking disease response. These data suggest that this combination therapy may represent a novel strategy to target RAS-driven cancers.

Conflict of interest statement

Competing Interests Statement:

Dr. McMahon served on external advisory boards for Novartis (June 2017), Genentech (December 2017) and Merck (June 2018). Dr. McMahon is the recipient of research funding from Pfizer, through a grant peer-reviewed, co-funded and awarded by the Melanoma Research Alliance.

Figures

Ext. Figure 1.. Flow cytometry analysis of…
Ext. Figure 1.. Flow cytometry analysis of autophagic flux reporter with autophagy inhibitors and inducers.
a-e: Autophagic flux was assessed by flow cytometry in Mia-PaCa2AFR cells following 48 hours treatment with control, chloroquine (CQ), SAR-405, temsirolimus, or trametinib. Experiments were repeated three times with similar results. f: Autophagic flux was assessed by fluorescent imaging in Mia-PaCa2AFR cells following 48 hours treatment with control, chloroquine (CQ), VPS34i (SAR-405), or trametinib. Experiments were repeated three times with similar results.
Ext. Figure 2.. Inhibition of RAS→RAF→MEK→ERK signaling…
Ext. Figure 2.. Inhibition of RAS→RAF→MEK→ERK signaling pathway induces autophagic flux (AF) as seen by p62 degradation and LC3 conversion in pancreatic cancer cells.
a & b: Cell lysates prepared from Mia-PaCa2 (a) or BxPC3 (b) cells treated with 0.1–100 nM of trametinib for 48 hours were analyzed by immunoblotting for the phosphorylation (p) or total (t) abundance of ERK1/2, p62, LC3, or actin as indicated. Experiments were repeated three times with similar results. c: Cell lysates prepared from Mia-PaCa2 cells treated with ARS-853 (KRASG12Ci), SCH772984 (ERKi), or cobimetinib (MEKi) for 48 hours were analyzed by immunoblotting for the phosphorylation (p) or total (t) abundance of ERK1/2, p62, LC3, or actin as indicated. Experiments were repeated three times with similar results. d: Cell lysates prepared from Mia-PaCa2AFR cells transiently expressing exogenous ULK1 WT, ULKM92A (dominant negative), AMPK WT, or AMPKK45R (dominant negative) were analyzed by immunoblotting for ULK1, AMPK, or actin as indicated. Experiments were repeated three times with similar results. e: Cell lysates prepared from Mia-PaCa2AFR cells lentivirally transduced with shRNAs targeting LKB1 or scrambled control were analyzed by immunoblotting for LKB1 or actin as indicated. Experiments were repeated three times with similar results.
Ext. Figure 3.. Trametinib and chloroquine are…
Ext. Figure 3.. Trametinib and chloroquine are synergistically cytotoxic in vitro.
Mia-PaCa2 cells, BxPC3 and PDX220 cells were treated for 48–96 as indicated with trametinib and chloroquine and analyzed for cell viability by ATPlite assay. Synergy scores were generated utilizing Combenefit Software. Experiments were repeated four times with similar results.
Ext. Figure 4.. Treatment of pancreatic tumors…
Ext. Figure 4.. Treatment of pancreatic tumors with trametinib and chloroquine results in decreased pERK and increased p62 abundance respectively.
Representative images of immunohistochemical analysis of sections of PDX 227 tumors that were treated with 1. vehicle (Control), 2. trametinib; 3. chloroquine or; 4. the combination of both agents. Sections were stained with H&E or with antisera against pERK1/2 or p62 as indicated. Experiments were repeated four times with similar results. Scale bar is 500 μM located in the bottom right of the upper left panel and is consistent for all images.
Ext. Figure 5.. Treatment of orthopically xenografted…
Ext. Figure 5.. Treatment of orthopically xenografted pancreatic tumors with trametinib and hydroxychloroquine demonstrates regression consistent with subcutaneous xenografts.
a: PDX220 tumors were orthotopically transplanted and after 3 weeks were imaged via FDG-PET/CT for baseline. They were then treated with trametinib, hydroxychloroquine or trametinib plus hydroxychloroquine for 2 weeks prior to re-imaging. n=3 for control; n=2 for HCQ; n=3 for trametinib; n=2 for trametinib+hydroxychloroquine. b & c: Quantification of total lesion glycolysis (b) and % change (c) for individual tumors within each treatment group.
Ext. Figure 6.. Regression of established NRAS…
Ext. Figure 6.. Regression of established NRAS driven melanoma tumors by combined inhibition of MEK1/2 plus chloroquine.
a. The growth of NRAS-mutated melanoma (NCI515677) PDX was assessed over 21 days in mice treated with: 1. vehicle (Control), 2. trametinib (1mg/kg), 3. chloroquine (25mg/kg) or; 4. the combination of both agents at the aforementioned doses as indicated. n=4 for all treatment groups except combination of both agents n=5. Center values are the mean; statistical testing was performed by two-sided t-test; ***p<0.001 vs. control; tttp<0.001 vs. trametinib. Error bars represent SD. b-e. The percentage weight change of HCI-Mel002 NRAS-mutated PDX was assessed over 21 days in mice treated with: b. vehicle (Control), c. trametinib (1mg/kg), d. chloroquine (50mg/kg) or; e. the combination of both agents at the aforementioned doses as indicated. However, side-effects of facial rash and hair loss were noted.
Ext. Figure 7.. Lack of autophagy induction…
Ext. Figure 7.. Lack of autophagy induction by MEK1/2 inhibition results in resistance to combined trametinib and chloroquine treatment.
a: Cell lysates prepared from two suitably manipulated KRASG12D/TP53Null mouse lung cancer-derived cell lines, SC196 or SC274 treated with 100nM of trametinib were analyzed by immunoblotting for the phosphorylation (p) or total (t) abundance of ERK1/2, p62, LC3, or actin as indicated. Experiments were repeated three times with similar results. b: SC196 and SC274 KRASG12D/TP53Null mouse lung cancer cells were treated for 48 hours respectively with trametinib and chloroquine and analyzed for cell viability by ATPlite assay. Synergy scores were generated utilizing Combenefit Software. Experiments were repeated four times with similar results. c & d: Autophagic flux was measured in SC196AFR (c) or SC274AFR (d) following treatment with 0.1–1000nM trametinib for 48 hours.. n=3; center values are the mean; statistical testing was performed by two-sided t-test of control high (red) versus experimental high; ***p<0.001 vs. control. Error bars represent SD. e & f: The growth of SC196 (e) or SC274 (f) KRASG12D/TP53Null mouse lung cancer derived tumors in xenografted mice treated with: 1. vehicle (Control), 2. trametinib (1mg/kg), 3. chloroquine (50mg/kg) or; 4. the combination of both agents was assessed over ~15 days as indicated. n=10 for all treatment groups.. Center values are the mean; statistical testing was performed by two-sided t-test;***p<0.001 vs. control; tttp<0.001 vs. trametinib. Error bars represent SD.
Figure 1.. Inhibition of RAS→RAF→MEK→ERK signaling pathway…
Figure 1.. Inhibition of RAS→RAF→MEK→ERK signaling pathway induces autophagic flux (AF) in pancreatic cancer cells
a: Pancreatic cancer cells expressing an autophagic flux reporter (AFR) were generated by ectopic expression of a chimeric fusion protein comprised of mCherry-EGFP-LC3 in which the increased ratio of red:green fluorescence assessed by flow cytomtery is indicative of elevated AF. b: Autophagic flux was assessed by flow cytometry in Mia-PaCa2AFR cells following 48 hours treatment with various pharmacological inhibitors (CQ 20 uM or SAR-405 10 uM) or inducers (temsirolimus 10 uM or trametinib 100 nM) of autophagy. n=3; center values are the mean; statistical testing was performed by two-sided t-test of control high (red) versus experimental high; ***p<0.001 vs. control (0 nM/uM). Error bars represent SD. c-h: Mia-PaCa2AFR, BxPC3AFR or PDX220AFR (derived from a human pancreatic cancer PDX) cells were treated for 48 hours with inhibitors of KRASG12C (ARS-853), ERK1/2 (SCH772984), or MEK1/2 (trametinib or cobimetinib) with autophagic flux assessed by flow cytometry. n=3; center values are the mean; statistical testing was performed by two-sided t-test of control high (red) versus experimental high; ***p<0.001, **p<0.01, *p<0.05 vs. control (0 nM/uM). Error bars represent SD. i & j: Cell lysates prepared from Mia-PaCa2 treated with 1–100nM trametinib for 48 hours (a) or PDX220 derived cells treated with 100nM of trametinib over a time course (b), were analyzed by immunoblotting for the phosphorylation (p) or total (t) abundance of ERK1/2, p62, LC3, LKB1 (pS428), AMPK (pT172), ULK1 (pS555) or actin as indicated. Experiments were repeated three times with similar results. k: Schematic model of the proposed mechanism by which inhibition of RAS→RAF→MEK→ERK signaling may elicit autophagic flux in pancreatic cancer cells. l & m: Autophagic flux was assessed by flow cytometry in Mia-PaCa2AFR cells transiently expressing exogenous ULK1 WT, ULK M92A (dominant negative) (d), AMPK WT, or AMPKK45R (dominant negative) (e) and treated with 1–100nM of trametinib for 48 hours. n=3; center values are the mean; statistical testing was performed by two-sided t-test of matched treatment control high (red) versus matched dominant negative treatment high or matched treatment WT versus matched dominant negative treatment high; ***p<0.001 vs. matched treatment control; tttp<0.001 vs. matched treatment WT. Error bars represent SD. n: Autophagic flux was assessed by flow cytometry in Mia-PaCa2AFR cells stably expressing shRNAs targeting LKB1 or scrambled control and treated with 1–100nM of trametinib for 48 hours. n=3; center values are the mean; statistical testing was performed by two-sided t-test; ***p<0.001 vs. matched treatment scrambled control. Error bars represent SD.
Figure 2.. Trametinib and chloroquine are synergistically…
Figure 2.. Trametinib and chloroquine are synergistically cytotoxic in vitro.
a: Mia-PaCa2, BxPC3 and PDX220 cells were treated for 48–96 as indicated with trametinib and chloroquine and analyzed for cell viability by ATPlite assay. Synergy graphs were generated utilizing Combenefit Software. Experiments were repeated 4 times with similar results. b-d: Mia-PaCa2, BxPC3 and PDX220 cells were treated for 48 hours as indicated with vehicle (Control; DMSO), trametinib 100nM, chloroquine (CQ) 20μM or trametinib plus chloroquine and analyzed for cell viability by CytoxRed assay using an Incucyte Microscope. n=3; center values are the mean; statistical testing was performed by one-way ANOVA; p

Figure 3.. Tumor cell autonomous inhibition of…

Figure 3.. Tumor cell autonomous inhibition of autophagy cooperates with MEK1/2 inhibition to elicit regression…

Figure 3.. Tumor cell autonomous inhibition of autophagy cooperates with MEK1/2 inhibition to elicit regression of xenografted pancreatic tumors
a: Mia-PaCa2AFR cells, engineered to express a doxycycline-regulated dominant-negative (DN) form of ATG4B (Mia-PaCa2AFR TetI-ATG4BDN) were treated with trametinib in the absence or presence of doxycycline with autophagic flux measured by flow cytometry. n=3; center values are the mean; statistical testing was performed by two-sided t-test of control high (red) versus experimental high; ***p<0.001 vs. trametinib treatment alone. Error bars represent SD. b: Immunoblot analysis of the expression of the autophagy indicator proteins p62 and LC3 in Mia-PaCa2AFR TetI-ATG4BDN treated with trametinib, doxycycline (to induce ATG4BDN expression) or both agents. This was repeated three times with similar results. c: The growth of xenografted tumors of Mia-PaCa2AFR/TetI-ATG4BDN cells was assessed over 20 days in mice treated with: 1. vehicle (Control) n=11; 2. Trametinib n=11; 3. Doxycycline n=10 or; 4. the combination of both agents n=12. Center values are the mean; statistical testing was performed by two-sided t-test; ***p<0.001 vs. control; tttp<0.001 vs. trametinib. Error bars represent SD. d: Representative images of immunohistochemical analysis of sections of xenografted Mia-PaCa2AFR TetI-ATG4BDN tumors that were treated with 1. vehicle (Control), 2. trametinib; 3. doxycycline or; 4. the combination of both agents. Sections were stained with H&E or with antisera against pERK1/2, ATG4B or p62 as indicated. Scale bar is 500 μM located in the bottom right of the upper left panel and is consistent for all images. e & f: The growth of tumor xenografts of Mia-PaCa2 (a) or BxPC3 (b) cells over ~60 days in mice treated with: 1. vehicle (Control); 2. trametinib (1mg/kg); 3. chloroquine (50mg/kg), or; 4. the combination of both agents at the aforementioned doses were assessed as indicated. Mia-PaCa2: control n=5, trametinib n=6, chloroquine n=5, combination of both agents n=4. BxPC3: n=6 for all treatment groups. Center values are the mean; statistical testing was performed by two-sided t-test; ***ptttp<0.001 vs. trametinib. Error bars represent SD. g & h: The growth of two pancreatic cancer patient derived xenografts (PDX220 or PDX227) in mice treated with: 1. vehicle (Control), 2. trametinib (1mg/kg), 3. hydroxychloroquine (40mg/kg in PDX220), chloroquine (50mg/kg in PDX227); 4. gemcitabine plus abraxane or; 5. the combination of trametinib plus CQ/HCQ at the aforementioned doses were assessed over ~30–40 days as indicated. PDX220: control n=6, trametinib n=5, hydroxychloroquine n=5, combination of both agents n=4, gemcitabine plus abraxane n=6. PDX227: n=5 for all groups except for gemcitabine plus abraxane n=6. Center values are the mean; statistical testing was performed by two-sided t-test; ***ptttp<0.001 vs. trametinib, xxxp<0.001, xxp<0.01 vs. gemcitabine plus abraxane. Error bars represent SD. i & j: The growth of NRAS-mutated melanoma (HCI-Mel002) PDX or a BRAF-mutated colorectal cancer PDX (HCI-CRC004) was assessed over 18–21 days in mice treated with: 1. vehicle (Control), 2. trametinib (1mg/kg), 3. chloroquine (50mg/kg) or; 4. the combination of both agents at the aforementioned doses as indicated. HCI-Mel002: control n=5, trametinib n=5, chloroquine n=4, combination of both agents n=4. HCI-CRC004: n=5 for all groups except combination of both agents n=4. Center values are the mean; statistical testing was performed by two-sided t-test; **p<0.01, *p<0.05 vs. control; tttp<0.001 vs. trametinib. Error bars represent SD.

Figure 4.. Treatment of a pancreatic cancer…

Figure 4.. Treatment of a pancreatic cancer patient with trametinib plus hydroxychloroquine (T/HCQ) lead to…

Figure 4.. Treatment of a pancreatic cancer patient with trametinib plus hydroxychloroquine (T/HCQ) lead to a reduction in tumor marker cancer antigen 19–9 (CA19–9) and overall tumor burden.
a: The patient’s blood-borne CA19–9 tumor marker was measured periodically throughout the the entire clinical course and is annotated with the dates and treatments administered. b, c, d & e: CT imaging 2 days after starting (b, d) the 2mg trametinib (q.d.) plus 1200mg (600mg b.i.d.) dosing of HCQ (which had been started with lower doses of HCQ two weeks previously) and two months post (c & e). The recurrent pancreatic bed lesion is dramatically reduced in size (comparing panel b to c), while the metastatic lesions in the liver are largely resolved (comparing panels b to c and d to e).
All figures (11)
Figure 3.. Tumor cell autonomous inhibition of…
Figure 3.. Tumor cell autonomous inhibition of autophagy cooperates with MEK1/2 inhibition to elicit regression of xenografted pancreatic tumors
a: Mia-PaCa2AFR cells, engineered to express a doxycycline-regulated dominant-negative (DN) form of ATG4B (Mia-PaCa2AFR TetI-ATG4BDN) were treated with trametinib in the absence or presence of doxycycline with autophagic flux measured by flow cytometry. n=3; center values are the mean; statistical testing was performed by two-sided t-test of control high (red) versus experimental high; ***p<0.001 vs. trametinib treatment alone. Error bars represent SD. b: Immunoblot analysis of the expression of the autophagy indicator proteins p62 and LC3 in Mia-PaCa2AFR TetI-ATG4BDN treated with trametinib, doxycycline (to induce ATG4BDN expression) or both agents. This was repeated three times with similar results. c: The growth of xenografted tumors of Mia-PaCa2AFR/TetI-ATG4BDN cells was assessed over 20 days in mice treated with: 1. vehicle (Control) n=11; 2. Trametinib n=11; 3. Doxycycline n=10 or; 4. the combination of both agents n=12. Center values are the mean; statistical testing was performed by two-sided t-test; ***p<0.001 vs. control; tttp<0.001 vs. trametinib. Error bars represent SD. d: Representative images of immunohistochemical analysis of sections of xenografted Mia-PaCa2AFR TetI-ATG4BDN tumors that were treated with 1. vehicle (Control), 2. trametinib; 3. doxycycline or; 4. the combination of both agents. Sections were stained with H&E or with antisera against pERK1/2, ATG4B or p62 as indicated. Scale bar is 500 μM located in the bottom right of the upper left panel and is consistent for all images. e & f: The growth of tumor xenografts of Mia-PaCa2 (a) or BxPC3 (b) cells over ~60 days in mice treated with: 1. vehicle (Control); 2. trametinib (1mg/kg); 3. chloroquine (50mg/kg), or; 4. the combination of both agents at the aforementioned doses were assessed as indicated. Mia-PaCa2: control n=5, trametinib n=6, chloroquine n=5, combination of both agents n=4. BxPC3: n=6 for all treatment groups. Center values are the mean; statistical testing was performed by two-sided t-test; ***ptttp<0.001 vs. trametinib. Error bars represent SD. g & h: The growth of two pancreatic cancer patient derived xenografts (PDX220 or PDX227) in mice treated with: 1. vehicle (Control), 2. trametinib (1mg/kg), 3. hydroxychloroquine (40mg/kg in PDX220), chloroquine (50mg/kg in PDX227); 4. gemcitabine plus abraxane or; 5. the combination of trametinib plus CQ/HCQ at the aforementioned doses were assessed over ~30–40 days as indicated. PDX220: control n=6, trametinib n=5, hydroxychloroquine n=5, combination of both agents n=4, gemcitabine plus abraxane n=6. PDX227: n=5 for all groups except for gemcitabine plus abraxane n=6. Center values are the mean; statistical testing was performed by two-sided t-test; ***ptttp<0.001 vs. trametinib, xxxp<0.001, xxp<0.01 vs. gemcitabine plus abraxane. Error bars represent SD. i & j: The growth of NRAS-mutated melanoma (HCI-Mel002) PDX or a BRAF-mutated colorectal cancer PDX (HCI-CRC004) was assessed over 18–21 days in mice treated with: 1. vehicle (Control), 2. trametinib (1mg/kg), 3. chloroquine (50mg/kg) or; 4. the combination of both agents at the aforementioned doses as indicated. HCI-Mel002: control n=5, trametinib n=5, chloroquine n=4, combination of both agents n=4. HCI-CRC004: n=5 for all groups except combination of both agents n=4. Center values are the mean; statistical testing was performed by two-sided t-test; **p<0.01, *p<0.05 vs. control; tttp<0.001 vs. trametinib. Error bars represent SD.
Figure 4.. Treatment of a pancreatic cancer…
Figure 4.. Treatment of a pancreatic cancer patient with trametinib plus hydroxychloroquine (T/HCQ) lead to a reduction in tumor marker cancer antigen 19–9 (CA19–9) and overall tumor burden.
a: The patient’s blood-borne CA19–9 tumor marker was measured periodically throughout the the entire clinical course and is annotated with the dates and treatments administered. b, c, d & e: CT imaging 2 days after starting (b, d) the 2mg trametinib (q.d.) plus 1200mg (600mg b.i.d.) dosing of HCQ (which had been started with lower doses of HCQ two weeks previously) and two months post (c & e). The recurrent pancreatic bed lesion is dramatically reduced in size (comparing panel b to c), while the metastatic lesions in the liver are largely resolved (comparing panels b to c and d to e).

References

    1. Siegel RL, Miller KD, and Jemal A, Cancer Statistics, 2017. CA Cancer J Clin, 2017. 67(1): p. 7–30.
    1. Li D, et al., Pancreatic cancer. Lancet, 2004. 363(9414): p. 1049–57.
    1. Gao J, et al., Integrative analysis of complex cancer genomics and clinical profiles using the cBioPortal. Sci Signal, 2013. 6(269): p. pl1.
    1. Cerami E, et al., The cBio cancer genomics portal: an open platform for exploring multidimensional cancer genomics data. Cancer Discov, 2012. 2(5): p. 401–4.
    1. Collisson EA, et al., A central role for RAF-->MEK-->ERK signaling in the genesis of pancreatic ductal adenocarcinoma. Cancer Discov, 2012. 2(8): p. 685–93.
    1. Infante JR, et al., A randomised, double-blind, placebo-controlled trial of trametinib, an oral MEK inhibitor, in combination with gemcitabine for patients with untreated metastatic adenocarcinoma of the pancreas. Eur J Cancer, 2014. 50(12): p. 2072–81.
    1. Kimura S, Noda T, and Yoshimori T, Dissection of the autophagosome maturation process by a novel reporter protein, tandem fluorescent-tagged LC3. Autophagy, 2007. 3(5): p. 452–60.
    1. Gump JM and Thorburn A, Sorting cells for basal and induced autophagic flux by quantitative ratiometric flow cytometry. Autophagy, 2014. 10(7): p. 1327–34.
    1. Klionsky DJ, et al., Guidelines for the use and interpretation of assays for monitoring autophagy (3rd edition). Autophagy, 2016. 12(1): p. 1–222.
    1. Ronan B, et al., A highly potent and selective Vps34 inhibitor alters vesicle trafficking and autophagy. Nat Chem Biol, 2014. 10(12): p. 1013–9.
    1. Patricelli MP, et al., Selective Inhibition of Oncogenic KRAS Output with Small Molecules Targeting the Inactive State. Cancer Discov, 2016. 6(3): p. 316–29.
    1. Gilmartin AG, et al., GSK1120212 (JTP-74057) is an inhibitor of MEK activity and activation with favorable pharmacokinetic properties for sustained in vivo pathway inhibition. Clin Cancer Res, 2011. 17(5): p. 989–1000.
    1. Rice KD, et al., Novel Carboxamide-Based Allosteric MEK Inhibitors: Discovery and Optimization Efforts toward XL518 (GDC-0973). ACS Med Chem Lett, 2012. 3(5): p. 416–21.
    1. Morris EJ, et al., Discovery of a novel ERK inhibitor with activity in models of acquired resistance to BRAF and MEK inhibitors. Cancer Discov, 2013. 3(7): p. 742–50.
    1. Lito P, et al., Allele-specific inhibitors inactivate mutant KRAS G12C by a trapping mechanism. Science, 2016. 351(6273): p. 604–8.
    1. Zheng B, et al., Oncogenic B-RAF negatively regulates the tumor suppressor LKB1 to promote melanoma cell proliferation. Mol Cell, 2009. 33(2): p. 237–47.
    1. Kim J, et al., AMPK and mTOR regulate autophagy through direct phosphorylation of Ulk1. Nat Cell Biol, 2011. 13(2): p. 132–41.
    1. Egan DF, et al., Phosphorylation of ULK1 (hATG1) by AMP-activated protein kinase connects energy sensing to mitophagy. Science, 2011. 331(6016): p. 456–61.
    1. Mihaylova MM and Shaw RJ, The AMPK signalling pathway coordinates cell growth, autophagy and metabolism. Nat Cell Biol, 2011. 13(9): p. 1016–23.
    1. Chou TC, Drug combination studies and their synergy quantification using the Chou-Talalay method. Cancer Res, 2010. 70(2): p. 440–6.
    1. Vessoni AT, et al., Chloroquine-induced glioma cells death is associated with mitochondrial membrane potential loss, but not oxidative stress. Free Radic Biol Med, 2016. 90: p. 91–100.
    1. Boya P, et al., Mitochondrial membrane permeabilization is a critical step of lysosome-initiated apoptosis induced by hydroxychloroquine. Oncogene, 2003. 22(25): p. 3927–36.
    1. Fujita N, et al., An Atg4B mutant hampers the lipidation of LC3 paralogues and causes defects in autophagosome closure. Mol Biol Cell, 2008. 19(11): p. 4651–9.
    1. Yang A, et al., Autophagy Sustains Pancreatic Cancer Growth through Both Cell-Autonomous and Nonautonomous Mechanisms. Cancer Discov, 2018. 8(3): p. 276–287.
    1. Awasthi N, et al., Comparative benefits of Nab-paclitaxel over gemcitabine or polysorbate-based docetaxel in experimental pancreatic cancer. Carcinogenesis, 2013. 34(10): p. 2361–9.
    1. Young NP, Crowley D, and Jacks T, Uncoupling cancer mutations reveals critical timing of p53 loss in sarcomagenesis. Cancer Res, 2011. 71(11): p. 4040–7.
    1. Levy JMM, Towers CG, and Thorburn A, Targeting autophagy in cancer. Nat Rev Cancer, 2017. 17(9): p. 528–542.
    1. Levy JM, et al., Autophagy inhibition improves chemosensitivity in BRAF(V600E) brain tumors. Cancer Discov, 2014. 4(7): p. 773–80.
    1. Mulcahy Levy JM, et al., Autophagy inhibition overcomes multiple mechanisms of resistance to BRAF inhibition in brain tumors. Elife, 2017. 6.
    1. Ma XH, et al., Targeting ER stress-induced autophagy overcomes BRAF inhibitor resistance in melanoma. J Clin Invest, 2014. 124(3): p. 1406–17.
    1. Eng CH, et al., Macroautophagy is dispensable for growth of KRAS mutant tumors and chloroquine efficacy. Proc Natl Acad Sci U S A, 2016. 113(1): p. 182–7.
    1. Rosenfeldt MT, et al., p53 status determines the role of autophagy in pancreatic tumour development. Nature, 2013. 504(7479): p. 296–300.
    1. Sousa CM, et al., Pancreatic stellate cells support tumour metabolism through autophagic alanine secretion. Nature, 2016. 536(7617): p. 479–83.
    1. Poillet-Perez L, et al., Autophagy maintains tumour growth through circulating arginine. Nature, 2018. 563(7732): p. 569–573.
    1. Honda A, et al., Potent, Selective, and Orally Bioavailable Inhibitors of VPS34 Provide Chemical Tools to Modulate Autophagy in Vivo. ACS Med Chem Lett, 2016. 7(1): p. 72–6.
    1. Egan DF, et al., Small Molecule Inhibition of the Autophagy Kinase ULK1 and Identification of ULK1 Substrates. Mol Cell, 2015. 59(2): p. 285–97.
    1. Kim KB, et al., Phase II study of the MEK1/MEK2 inhibitor Trametinib in patients with metastatic BRAF-mutant cutaneous melanoma previously treated with or without a BRAF inhibitor. J Clin Oncol, 2013. 31(4): p. 482–9.
    1. White NJ, The treatment of malaria. N Engl J Med, 1996. 335(11): p. 800–6.
    1. Ben-Zvi I, et al., Hydroxychloroquine: from malaria to autoimmunity. Clin Rev Allergy Immunol, 2012. 42(2): p. 145–53.
    1. Di Veroli GY, et al., Combenefit: an interactive platform for the analysis and visualization of drug combinations. Bioinformatics, 2016. 32(18): p. 2866–8.

Source: PubMed

3
Abonnieren