Sorafenib is U.S. Food and Drug Adminstration–approved for the treatment of renal cell carcinoma and hepatocellular carcinoma and has been combined with numerous other targeted therapies and chemotherapies in the treatment of many cancers. Unfortunately, as with other RAF inhibitors, patients treated with sorafenib have a 5% to 10% rate of developing cutaneous squamous cell carcinoma (cSCC)/keratoacanthomas. Paradoxical activation of extracellular signal–regulated kinase (ERK) in BRAF wild-type cells has been implicated in RAF inhibitor–induced cSCC. Here, we report that sorafenib suppresses UV-induced apoptosis specifically by inhibiting c-jun–NH2–kinase (JNK) activation through the off-target inhibition of leucine zipper and sterile alpha motif–containing kinase (ZAK). Our results implicate suppression of JNK signaling, independent of the ERK pathway, as an additional mechanism of adverse effects of sorafenib. This has broad implications for combination therapies using sorafenib with other modalities that induce apoptosis. Mol Cancer Ther; 13(1); 221–9. ©2013 AACR.

Sorafenib is a multikinase inhibitor originally designed to target CRAF, but has been found to effectively inhibit multiple kinases, including BRAF, VEGFR2, VEGFR3, PDGFR-β, FLT-3, and c-KIT (1, 2). In multiple clinical trials, sorafenib was well tolerated, but commonly associated with dermatologic toxicities, including cutaneous squamous cell carcinoma (cSCC) and keratoacanthomas (3–7).

All RAF inhibitors tested in clinical trials, including vemurafenib, dabrafenib, and sorafenib, induce hyperproliferative epidermal lesions, including cSCC and keratoacanthoma. Vemurafenib seems to have the highest rate: vemurafenib causes keratoacanthomas and SCCs in approximately 22% of patients (8–10), whereas sorafenib causes lesions in about 7% of patients (4, 6). These cutaneous toxicities associated with RAF inhibitors have been attributed to paradoxical extracellular signal–regulated kinase (ERK) activation, in which BRAF wild-type cells paradoxically activate MAP–ERK kinase (MEK) and ERK activity through increased CRAF activation (11–15). Consistent with this notion, RAS mutations are overrepresented in these lesions (particularly for vemurafenib), and combined MEK inhibitor (MEKi) therapy partially suppresses lesion formation (16–19).

However, suppression by MEKi may simply reflect an intrinsic need for MEK signaling in this tumor type and paradoxical ERK activation by sorafenib is short-lived for only a few hours, with modest effects on phospho-ERK levels and proliferation in nontarget tissues such as keratinocytes in human skin (5). Furthermore, although sorafenib-induced lesions have RAS mutations (5), a powerful enabler of paradoxical ERK activation, they were found in a small minority of samples, significantly less frequently than in vemurafenib-induced lesions (16, 17). Therefore, we sought to identify whether there might be other mechanisms contributing to sorafenib-induced cSCC.

Cells

HaCaT cells were originally obtained from Norbert Fusenig (German Cancer Research Center, DKFZ, Heidelberg, Germany) through Stephen Ullrich (MD Anderson Cancer Center, Houston, TX) in March 2011. These cells were authenticated twice, in March 2011 and in May 2012 by short tandem repeat (STR) DNA fingerprinting using the AmpFLSTR Identifiler Kit according to the manufacturer's instructions (Applied Biosystems; cat 4322288). The STR profiles were compared with known American Type Culture Collection (ATCC) fingerprints (ATCC.org), to the Cell Line Integrated Molecular Authentication database (CLIMA) version 0.1.200808 (http://bioinformatics.istge.it/clima/; Nucleic Acids Research 37:D925-D932 PMCID: PMC2686526) and to the MD Anderson Cancer Center fingerprint database. Normal human epidermal keratinocytes (NHEK) were obtained from Lonza in June 2012, aliquoted, and used according to the manufacturer's instructions, and not further tested.

Cell culture and UV irradiation

HaCaT cells were cultured in Dulbecco's Modified Eagle Medium (DMEM)/Ham's F-12 50/50 (Cellgro) supplemented with 10% FBS (Sigma), glutamine, and Primocin (Invivogen). NHEKs (Lonza) were cultured in media according to the manufacturer's instructions. Irradiation was performed using an FS40 sunlamp dosed by an IL1700 radiometer. Following irradiation, cells were treated with sorafenib (LC Laboratories) or dimethyl sulfoxide (DMSO; 1:2,000).

Flow cytometry

Adherent cells were trypsinized and nonadherant cells were collected for staining with Annexin V, TMRE (tetramethylrhodamine), and Cytox Blue. TMRE (Invitrogen) was used as a measure of mitochondrial membrane potential, Annexin V–APC (Invitrogen) as a probe for apoptosis, and Cytox Blue (Invitrogen) as an indicator for dead cells. Data were collected and analyzed using a flow cytometer (FACScalibur; Becton Dickinson) and FlowJo Software (Tree Star). Data were calculated and charts were plotted using GraphPad Prism 5 software.

Western blot analysis

Cell were lysed in standard buffers with protease inhibitors (Roche) and phosphatase inhibitors (Santa Cruz Biotechnology) with extracts run on SDS/PAGEs and transferred to Immobilon-P transfer membrane (Millipore). Western blot analysis was blocked in TBST (10 mmol/L Tris-HCL pH8, 150 mmol/L NaCl, 0.5% Tween) containing 2.5% bovine serum albumin, probed with primary antibodies (all Cell Signaling Technology), corresponding horseradish peroxidase–conjugated secondary antibodies, and signals detected using the ECL Kit (Amersham).

Lentiviral knockdown experiments

Lentiviral short hairpin RNA (shRNA) knockdown was accomplished using standard lentiviral methods using 293T cells and psPAX2 and VSVG packaging plasmids. shRNA clones against leucine zipper and sterile alpha motif–containing kinase (ZAK; clones: V2LHS_239842; V3LHS_336769) and TAOK2 (clone: V3LHS_315551), as well as a nonsilencing shRNA were obtained from Open Biosystems in the GIPZ vector. Following transduction, cells were puromycin-selected to obtain a stably infected population and fluorescence-activated cell sorted (FACS) to obtain cells with high-level suppression. Degree of mRNA suppression was quantified by quantitative PCR using TaqMan probes using internally controlled (two-color, same well) GAPDH (glyceraldehyde-3-phosphate dehydrogenase) probes to ensure proper normalization.

Mice experiments

For chronically irradiated SKH-1E hairless mice (Charles River Laboratories), 3-month-old female mice were irradiated thrice weekly for a total weekly dose of 12.5 kJ/m2 UVB (solar simulator; Oriel). At 72 days, sorafenib treatment was started by oral gavage (12.5% Cremphor, 12.5% ethanol in water) at 50 mg/kg/d.

Immunohistochemistry

cSCCs biopsied from patients treated with or without sorafenib were obtained under Institutional Review Board approval (LAB08-0750). Staining levels were quantified by counting positively labeled cells and dividing by the total area of the tumor tissue (malignant keratinocytes) within each sample. To measure tumor areas, all samples were photographed, tumor cells outlined, and total pixel numbers calculated using image analysis tools in Adobe Photoshop and standardized to a hemacytometer to convert to mm2.

Soft agar assays

For growth assays, transformed and immortal cells were generated by retroviral infection. Wild-type (WT) mouse embryonic fibroblasts (MEF) and Craf-deficient (Craf−/−) MEFs infected with HRAS (pBABE puro HRASV12) and E1A (pLS adenovirus E1A-12S) expressing lentiviruses were selected with puromycin. Cells were seeded in soft agar at 2 × 103 in 24-well plate format in growth medium containing 0.3% agar over a base layer of 0.6% agar, with varying concentrations of sorafenib or with DMSO (Sigma-Aldrich) vehicle control.

Sorafenib suppresses UV-induced apoptosis in keratinocytes

Because many of the cSCC/keratoacanthoma induced by RAFi occur on sun-exposed areas, we hypothesized instead that sorafenib could affect UV-induced apoptosis. When we exposed HaCaT keratinocyte cell lines and primary NHEKs to 500 J/m2 of UVB, they underwent significant apoptosis by 24 hours (Fig. 1A and B) as assessed by FACS for Annexin V+, TMRE-low cells. Treatment with 1 μmol/L sorafenib resulted in significant suppression of apoptosis as compared DMSO-treated controls, by 40% for HaCaT cells (51.0% ± 3.6% vs. 30.6% ± 4.1%) and 81% for NHEKs (34.5% ± 1.8% vs. 6.6% ± 0.5%; Fig. 1A and B). This suppression of apoptosis could also be observed by assessing cleaved caspase-3 levels by Western blot analysis (Supplementary Fig. S1).

Figure 1.

Sorafenib suppresses UV-induced apoptosis. HaCaT and NHEK cells were either unirradiated or irradiated with 500 J/m2 of UVB in the absence (o, 1:2,000 DMSO) or presence (+) of 1 μmol/L sorafenib and isolated for FACS analysis and protein extracts 24 hours later. HaCaT (A) and NHEK (B)cells show 45% and 80% suppression, respectively, of apoptosis in the presence of sorafenib as measured by FACS for Annexin V+, TMRE-low cells (n = 6 for each condition; ***, P < 0.001). Western blot analysis of HaCaT (C) and NHEK (D) cells probed for the MAPKs JNK and ERK demonstrated strong phospho-JNK induction following irradiation and significant suppression by sorafenib. Phospho-ERK was slightly suppressed following irradiation, and at all time points, paradoxical hyperactivation in the presence of sorafenib was observed, particularly in HaCaT cells. Importantly, phospho-MKK4 and phospho-MKK7, upstream kinases that activate JNK, were both strongly induced by UV radiation and strongly suppressed by sorafenib.

Figure 1.

Sorafenib suppresses UV-induced apoptosis. HaCaT and NHEK cells were either unirradiated or irradiated with 500 J/m2 of UVB in the absence (o, 1:2,000 DMSO) or presence (+) of 1 μmol/L sorafenib and isolated for FACS analysis and protein extracts 24 hours later. HaCaT (A) and NHEK (B)cells show 45% and 80% suppression, respectively, of apoptosis in the presence of sorafenib as measured by FACS for Annexin V+, TMRE-low cells (n = 6 for each condition; ***, P < 0.001). Western blot analysis of HaCaT (C) and NHEK (D) cells probed for the MAPKs JNK and ERK demonstrated strong phospho-JNK induction following irradiation and significant suppression by sorafenib. Phospho-ERK was slightly suppressed following irradiation, and at all time points, paradoxical hyperactivation in the presence of sorafenib was observed, particularly in HaCaT cells. Importantly, phospho-MKK4 and phospho-MKK7, upstream kinases that activate JNK, were both strongly induced by UV radiation and strongly suppressed by sorafenib.

Close modal

The stress-activated c-jun–NH2–kinase N-terminal (JNK) mitogen-activated protein kinase (MAPK) pathway is critically important in UV-induced apoptosis (20), so we explored its activation in cells treated with and without sorafenib after UV irradiation. Phospho-JNK levels were highly induced in DMSO-treated cells following UV irradiation, and potently suppressed in the presence of 1 μmol/L sorafenib in both HaCaT (Fig. 1C) and NHEK (Fig. 1D) cells. Phosphorylation of MKK4 (MAP2K4) and MKK7 (MAP2K7), two kinases required for the activation of JNK (21–23), were also upregulated significantly by UV treatment particularly at 1 and 6 hours (lanes 3, 5, and 7; Fig. 1C and D) and suppressed by drug treatment (lanes 4, 6, and 8; Fig. 1C). ERK signaling, as seen by phospho-ERK levels, remained intact, with paradoxical hyperactivation following sorafenib treatment (Fig. 1C and D).

Sorafenib suppresses apoptosis and JNK signaling independently of ERK activation

Because sorafenib exerts potent effects on ERK signaling (5, 14), we sought to probe whether its suppression of JNK-dependent apoptosis could be the result of cross talk between ERK and JNK signaling. Because sorafenib induces a dose-dependent activation of the MAPK pathway in BRAF wild-type cells (5, 14), we separated the effects on ERK signaling by simultaneously treating cells with the MEK inhibitor selumetinib (24). NHEK cells and HaCaT cells were treated with sorafenib singly and in combination with selumetinib and then UV irradiated. Sorafenib suppressed UV-induced apoptosis strongly, regardless of whether selumetinib was present (Fig. 2A and B). Selumetinib did not affect sorafenib-mediated suppression of apoptosis or JNK activation (lanes 5–8; Fig. 2C and D) despite potent abrogation of ERK phosphorylation (lanes 3–4, 7–8; Fig. 2C and D).

Figure 2.

Sorafenib suppresses apoptosis and JNK signaling independently of MEK/ERK signaling. To test the relevance of MEK/ERK signaling in the suppression of JNK-mediated apoptosis, HaCaT and NHEK cells were irradiated with 500 J/m2 of UVB in the absence (“o,” 1:2,000 DMSO) or presence of 1 μmol/L sorafenib singly or in combination with 0.6 μmol/L (NHEK) or 1.2 μmol/L (HaCaT) selumetinib (MEKi) and isolated for FACS analysis and protein extracts 24 hours later. HaCaT (A) and NHEK (B) cells exhibited a strong suppression of UV-induced apoptosis by sorafenib (Annexin V+, TMRE-low cells; n = 6 for each condition; ***, P < 0.001) that was not substantially by the addition of MEKi. HaCaT (C) and NHEK (D) cells showed induction of phospho-JNK at 24 hours following irradiation, by Western blot analysis in the presence (lane 7) and absence (lane 5) of MEKi. The addition of MEKi with sorafenib did not affect the suppression of JNK activation (compare lanes 7 vs. 8) despite potent suppression of phospho-ERK.

Figure 2.

Sorafenib suppresses apoptosis and JNK signaling independently of MEK/ERK signaling. To test the relevance of MEK/ERK signaling in the suppression of JNK-mediated apoptosis, HaCaT and NHEK cells were irradiated with 500 J/m2 of UVB in the absence (“o,” 1:2,000 DMSO) or presence of 1 μmol/L sorafenib singly or in combination with 0.6 μmol/L (NHEK) or 1.2 μmol/L (HaCaT) selumetinib (MEKi) and isolated for FACS analysis and protein extracts 24 hours later. HaCaT (A) and NHEK (B) cells exhibited a strong suppression of UV-induced apoptosis by sorafenib (Annexin V+, TMRE-low cells; n = 6 for each condition; ***, P < 0.001) that was not substantially by the addition of MEKi. HaCaT (C) and NHEK (D) cells showed induction of phospho-JNK at 24 hours following irradiation, by Western blot analysis in the presence (lane 7) and absence (lane 5) of MEKi. The addition of MEKi with sorafenib did not affect the suppression of JNK activation (compare lanes 7 vs. 8) despite potent suppression of phospho-ERK.

Close modal

Multiple upstream kinases of JNK are inhibited by sorafenib

We then sought to determine how sorafenib suppresses JNK activation and apoptosis. Given that MKK4 and MKK7 activation were also suppressed at 1 μmol/L sorafenib (Fig. 1C and D), we reasoned that sorafenib must affect upstream kinases. Comprehensive kinase profiling of sorafenib has been reported using a quantitative binding assay platform (25, 26); therefore, we performed in vitro kinase assays against a panel of 38 kinases reported to be upstream of JNK (21, 27) and other kinases previously tested against sorafenib using a 10-dose, 3-fold serial dilution starting at 10 μmol/L (Supplementary Table S1). None of the JNKs is directly inhibited.

We identified ZAK, TAO1/TAOK2, MAPK14 (p38α), and MAPK11 (p38β) as having low in vitro IC50 values below 500 nmol/L (Supplementary Table S1). Using p38 inhibitors BIRB796 and LY2228820, we were unable to observe any suppression of UV-induced apoptosis (data not shown). Therefore, we focused on ZAK and TAOK2, which have the lowest biochemical IC50 values of 48.6 and 59.8 nmol/L, respectively (Fig. 3A). These IC50 values are in the same nanomolar range as those previously reported for BRAF and CRAF (2) and in our panel of assays (Supplementary Table S1).

Figure 3.

Sorafenib exhibits significant off-target effects on ZAK and TAOK2 kinases. A, in vitro kinase assays for ZAK and TAOK2 were performed across a 10-point concentration range from 0.03 to 10 μmol/L, revealed significant inhibition of kinase activity within the nmol/L range for sorafenib. B, lentiviral shRNA knockdown of ZAK singly (clones ZAK-1 and ZAK-4) or in combination with TAOK2 was performed, with both exhibiting significant knockdown of ZAK protein. Knockdown of TAOK2 was quantitated using TaqMan qRT-PCR, showing 91% knockdown (***, P < 0.001). C, lentiviral shRNA knockdown of ZAK singly (two clones ZAK-1 and ZAK-4) or in combination with TAOK2 (ZAK-1 only) was performed, revealing potent suppression of apoptosis as measured by FACS for Annexin V+, TMRE-low cells (n = 9; **, P < 0.01; ***, P < 0.001; NS, not significant) at 24 hours following single dose UVB irradiation at 1 kJ/m2. ZAK single knockdowns exhibited 38% (ZAK-1) and 59% (ZAK-4) suppression, whereas ZAK/TAOK2 double knockdown cells and drug-treated cells exhibited 50% and 55% suppression of apoptosis, respectively. Differences between the ZAK-4 shRNA single knockdown, ZAK/TAOK2 double knockdown, and sorafenib-treated scramble (“SCR”) cells were not statistically significant. D, Western blot analysis of lysates obtained at 1 and 6 hours after UV irradiation show potent induction of phospho-JNK following UV irradiation (both lanes 5). ZAK-1 shRNA single knockdown produced suppression of phospho-JNK activation comparable with those of ZAK/TAOK2 double knockdown and sorafenib treatment (lanes 6–8).

Figure 3.

Sorafenib exhibits significant off-target effects on ZAK and TAOK2 kinases. A, in vitro kinase assays for ZAK and TAOK2 were performed across a 10-point concentration range from 0.03 to 10 μmol/L, revealed significant inhibition of kinase activity within the nmol/L range for sorafenib. B, lentiviral shRNA knockdown of ZAK singly (clones ZAK-1 and ZAK-4) or in combination with TAOK2 was performed, with both exhibiting significant knockdown of ZAK protein. Knockdown of TAOK2 was quantitated using TaqMan qRT-PCR, showing 91% knockdown (***, P < 0.001). C, lentiviral shRNA knockdown of ZAK singly (two clones ZAK-1 and ZAK-4) or in combination with TAOK2 (ZAK-1 only) was performed, revealing potent suppression of apoptosis as measured by FACS for Annexin V+, TMRE-low cells (n = 9; **, P < 0.01; ***, P < 0.001; NS, not significant) at 24 hours following single dose UVB irradiation at 1 kJ/m2. ZAK single knockdowns exhibited 38% (ZAK-1) and 59% (ZAK-4) suppression, whereas ZAK/TAOK2 double knockdown cells and drug-treated cells exhibited 50% and 55% suppression of apoptosis, respectively. Differences between the ZAK-4 shRNA single knockdown, ZAK/TAOK2 double knockdown, and sorafenib-treated scramble (“SCR”) cells were not statistically significant. D, Western blot analysis of lysates obtained at 1 and 6 hours after UV irradiation show potent induction of phospho-JNK following UV irradiation (both lanes 5). ZAK-1 shRNA single knockdown produced suppression of phospho-JNK activation comparable with those of ZAK/TAOK2 double knockdown and sorafenib treatment (lanes 6–8).

Close modal

ZAK is critically important for JNK activation upstream of MKK4 and MKK7 (28) and is important for doxorubicin-induced apoptosis (29, 30). TAOK2 has been implicated as an activator of JNK as well (31–35). To examine the requirements for ZAK and TAOK2 in promoting JNK activation and apoptosis more directly, we carried out lentiviral shRNA knockdown experiments in HaCaT cells. HaCaT cells with more than 85% knockdown of ZAK by quantitative real-time PCR (qRT-PCR, TaqMan; Fig. 3B) were created using two lentiviral shRNA clones (pGIPZ; Thermo), ZAK-1 and ZAK-4. Cells expressing shRNA ZAK-1 were then infected to express shRNA against TAOK2 as well (Fig. 3B). Both ZAK single knockdown and ZAK/TAOK2 double knockdown cells lack detectable ZAK protein expression and ZAK/TAOK2 double knockdown cells showed 91% reduction in TAOK2 mRNA (TaqMan; Fig. 3B). Cells expressing shRNA ZAK-1 and ZAK-4 were substantially more resistant than scramble shRNA-expressing (SCR) cells to UV-induced apoptosis (Fig. 3C). Knockdown of ZAK alone essentially accounted for the entire effect of sorafenib on UV-induced apoptosis (Fig. 3C). ZAK-1-shRNA–expressing HaCaT cells showed statistically significant, but small differences in suppression of apoptosis as compared with ZAK/TAOK2 double knockdown (12%) and sorafenib-treated SCR cells (17%; Fig. 3C). Importantly, JNK activation was substantially suppressed in ZAK-1-shRNA and ZAK/TAOK2 double knockdown cells at 1 and 6 hours after irradiation (Fig. 3D). The suppression is most evident by treatment with 1 μmol/L sorafenib (lanes 6, Fig. 3D), but it is clear than both single ZAK knockdown and double ZAK/TAOK2 knockdown both result in significant suppression of phospho-JNK (lanes 7 and 8, Fig. 3D).

Sorafenib suppresses JNK activation and apoptosis in vivo

To address whether sorafenib-mediated suppression of JNK signaling and apoptosis occurs in vivo, we examined cSCC arising in patients treated with sorafenib and compared them with sporadic cSCC that were histologically similar, arising in individuals never treated with sorafenib (Fig. 4A and B). Phospho-JNK (Fig. 4A) and cleaved caspase-3 (Fig. 4B) expression were probed using immunohistochemistry and quantified following normalization by unit area (mm2) of tumor tissue (malignant keratinocytes) only (Fig. 4C). Sporadic cSCC arising in patients never treated with sorafenib (n = 9) had substantially greater expression of phospho-JNK (P = 0.0045; Fig. 4A and C) and cleaved caspase-3 (P = 0.003; Fig. 4B and C) as compared with lesions arising in sorafenib-treated patients (n = 13; Fig. 4A–C). Therefore, there are significant reductions in phospho-JNK (46%) and cleaved caspase-3 expression (88%) in human cSCC, showing that suppression of JNK activity and apoptosis occur in vivo in patients treated with sorafenib.

Figure 4.

Sorafenib suppresses apoptosis and JNK signaling in vivo. A and B, cSCC samples from sorafenib-treated patients and nontreated patients (sporadic, first row) were analyzed by immunohistochemistry for phospho-JNK and cleaved caspase-3 expression. cSCC arising in sorafenib-treated patients (A and B, second row) show decreased expression of phospho-JNK (A) and cleaved caspase-3 (B) as compared with sporadic cSCC in patients never treated with sorafenib (second row). Scale bar, 100 μm. C, comparisons of stained cells normalized to mm2 of tumor area revealed significant suppression of both phospho-JNK and cleaved caspase-3 expression in cSCC arising in sorafenib-treated patients (**, P < 0.01).

Figure 4.

Sorafenib suppresses apoptosis and JNK signaling in vivo. A and B, cSCC samples from sorafenib-treated patients and nontreated patients (sporadic, first row) were analyzed by immunohistochemistry for phospho-JNK and cleaved caspase-3 expression. cSCC arising in sorafenib-treated patients (A and B, second row) show decreased expression of phospho-JNK (A) and cleaved caspase-3 (B) as compared with sporadic cSCC in patients never treated with sorafenib (second row). Scale bar, 100 μm. C, comparisons of stained cells normalized to mm2 of tumor area revealed significant suppression of both phospho-JNK and cleaved caspase-3 expression in cSCC arising in sorafenib-treated patients (**, P < 0.01).

Close modal

To model the emergence of UV-driven cSCC under controlled conditions, we exposed two cohorts of SKH1-E hairless mice to chronic low-dose (12.5 kJ/m2 weekly) UV using solar simulators (Oriel). In this model of UV-driven cSCC development, we irradiated the mice for 72 days before initiation of drug (50 mg/kg daily, oral gavage) or control vehicle treatment. Papillomas were observed within 30 days of drug administration (Fig. 5A) and were histologically well differentiated (Fig. 5B). Some progressed to invasive cSCC and although the kinetics of lesion development were similar, drug-treated animals had 2.3-fold more lesions (P < 10−4; Fig. 5C), with no significant differences in the average size of lesions (2.77 ± 0.26 mm in diameter in controls vs. 3.00 ± 0.20 mm sorafenib-treated; P = 0.49). When we quantified the effects of sorafenib on JNK activation and apoptosis in these papillomas and cSCC, we found decreases in both phospho-JNK expression (51%; P = 0.037; n = 11 pairs) and cleaved caspase-3 expression (72%; P = 0.001; n = 11 pairs; Fig. 5D) in sorafenib-treated mice as compared with control-treated mice.

Figure 5.

Sorafenib increases cSCC development in the UV-driven hairless mouse model. A and C, chronically irradiated SKH-1E hairless mice were treated with sorafenib (n = 5, SOR) and vehicle (n = 5, CON) starting at day 72. Tumors were induced within 30 days of sorafenib treatment and drug-treated (SOR) mice had significantly greater numbers of lesions than controls (CON; A, day 138). Papillomas were histologically well differentiated (B). Although tumors exhibited similar kinetics, there were 2.3-fold more tumors in sorafenib-treated mice as compared with vehicle-treated mice on average (C: ***, P < 0.001). D, tumors from sorafenib-treated animals showed lower levels of phospho-JNK (P = 0.037; *, P < 0.05) and cleaved caspase-3 (P = 0.001; **, P < 0.01) as compared with control-treated animals (n = 6 pairs).

Figure 5.

Sorafenib increases cSCC development in the UV-driven hairless mouse model. A and C, chronically irradiated SKH-1E hairless mice were treated with sorafenib (n = 5, SOR) and vehicle (n = 5, CON) starting at day 72. Tumors were induced within 30 days of sorafenib treatment and drug-treated (SOR) mice had significantly greater numbers of lesions than controls (CON; A, day 138). Papillomas were histologically well differentiated (B). Although tumors exhibited similar kinetics, there were 2.3-fold more tumors in sorafenib-treated mice as compared with vehicle-treated mice on average (C: ***, P < 0.001). D, tumors from sorafenib-treated animals showed lower levels of phospho-JNK (P = 0.037; *, P < 0.05) and cleaved caspase-3 (P = 0.001; **, P < 0.01) as compared with control-treated animals (n = 6 pairs).

Close modal

Paradoxical ERK activation is separable from JNK activity suppression

Although the effects of sorafenib on JNK-dependent apoptosis is clear and independent of ERK activity (Fig. 2), the relative contribution of paradoxical ERK activation versus JNK pathway inhibition to tumorigenesis has not been precisely addressed. To study this, we used the fact that paradoxical ERK activation requires CRAF (11–13). We used isogenic, matched WT and Craf−/− MEFs and transformed them with adenovirus E1A and human HRASG12V to enable anchorage-independent growth. WT and matched Craf−/− MEFs were plated in soft agar assays (16) and treated with sorafenib. Both WT and Craf−/− MEFs exhibited a significant colony formation advantage in the presence of sorafenib (compare Fig. 6A with B). When normalized to untreated transformed WT and Craf−/− MEFs, sorafenib treatment resulted in a 1.92-fold expansion of colony numbers in WT MEFs and a 1.66-fold expansion in Craf−/− MEFs. On the basis of this analysis, we estimated that the effect of paradoxical ERK activation to be 13.5% and other effects, including inhibition of JNK activity, to account for the rest (86.5%) of the total colony growth advantage (Fig. 6C).

Figure 6.

Paradoxical ERK activation and JNK pathway inhibition make significant and separable contributions to sorafenib-induced growth. A and B, we envision two separable, parallel mechanisms by which sorafenib contributes to cSCC development. Drug-induced paradoxical ERK activation and inhibition of JNK signaling occur in parallel, but the former depends on intact CRAF. A and B, representative soft agar colonies of E1A and HRASG12V-transformed wild-type (WT; first row) and Craf−/− (second row) MEFs, following exposure to 1.0 μmol/L sorafenib (B) more than 4 to 6 weeks show significant colony-forming advantages conferred by sorafenib. C, the fold-change in colony counts of transformed WT (n = 36) and Craf−/− (n = 22) MEFs demonstrate a drug-induced increase in colonies, for both WT (1.92-fold) and Craf−/− (1.66-fold) MEFs. The difference between colony formation advantages conferred by 1.0 μmol/L sorafenib in WT versus Craf−/− MEFs was interpreted to reflect the contribution of paradoxical ERK signaling (red arrow), which depends upon Craf, and is 13.5% of the total effect reflected in the first two columns. The remainder of the total effect is composed of other effects, including JNK inhibition (blue arrow). All differences within each MEF population were significant (*, P < 0.05; **, P < 0.01).

Figure 6.

Paradoxical ERK activation and JNK pathway inhibition make significant and separable contributions to sorafenib-induced growth. A and B, we envision two separable, parallel mechanisms by which sorafenib contributes to cSCC development. Drug-induced paradoxical ERK activation and inhibition of JNK signaling occur in parallel, but the former depends on intact CRAF. A and B, representative soft agar colonies of E1A and HRASG12V-transformed wild-type (WT; first row) and Craf−/− (second row) MEFs, following exposure to 1.0 μmol/L sorafenib (B) more than 4 to 6 weeks show significant colony-forming advantages conferred by sorafenib. C, the fold-change in colony counts of transformed WT (n = 36) and Craf−/− (n = 22) MEFs demonstrate a drug-induced increase in colonies, for both WT (1.92-fold) and Craf−/− (1.66-fold) MEFs. The difference between colony formation advantages conferred by 1.0 μmol/L sorafenib in WT versus Craf−/− MEFs was interpreted to reflect the contribution of paradoxical ERK signaling (red arrow), which depends upon Craf, and is 13.5% of the total effect reflected in the first two columns. The remainder of the total effect is composed of other effects, including JNK inhibition (blue arrow). All differences within each MEF population were significant (*, P < 0.05; **, P < 0.01).

Close modal

Compared with other RAF inhibitors such as vemurafenib, the incidence of cSCC associated with sorafenib is relatively low; however, this association is well established in multiple trials (4–6, 8–10). Although these proliferations are indolent and often easily treated, we further explored the mechanism of development of these lesions to make the broader point that adverse reactions are an opportunity to study how off-target pathways may result in biologically significant effects. As the use of sorafenib continues to expand, a thorough understanding of these effects is important in knowing how effects on nontarget tissues occur.

The selectivity of kinase inhibitors remains a crude estimate of the entire spectrum of potential biologic effects. Although sorafenib is not generally regarded as being highly selective, its inhibition of several kinases relevant for cancer cell signaling likely contributes to its clinical efficacy in vivo (1), and it is still unclear to what extent individual targeted kinases contribute to the observed efficacy in certain settings (36). What has remained relatively underexplored in the study of targeted inhibitors is how unintended targets such as ZAK affect the biology of nontarget cells (keratinocytes).

The prevailing explanation for why cSCC development is accelerated in RAF inhibitor–treated patients is paradoxical ERK activation, which occur most prominently in cells with mutant RAS. RAS mutations occur, along with mutations in TGFβR1 and TP53, in cSCC arising in sorafenib-treated patients (4, 5); however, RAS mutations do not seem to be as overrepresented as they are in vemurafenib-induced lesions (16, 17), perhaps suggesting that the relative contribution of paradoxical ERK activation may be lower in sorafenib-exposed cells. Furthermore, paradoxical ERK activation seems to be short-lived (5). Therefore, these findings also suggest that other mechanisms play a role in accelerating the development of cSCC.

Because these cSCC arise relatively quickly and many of the lesions are on sun-exposed areas (4, 6), we specifically chose to look at UV exposure as a predisposing factor, because it is the predominant environmental risk factor for skin cancer. In the course of our studies, we examined multiple signaling pathways, including ERK, p38, and JNK and focused on the latter because of its demonstrated and critical role of UV-induced apoptosis (20, 37).

We have discovered a novel effect of sorafenib in inhibiting UV-induced JNK activation and apoptosis. This effect is independent of ERK signaling, and is not the result of paradoxical ERK activation observed with RAF inhibitors. After profiling several kinases upstream of JNK and p38, we showed that TAOK2 and ZAK were the most likely targets and that inhibition of ZAK is predominantly responsible for this effect. Sorafenib has been previously shown to suppress ZAK and downstream JNK signaling (28–30, 38). Indeed, multiple inhibitors are known to inhibit ZAK, including nilotinib (30), suggesting it may be a promiscuous target of multiple small molecule kinase inhibitors. Whether this is due to structural similarities is not currently known.

We show for the first time that sorafenib suppresses JNK-dependent UV-induced apoptosis in cells and in epidermal squamous proliferations of both treated patients and mice. In addition, sorafenib-induced suppression of JNK signaling contributes to a significant portion of the acceleration of soft agar colony formation in vitro. cSCC arising in patients treated with sorafenib have substantially less phospho-JNK expression and apoptosis than sporadic cSCC arising in patients never treated with sorafenib (Fig. 4). Similarly, in our mouse model of UV-driven cSCC, both phospho-JNK expression and cleaved caspase-3 expression are inhibited by sorafenib (Fig. 5). Finally, we used a soft agar colony assay to test the relative contributions of paradoxical ERK activation and JNK-mediated apoptosis by using Craf−/− MEFs, which do not exhibit paradoxical ERK activation. In this assay, paradoxical ERK activation played a small role in colony growth (Fig. 6). However, our findings do not exclude an absolute requirement for either paradoxical ERK activation or JNK inhibition in the acceleration of cSCC development; our results show that both cooperate in this process. Although we do not exclude the possibility that additional pathways are involved, we have demonstrated that drug-induced suppression of JNK signaling plays a role in UV-exposed keratinocytes and in cSCC acceleration, thus challenging the notion that paradoxical ERK activation alone drives this process. Our results have important implications for the effects of sorafenib on nontarget tissues and suggest that toxicities that are related to JNK signaling are impacted significantly by sorafenib.

N.L. Busaidy has received commercial research grant support from Bayer. No potential conflicts of interest were disclosed by the other authors.

Conception and design: K.Y. Tsai

Development of methodology: H. Vin, G. Ching, V. Chitsazzadeh, D.W. Dwyer, K.Y. Tsai

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): H. Vin, G. Ching, S.S. Ojeda, C.H. Adelmann, V. Chitsazzadeh, D.W. Dwyer, H. Ma, M. Baccarini, J.L. Curry, A.M. Ciurea, M. Duvic, N.L. Busaidy, N.M. Tannir, K.Y. Tsai

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): H. Vin, G. Ching, S.S. Ojeda, C.H. Adelmann, V. Chitsazzadeh, N.M. Tannir, K.Y. Tsai

Writing, review, and/or revision of the manuscript: H. Vin, J.L. Curry, M. Duvic, N.L. Busaidy, K.Y. Tsai

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): G. Ching, V. Chitsazzadeh, D.W. Dwyer, H. Ma, K. Ehrenreiter, R. Ruggieri, N.M. Tannir, K.Y. Tsai

Study supervision: H. Ma, N.M. Tannir, K.Y. Tsai

The authors thank the assistance of Trellis Thompson in initial cell culture experiments, Sherie Mudd, Humaira Khan, Hafsa Ahmed, and Patricia Sheffield for histology, Nasser Kazimi and Omid Tavana for assistance in UV radiation experiments, Brian Rabinovich for advice on lentiviral transduction, and the South Campus Vivarium for mouse maintenance. K.Y. Tsai thanks Ronald P. Rapini for departmental support as well as Tyler Jacks, Gordon B. Mills, Patrick Hwu, and Jeffrey N. Myers for critical discussions and mentorship.

This work was supported by DX Biosciences Cancer Research Fund, MD Anderson Cancer Center IRG Program, American Skin Association, institutional funds, Elsa U. Pardee Foundation, and NCI CA16672 (FACS, Characterized Cell Line, DNA Analysis Facility Cores) (to K.Y. Tsai).

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1.
Iyer
R
,
Fetterly
G
,
Lugade
A
,
Thanavala
Y
. 
Sorafenib: a clinical and pharmacologic review
.
Expert Opin Pharmacother
2010
;
11
:
1943
55
.
2.
Wilhelm
SM
,
Carter
C
,
Tang
L
,
Wilkie
D
,
McNabola
A
,
Rong
H
, et al
BAY 43-9006 exhibits broad spectrum oral antitumor activity and targets the RAF/MEK/ERK pathway and receptor tyrosine kinases involved in tumor progression and angiogenesis
.
Cancer Res
2004
;
64
:
7099
109
.
3.
Autier
J
,
Escudier
B
,
Wechsler
J
,
Spatz
A
,
Robert
C
. 
Prospective study of the cutaneous adverse effects of sorafenib, a novel multikinase inhibitor
.
Arch Dermatol
2008
;
144
:
886
92
.
4.
Arnault
JP
,
Wechsler
J
,
Escudier
B
,
Spatz
A
,
Tomasic
G
,
Sibaud
V
, et al
Keratoacanthomas and squamous cell carcinomas in patients receiving sorafenib
.
J Clin Oncol
2009
;
27
:
e59
61
.
5.
Arnault
JP
,
Mateus
C
,
Escudier
B
,
Tomasic
G
,
Wechsler
J
,
Hollville
E
, et al
Skin tumors induced by sorafenib; paradoxic RAS-RAF pathway activation and oncogenic mutations of HRAS, TP53, and TGFBR1
.
Clin Cancer Res
2012
;
18
:
263
72
.
6.
Dubauskas
Z
,
Kunishige
J
,
Prieto
VG
,
Jonasch
E
,
Hwu
P
,
Tannir
NM
. 
Cutaneous squamous cell carcinoma and inflammation of actinic keratoses associated with sorafenib
.
Clin Genitourinary Cancer
2009
;
7
:
20
3
.
7.
Kong
HH
,
Cowen
EW
,
Azad
NS
,
Dahut
W
,
Gutierrez
M
,
Turner
ML
. 
Keratoacanthomas associated with sorafenib therapy
.
J Am Acad Dermatol
2007
;
56
:
171
2
.
8.
Chapman
PB
,
Hauschild
A
,
Robert
C
,
Haanen
JB
,
Ascierto
P
,
Larkin
J
, et al
Improved survival with vemurafenib in melanoma with BRAF V600E mutation
.
N Engl J Med
2011
;
364
:
2507
16
.
9.
Flaherty
KT
,
Puzanov
I
,
Kim
KB
,
Ribas
A
,
McArthur
GA
,
Sosman
JA
, et al
Inhibition of mutated, activated BRAF in metastatic melanoma
.
N Engl J Med
2010
;
363
:
809
19
.
10.
Sosman
JA
,
Kim
KB
,
Schuchter
L
,
Gonzalez
R
,
Pavlick
AC
,
Weber
JS
, et al
Survival in BRAF V600-mutant advanced melanoma treated with vemurafenib
.
N Engl J Med
2012
;
366
:
707
14
.
11.
Heidorn
SJ
,
Milagre
C
,
Whittaker
S
,
Nourry
A
,
Niculescu-Duvas
I
,
Dhomen
N
, et al
Kinase-dead BRAF and oncogenic RAS cooperate to drive tumor progression through CRAF
.
Cell
2010
;
140
:
209
21
.
12.
Hatzivassiliou
G
,
Song
K
,
Yen
I
,
Brandhuber
BJ
,
Anderson
DJ
,
Alvarado
R
, et al
RAF inhibitors prime wild-type RAF to activate the MAPK pathway and enhance growth
.
Nature
2010
;
464
:
431
5
.
13.
Poulikakos
PI
,
Zhang
C
,
Bollag
G
,
Shokat
KM
,
Rosen
N
. 
RAF inhibitors transactivate RAF dimers and ERK signalling in cells with wild-type BRAF
.
Nature
2010
;
464
:
427
30
.
14.
Karreth
FA
,
DeNicola
GM
,
Winter
SP
,
Tuveson
DA
. 
C-Raf inhibits MAPK activation and transformation by B-Raf(V600E)
.
Mol Cell
2009
;
36
:
477
86
.
15.
Halaban
R
,
Zhang
W
,
Bacchiocchi
A
,
Cheng
E
,
Parisi
F
,
Ariyan
S
, et al
PLX4032, a selective BRAF(V600E) kinase inhibitor, activates the ERK pathway and enhances cell migration and proliferation of BRAF melanoma cells
.
Pigment Cell Melanoma Res
2010
;
23
:
190
200
.
16.
Su
F
,
Viros
A
,
Milagre
C
,
Trunzer
K
,
Bollag
G
,
Spleiss
O
, et al
RAS mutations in cutaneous squamous-cell carcinomas in patients treated with BRAF inhibitors
.
N Engl J Med
2012
;
366
:
207
15
.
17.
Oberholzer
PA
,
Kee
D
,
Dziunycz
P
,
Sucker
A
,
Kamsukom
N
,
Jones
R
, et al
RAS mutations are associated with the development of cutaneous squamous cell tumors in patients treated with RAF inhibitors
.
J Clin Oncol
2012
;
30
:
316
21
.
18.
Flaherty
KT
,
Infante
JR
,
Daud
A
,
Gonzalez
R
,
Kefford
RF
,
Sosman
J
, et al
Combined BRAF and MEK Inhibition in Melanoma with BRAF V600 Mutations
.
N Engl J Med
2012
;
367
:
1694
703
.
19.
Weber
JS
,
Flaherty
KT
,
Infante
JR
,
Falchook
GS
,
Kefford
R
,
Daud
A
, et al
Updated safety and efficacy results from a phase I/II study of the oral BRAF inhibitor dabrafenib (GSK2118436) combined with the oral MEK 1/2 inhibitor trametinib (GSK1120212) in patients with BRAFi-naive metastatic melanoma
.
J Clin Oncol
2012
.
[Epub ahead of print]
.
20.
Tournier
C
,
Hess
P
,
Yang
DD
,
Xu
J
,
Turner
TK
,
Nimnual
A
, et al
Requirement of JNK for stress-induced activation of the cytochrome c-mediated death pathway
.
Science
2000
;
288
:
870
4
.
21.
Haeusgen
W
,
Herdegen
T
,
Waetzig
V
. 
The bottleneck of JNK signaling: molecular and functional characteristics of MKK4 and MKK7
.
Eur J Cell Biol
2011
;
90
:
536
44
.
22.
Tournier
C
,
Whitmarsh
AJ
,
Cavanagh
J
,
Barrett
T
,
Davis
RJ
. 
Mitogen-activated protein kinase kinase 7 is an activator of the c-Jun NH2-terminal kinase
.
Proc Natl Acad Sci U S A
1997
;
94
:
7337
42
.
23.
Yang
D
,
Tournier
C
,
Wysk
M
,
Lu
HT
,
Xu
J
,
Davis
RJ
, et al
Targeted disruption of the MKK4 gene causes embryonic death, inhibition of c-Jun NH2-terminal kinase activation, and defects in AP-1 transcriptional activity
.
Proc Natl Acad Sci U S A
1997
;
94
:
3004
9
.
24.
Haass
NK
,
Sproesser
K
,
Nguyen
TK
,
Contractor
R
,
Medina
CA
,
Nathanson
KL
, et al
The mitogen-activated protein/extracellular signal-regulated kinase kinase inhibitor AZD6244 (ARRY-142886) induces growth arrest in melanoma cells and tumor regression when combined with docetaxel
.
Clin Cancer Res
2008
;
14
:
230
9
.
25.
Inaba
H
,
Rubnitz
JE
,
Coustan-Smith
E
,
Li
L
,
Furmanski
BD
,
Mascara
GP
, et al
Phase I pharmacokinetic and pharmacodynamic study of the multikinase inhibitor sorafenib in combination with clofarabine and cytarabine in pediatric relapsed/refractory leukemia
.
J Clin Oncol
2011
;
29
:
3293
300
.
26.
Davis
MI
,
Hunt
JP
,
Herrgard
S
,
Ciceri
P
,
Wodicka
LM
,
Pallares
G
, et al
Comprehensive analysis of kinase inhibitor selectivity
.
Nat Biotechnol
2011
;
29
:
1046
51
.
27.
Keshet
Y
,
Seger
R
. 
The MAP kinase signaling cascades: a system of hundreds of components regulates a diverse array of physiological functions
.
Methods Mol Biol
2010
;
661
:
3
38
.
28.
Yang
JJ
. 
Mixed lineage kinase ZAK utilizing MKK7 and not MKK4 to activate the c-Jun N-terminal kinase and playing a role in the cell arrest
.
Biochem Biophys Res Commun
2002
;
297
:
105
10
.
29.
Sauter
KA
,
Magun
EA
,
Iordanov
MS
,
Magun
BE
. 
ZAK is required for doxorubicin, a novel ribotoxic stressor, to induce SAPK activation and apoptosis in HaCaT cells
.
Cancer Biol Ther
2010
;
10
:
258
66
.
30.
Wong
J
,
Smith
LB
,
Magun
EA
,
Engstrom
T
,
Kelley-Howard
K
,
Jandhyala
DM
, et al
Small molecule kinase inhibitors block the ZAK-dependent inflammatory effects of doxorubicin
.
Cancer Biol Ther
2013
;
14
:
56
63
.
31.
de Anda
FC
,
Rosario
AL
,
Durak
O
,
Tran
T
,
Graff
J
,
Meletis
K
, et al
Autism spectrum disorder susceptibility gene TAOK2 affects basal dendrite formation in the neocortex
.
Nat Neurosci
2012
;
15
:
1022
31
.
32.
Wu
MF
,
Wang
SG
. 
Human TAO kinase 1 induces apoptosis in SH-SY5Y cells
.
Cell Biol Int
2008
;
32
:
151
6
.
33.
Chen
Z
,
Cobb
MH
. 
Regulation of stress-responsive mitogen-activated protein (MAP) kinase pathways by TAO2
.
J Biol Chem
2001
;
276
:
16070
5
.
34.
Zihni
C
,
Mitsopoulos
C
,
Tavares
IA
,
Ridley
AJ
,
Morris
JD
. 
Prostate-derived sterile 20-like kinase 2 (PSK2) regulates apoptotic morphology via C-Jun N-terminal kinase and Rho kinase-1
.
J Biol Chem
2006
;
281
:
7317
23
.
35.
Moore
TM
,
Garg
R
,
Johnson
C
,
Coptcoat
MJ
,
Ridley
AJ
,
Morris
JD
. 
PSK, a novel STE20-like kinase derived from prostatic carcinoma that activates the c-Jun N-terminal kinase mitogen-activated protein kinase pathway and regulates actin cytoskeletal organization
.
J Biol Chem
2000
;
275
:
4311
22
.
36.
Heinrich
MC
,
Marino-Enriquez
A
,
Presnell
A
,
Donsky
RS
,
Griffith
DJ
,
McKinley
A
, et al
Sorafenib inhibits many kinase mutations associated with drug-resistant gastrointestinal stromal tumors
.
Mol Cancer Ther
2012
;
11
:
1770
80
.
37.
Derijard
B
,
Hibi
M
,
Wu
IH
,
Barrett
T
,
Su
B
,
Deng
T
, et al
JNK1: a protein kinase stimulated by UV light and Ha-Ras that binds and phosphorylates the c-Jun activation domain
.
Cell
1994
;
76
:
1025
37
.
38.
Nakamura
K
,
Hatano
E
,
Narita
M
,
Miyagawa-Hayashino
A
,
Koyama
Y
,
Nagata
H
, et al
Sorafenib attenuates monocrotaline-induced sinusoidal obstruction syndrome in rats through suppression of JNK and MMP-9
.
J Hepatol
2012
;
57
:
1037
43
.