Purpose: This phase I clinical trial was conducted to determine the safety, efficacy, and molecular effects of sorafenib with temsirolimus in patients with advanced melanoma.

Patients and Methods: Patients with stage IV or unresectable or recurrent stage III melanoma and Eastern Cooperative Oncology Group performance status of 0 to 1 were eligible. Sorafenib was given orally once or twice daily and temsirolimus was given i.v. weekly, both starting on day 1, with a 4-week cycle. Responses were assessed every 2 cycles per Response Evaluation Criteria in Solid Tumors criteria. Consenting patients with accessible tumors underwent optional tumor biopsies before treatment and after the second infusion of temsirolimus. Tumor biopsies were analyzed for activating mutations in BRAF and NRAS, and for expression of P-extracellular signal-regulated kinase (P-ERK) and P-S6 proteins.

Results: A total of 25 patients were accrued to the study. The maximum tolerated doses were sorafenib 400 mg every morning and 200 mg every evening and temsirolimus 25 mg i.v. weekly. Dose-limiting toxicities included thrombocytopenia, hand-foot syndrome, serum transaminase elevation, and hypertriglyceridemia. There were no complete or partial responses with the combination; 10 patients achieved stabilization of disease as their best response. The median progression-free survival was 2.1 months. Matching pretreatment and day 15 tumor biopsies showed marked inhibition of P-S6 with treatment in 3 of 4 evaluable patients, but minimal inhibition of P-ERK.

Conclusions: Combination therapy with sorafenib and temsirolimus resulted in significant toxicity at higher dose levels, failed to achieve any clinical responses in genetically unselected patient population, and did not inhibit P-ERK. Clin Cancer Res; 18(4); 1120–8. ©2012 AACR.

Translational Relevance

Targeted therapies against the RAS-RAF-MEK-ERK signaling pathway have recently shown dramatic activity in patients with melanoma, but their clinical benefit has been limited by the rapid development of resistance. A number of lines of evidence support that combined inhibition of the RAS-RAF-MEK-ERK and the PI3K-AKT pathways may be an effective strategy to overcome resistance. This article describes the results of the first clinical trial in melanoma assessing both the antitumor effects and molecular correlates of combined therapy against both of these pathways, with sorafenib (RAF inhibitor) and temsirolimus (mTOR inhibitor). The combination had significant toxicity and failed to achieve clinical responses in any patients, including patients with activating BRAF mutations. Pharmacodynamic analysis supports that the regimen was able to inhibit signaling by mTOR, but it did not inhibit ERK activation. The results reinforce the need for pharmacodynamic analysis in the evaluation of novel targeted therapies and combinations.

The majority of melanomas have a mutation in the RAS-RAF-MEK-ERK signaling pathway, including activating mutations in the BRAF (∼45%) and NRAS (∼20%) genes (1). Whereas activating mutations in BRAF are thought to predominantly activate downstream signaling via mitogen-activated protein/extracellular signal-regulated kinase (ERK) kinase (MEK) and ERK, RAS GTPases activate additional signaling pathways (2). In particular, the PI3K-AKT-mTOR pathway plays a critical role in oncogenic signaling by RAS, and it is a promising combinatorial target for NRAS-mutant tumors (3, 4). The PI3K-AKT-mTOR pathway is also activated in cancer by loss of function of the lipid phosphatases PTEN, which is detected in 10% to 30% of melanomas (5–7). PTEN loss is mutually exclusive with NRAS mutations, but often occurs in melanomas with a concurrent BRAF mutation. Functionally, loss of PTEN complements activating BRAF mutations to transform melanocytes in a murine model (8). Furthermore, activation of PI3K-AKT-mTOR signaling has been implicated in both primary and secondary resistance to BRAF and MEK inhibitors (9–12). Thus, there is a strong rationale to test the clinical effects of combined inhibition of the RAS-RAF-MEK-ERK and PI3K-AKT-mTOR pathways in melanoma.

Sorafenib (also called BAY 43-9006) is a small molecule inhibitor of several protein kinases, including wild-type and mutant BRAF, CRAF, c-KIT, and VEGFRs (13). Sorafenib is effective at inhibiting the growth and survival of melanoma cell lines and xenografts with BRAF or NRAS mutations in preclinical models (14). However, it achieves a clinical response in only 3% of patients (15, 16). One of these clinical studies evaluated the effects of sorafenib treatment on cyclin D1 and Ki67 expression in patient specimens (16). However, to date, the effects of sorafenib treatment on ERK phosphorylation, which correlates with tumor regression with the selective BRAF inhibitor vemurafenib, has only been reported in one melanoma patient (17, 18).

Combined treatment with sorafenib and rapamycin, an mTOR inhibitor, decreases proliferation and invasion and induces apoptosis in BRAF-mutant human melanoma cell lines (19). Synergistic growth inhibition with this combination is observed in human melanoma cell lines with both mutant and wild-type BRAF (20). Clinical testing of mTOR inhibition in melanoma has previously been reported with temsirolimus (also called CCI-779), an mTOR inhibitor structurally related to rapamycin (21). Temsirolimus was well tolerated, but only 1 of 33 metastatic melanoma patients achieved a clinical response. Although previous studies have examined the degree of inhibition of the phosphorylation (activation) of the mTOR effector S6 in circulating cells and head and neck carcinomas, no assessments have been reported in melanoma patients (22, 23).

We have conducted a phase I clinical trial of concurrent treatment with sorafenib and temsirolimus. The primary endpoint of the study was to evaluate the safety and toxicity of this regimen, and to determine the maximum tolerated dose (MTD) for further testing. The secondary endpoint was to determine the clinical activity of this treatment in metastatic melanoma. Patients with accessible tumors underwent optional biopsies before and during treatment with sorafenib and temsirolimus for quantitative proteomic analysis by reverse phase protein arrays (RPPA; refs. 24, 25). The presence of activating mutations in BRAF and NRAS, and effects on P-ERK and P-S6 protein expression, were compared with the clinical efficacy of the combination.

Patient selection

Patients with histologically confirmed stage IV or recurrent or unresectable stage III melanoma of nonuveal origin were enrolled. Major inclusion criteria included: Eastern Cooperative Oncology Group (ECOG) performance status of 1 or less, measurable disease defined by Response Evaluation Criteria in Solid Tumors (RECIST) version 1.0, adequate organ functions, no active or symptomatic brain metastases and no steroid support for palliation, and uncontrolled blood pressure. There was no restriction in the number of prior therapies.

Treatment plan

The protocol for this study was approved by the Institutional Review Board of The University of Texas MD Anderson Cancer Center, and all patients gave written informed consent before enrollment. The doses of oral sorafenib and intravenous temsirolimus in the first patient cohort were 400 mg twice a day and 15 mg weekly, respectively (dose level 1). Table 1 lists the dose escalation scheme. One cycle was defined as 4 weeks of treatment. The maximum number of treatment cycles planned was 6; patients who continued to have disease regression were allowed to receive more than 6 cycles, as long as they tolerated treatment well. Intrapatient dose escalation was not permitted.

Table 1.

Dose escalation scheme with patient mutation status

Dose levelSorafenib (mg orally)Temsirolimus (mg i.v. weekly)Total number treatedBRAF, nNRAS, nWT, nNE, n
400 every day 15    
1a 400 twice daily 15 
1a 200 twice daily 25   
1bb 400 in am 25 10  
 200 in pm       
400 twice daily 25   
400 twice daily 50      
400 twice daily 75      
Dose levelSorafenib (mg orally)Temsirolimus (mg i.v. weekly)Total number treatedBRAF, nNRAS, nWT, nNE, n
400 every day 15    
1a 400 twice daily 15 
1a 200 twice daily 25   
1bb 400 in am 25 10  
 200 in pm       
400 twice daily 25   
400 twice daily 50      
400 twice daily 75      

NOTE: Dose levels 1a and 1b were added after the trial was initiated.

Abbreviations: WT, wild-type BRAF and NRAS; NE, not evaluable.

aStarting dose level.

bMTD: This dose level is recommended for a phase II study.

Toxicity evaluation

Toxicity was evaluated according to the U.S. National Cancer Institute Common Terminology Criteria for Adverse Events v3.0 (https://webapps.ctep.nci.nih.gov/webobjs/ctc/webhelp/welcome_to_ctcae.htm). Patients were assessed for safety every 2 weeks during the first 2 cycles and then once every cycle. Complete blood counts with differential counts and serum chemistry were obtained once a week during the first 2 cycles and then every other week. Dose-limiting toxicity (DLT) was defined as grade 4 hematologic toxicity, grade 3 or more nonhematologic toxicity, or any grade 2 nonhematologic, or grade 3 hematologic toxicity requiring a dose reduction or treatment interruption for more than 7 days during the first cycle. Grade 3 to 4 nausea, vomiting, or diarrhea were only considered DLTs if they occurred despite optimal medical management. Grade 3 electrolytes, uric acid, phosphorus abnormalities were not considered DLTs if they were correctable within 1 week.

Response evaluation

Responses were evaluated after every 2 cycles of the treatment using computed tomography and/or MRI, as well as by direct ruler measurement for clearly visible cutaneous lesions. Clinical responses were evaluated using the international criteria proposed by the RECIST Committee (26).

Population pharmacokinetic analysis

Blood samples were collected on consenting patients at the following time points of cycle one: Day 1: pretreatment, 1, 4, 6, 8, 10, and 24 hours after start of administration of sorafenib; Day 15: pretreatment, 1, 4, 6, 8, 10, and 24 hours after treatment. Blood samples were collected in 5-mL sodium heparin tubes and obtained in Clinical and Translational Research Center at The University of Texas MD Anderson Cancer Center. Following collection, they were immediately centrifuged at 1,500 rpm for 10 minutes at 4°C for separation. Plasma samples were aliquoted and stored in labeled cryovials at approximately −70°C until analysis. Plasma sorafenib concentrations were determined by liquid chromatography/tandem mass spectrometry. A Waters Acquity UPLC system (Waters) with a 4°C autosampler was run in tandem with a Waters Quattro Premier mass spectrometer using MassLynx/Quanlynx software for data acquisition and integration. Sorafenib was extracted using a liquid–liquid method by adding 4 mL methyl tert-butyl ether to 100 μL of plasma and vortexed for 5 minutes. The resulting mixture was centrifuged at 2,000 × g for 8 minutes at 4°C and frozen at −80°C for 30 minutes. The resulting supernatant was evaporated to dryness at 40°C under gentle nitrogen gas stream and reconstituted with 100 μL of 50:50, methanol:water (v:v). Chromatographic separation was carried out following a 10 μL injection on a Phenomenex Kinetex C18 column (100 × 2.1 mm, 2.6 μm) over 5 minutes at 0.4 mL/min using 50:50, 0.2% formic acid:methanol (v:v) with a linear gradient to 98% methanol over the first 2 minutes. The monitored transition for sorafenib was 465 > 271 m/z with a peak retention time of 2.52 minutes in positive electrospray ionization mode (ESI+). The calibration curve ranged from 1 to 10,000 ng/mL in single donor human plasma. Pharmacokinetic parameter estimates were generated from individual patient concentration–time data using a noncompartmental model (WinNonLin v5.2; Pharsight Corp.).

Tumor biomarker studies

After signing informed consent, each patient underwent a tumor biopsy (punch or core-needle biopsy), which was embedded in Optimum Cutting Temperature (OCT) solution, unless frozen or OCT-embedded tumor samples obtained within 3 months before signing informed consent were available. An optional tumor biopsy was conducted within 6 hours after the completion of temsirolimus infusion on day 15 of the first treatment cycle. Hematoxylin and eosin stain (H&E) slides were generated from each tumor, which were reviewed by an experienced dermatopathologist. Tumors with regions of at least 70% viable tumor cells of sufficient size for DNA and protein extraction underwent H&E-guided macrodissection (7). Shears of the tumor-enriched isolates were prepared using a DNAse-free, RNAse-free microtome at −20°C, then were stored at −80°C. DNA was extracted from the samples and underwent mass spectroscopy–based genotyping for mutations in BRAF (exon 15) and NRAS (exons 1and 2), as previously described (7, 27). Proteins were extracted from the samples as previously described (7). Protein lysates were normalized to the same concentration, denatured, and then submitted for RPPA analysis through the MD Anderson Cancer Center RPPA Core Facility (http://www.mdanderson.org/education-and-research/resources-for-professionals/scientific-resources/core-facilities-and-services/functional-proteomics-rppa-core/index.html). Log2 expression data for each protein was corrected for measured differences in total protein loading as previously described (7). A detailed description of the RPPA procedure is provided in the Supplementary Materials.

Statistical analysis

The MTD was defined as the highest dose at which the incidence of DLT was less than 33%. A conventional 3+3 dose escalation design with predefined dose levels of sorafenib and temsirolimus was implemented. At the MTD, a total of 10 patients were enrolled to ensure that the safety profile was acceptable. Overall clinical response included both complete and partial responses. Progression-free survival (PFS) and overall survival (OS) were measured from day 1 of treatment. One patient discontinued the study due to toxicity before disease progression and used different therapies; this patient was censored for PFS at the time of study discontinuation. Patients that did not experience death were censored at the time of their last clinic follow-up. The duration of clinical response was measured from the time a clinical response was first achieved until the time of disease progression or death, whichever occurred first.

The association between tumor mutation status and clinical response was determined using Fisher exact test. Kaplan–Meier survival curves were used to estimate PFS and OS; log-rank testing was used to compare curves. Pharmacodynamic effects of treatment with sorafenib and temsirolimus on P-ERK and P-S6 were determined by calculating the ratio of phospho:total protein expression in each sample. Pretreatment and day 15 ratios were compared using paired Student t test.

Patient characteristics

Twenty-five patients were enrolled and treated in the study (Table 2). The median age of the patients was 51 years. Forty percent of the patients had stage III melanoma, 12% had stage IV M1a, 28% stage IV M1b, and 20% stage IV M1c, respectively. Fifty-six percent of the patients had received at least 2 prior systemic therapies for metastatic melanoma.

Table 2.

Patient characteristics

CharacteristicNo. of patients (%)
Total patients enrolled in the study 25 (100) 
Sex 
 Male 18 (72) 
 Female 7 (28) 
Age (y) 
 Median (range) 51 [22–76] 
ECOG performance status 
 0 10 (40) 
 1 15 (60) 
Disease stage 
 III (unresectable or recurrent) 10 (40) 
 IV 
  M1a 3 (12) 
  M1b 7 (28) 
  M1c 5 (20) 
Serum lactate dehydrogenase level 
 Normal 19 (76) 
 Higher than upper limit of normal 6 (24) 
Prior systemic treatment 
 0 4 (16) 
 1 7 (28) 
 2 8 (32) 
 3 4 (16) 
 4 2 (8) 
CharacteristicNo. of patients (%)
Total patients enrolled in the study 25 (100) 
Sex 
 Male 18 (72) 
 Female 7 (28) 
Age (y) 
 Median (range) 51 [22–76] 
ECOG performance status 
 0 10 (40) 
 1 15 (60) 
Disease stage 
 III (unresectable or recurrent) 10 (40) 
 IV 
  M1a 3 (12) 
  M1b 7 (28) 
  M1c 5 (20) 
Serum lactate dehydrogenase level 
 Normal 19 (76) 
 Higher than upper limit of normal 6 (24) 
Prior systemic treatment 
 0 4 (16) 
 1 7 (28) 
 2 8 (32) 
 3 4 (16) 
 4 2 (8) 

Treatment

One of the first 3 patients treated at dose level 1 had a DLT (grade 3 hand-foot syndrome and thrombocytopenia requiring dose reduction; Table 3). However, no other DLTs were observed among 3 patients treated at dose level 0, nor among an additional 3 patients treated at dose level 1. At dose level 2, both of 2 patients had a DLT (grade 3 serum transaminase elevation and hand-foot syndrome). To better define a MTD of the combination, 2 additional dose levels (level 1a and 1b) were evaluated. All 4 patients at dose level 1a tolerated the treatment without DLTs. The next 6 patients were treated at dose level 1b with only 1 patient experiencing reversible grade 3 hypertriglyceridemia. Four additional patients were treated at dose level 1b, and 2 of them experienced DLTs of grade 3 or more thrombocytopenia requiring dose reduction. Dose level 1b (sorafenib 400 mg in am and 200 mg in pm daily and temsirolimus 25 mg weekly) was selected as the MTD.

Table 3.

Grade 3/4 toxicities

Dose levelSorafenib (mg)Temsirolimus (mg weekly)Toxicity
Grade 3Grade 4
0 (n = 3) 400 every day 15 Arthralgia (1) None 
   Vomiting (1)  
1 (n = 6) 400 twice daily 15 Hypophosphotemia (4) None 
   Fatigue (3)  
   Diarrhea (2)  
   Thrombocytopenia (1)  
   Dyspnea (1)  
   Hand-Foot Syndrome (1)  
   AST/ALT elevation (1)  
1a (n = 4) 200 twice daily 25 Lymphopenia (1) None 
   Anorexia (1)  
   Hypophosphotemia (1)  
   Fatigue (1)  
1b (n = 10) 400/200 25 Fatigue (3) Thrombocytopenia (1) 
   Hypophosphotemia (2)  
   Thrombocytopenia (1)  
   Lymphopenia (1)  
   Hypokalemia (1)  
   Cough (1)  
   Hypertriglyceridemia (1)  
   AST/ALT elevation (1)  
2 (n = 2) 400 twice daily 25 Fatigue (2) None 
   Lymphopenia (1)  
   Hand-Foot Syndrome (1)  
   AST/ALT elevation (1)  
   Hypophosphotemia (1)  
Dose levelSorafenib (mg)Temsirolimus (mg weekly)Toxicity
Grade 3Grade 4
0 (n = 3) 400 every day 15 Arthralgia (1) None 
   Vomiting (1)  
1 (n = 6) 400 twice daily 15 Hypophosphotemia (4) None 
   Fatigue (3)  
   Diarrhea (2)  
   Thrombocytopenia (1)  
   Dyspnea (1)  
   Hand-Foot Syndrome (1)  
   AST/ALT elevation (1)  
1a (n = 4) 200 twice daily 25 Lymphopenia (1) None 
   Anorexia (1)  
   Hypophosphotemia (1)  
   Fatigue (1)  
1b (n = 10) 400/200 25 Fatigue (3) Thrombocytopenia (1) 
   Hypophosphotemia (2)  
   Thrombocytopenia (1)  
   Lymphopenia (1)  
   Hypokalemia (1)  
   Cough (1)  
   Hypertriglyceridemia (1)  
   AST/ALT elevation (1)  
2 (n = 2) 400 twice daily 25 Fatigue (2) None 
   Lymphopenia (1)  
   Hand-Foot Syndrome (1)  
   AST/ALT elevation (1)  
   Hypophosphotemia (1)  

Toxicity

The most common grade 3 or 4 nonhematologic adverse events were fatigue and hypophosphotemia (Table 4). The most common grade 3/4 hematologic toxicities were thrombocytopenia (n = 3) and lymphopenia (n = 3; Table 4). Seventeen (68%) patients required dose reduction during the course of the treatment. All adverse events resolved upon treatment discontinuation.

Table 4.

Dose-limiting toxicities per dose level

Dose levelSorafenib (mg)Temsirolimus (mg weekly)Number of DLTsDLTs
400 every day 15 0/3 None 
400 twice daily 15 1a/6 G3 Hand-Foot syndrome 
    G3 Thrombocytopenia (requiring dosing reduction) 
1a 200 twice daily 25 0/4 None 
1b 400/200 25 3/10 G3 Hypertriglyceridemia 
    G4 Thrombocytopenia 
    G3 Thrombocytopenia (requiring dosing reduction) 
400 twice daily 25 2/2 G3 serum transaminases elevationb 
    G3 Hand-Foot syndrome 
Dose levelSorafenib (mg)Temsirolimus (mg weekly)Number of DLTsDLTs
400 every day 15 0/3 None 
400 twice daily 15 1a/6 G3 Hand-Foot syndrome 
    G3 Thrombocytopenia (requiring dosing reduction) 
1a 200 twice daily 25 0/4 None 
1b 400/200 25 3/10 G3 Hypertriglyceridemia 
    G4 Thrombocytopenia 
    G3 Thrombocytopenia (requiring dosing reduction) 
400 twice daily 25 2/2 G3 serum transaminases elevationb 
    G3 Hand-Foot syndrome 

NOTE: Dose level 1b was selected as MTD.

aOne patient had both DLTs at dose level 1.

bBoth serum alanine aminotransferase and aspartate aminotransferase elevation.

Pharmacokinetics

The plasma concentration–time profiles of sorafenib were characterized by a relatively slow absorption and elimination phase with primary and secondary peaks in a number of patients, as seen in a previous single-agent phase 1 study (18). Sorafenib mean peak plasma concentrations (Cmax) and mean daily exposure, measured as the average concentration over 24 hours (Cavg), was 2- to 4-fold greater at steady state on day 15 when compared with day 1 with low-to-moderate interpatient variability across all dosing levels (Table 5). Sorafenib mean Cavg at day 15 with 400 mg twice daily dosing was 8.45 mg/L, which was more than a dose proportional increase when compared with 400 mg every day (3.16 mg/L); mean Cmax increased proportionately (11.83 vs. 5.38 mg/L). Mean Cavg with sorafenib dosing of 400 mg am/200 mg pm on day 15 was 4.55 mg/L, which was similar to the result observed in the one patient treated with 200 mg twice daily with available day 15 data (Cavg 5.12 mg/L). The sorafenib mean Cmax was similar for all dose levels on day 1 but became more dose proportional by day 15 (except for 200 mg twice daily). This trend and with the mean Cmax levels at 400 mg twice daily were similar to those observed in a review of 4 phase I trials with single-agent sorafenib in patients with advanced cancer (28).

Table 5.

Sorafenib pharmacokinetics

Sorafenib concentration (mg/L)
PatientSorafenib dose (mg, frequency)Temsirolimus dose (mg, weekly)Cavg/Cmax, day 1Cavg/Cmax, day 15
400, twice daily 15 1.43/2.09 7.10/8.91 
400, twice daily 15 2.07/2.53 7.70/12.46 
400, twice daily 15 1.23/1.94 9.79/13.53 
400, twice daily 15 2.70/3.70 8.80/12.46 
400, twice daily 15 2.23/3.61 8.84/11.80 
Mean   1.93/2.77 8.45/11.83 
SD   0.60/0.83 1.06/1.75 
CV%   31.1/30.0 12.5/14.8 
400, every day 15 1.33/1.82 3.45/4.87 
400, every day 15 1.40/2.94 3.49/7.01 
400, every day 15 1.11/1.78 2.53/4.27 
Mean   1.28/2.18 3.16/5.38 
SD   0.15/0.66 0.54/1.44 
CV%   11.8/30.2 17.2/26.8 
12 200, twice daily 25 0.73/1.04 5.12/6.75 
13 200, twice daily 25 0.67/2.47 ND 
14 200, twice daily 25 0.83/2.96 ND 
15 200, twice daily 25 0.47/1.42 ND 
Mean   0.68/1.97 ND 
SD   0.15/0.89 ND 
CV%   22.5/45.3 ND 
16 400 am, 200 pm 25 0.90/1.40 4.19/6.80 
17 400 am, 200 pm 25 1.30/2.19 3.17/5.39 
20 400 am, 200 pm 25 1.00/1.66 6.28/9.08 
Mean   1.07/1.75 4.55/7.09 
SD   0.21/0.40 1.59/1.86 
CV%   19.5/23.0 34.9/26.3 
Sorafenib concentration (mg/L)
PatientSorafenib dose (mg, frequency)Temsirolimus dose (mg, weekly)Cavg/Cmax, day 1Cavg/Cmax, day 15
400, twice daily 15 1.43/2.09 7.10/8.91 
400, twice daily 15 2.07/2.53 7.70/12.46 
400, twice daily 15 1.23/1.94 9.79/13.53 
400, twice daily 15 2.70/3.70 8.80/12.46 
400, twice daily 15 2.23/3.61 8.84/11.80 
Mean   1.93/2.77 8.45/11.83 
SD   0.60/0.83 1.06/1.75 
CV%   31.1/30.0 12.5/14.8 
400, every day 15 1.33/1.82 3.45/4.87 
400, every day 15 1.40/2.94 3.49/7.01 
400, every day 15 1.11/1.78 2.53/4.27 
Mean   1.28/2.18 3.16/5.38 
SD   0.15/0.66 0.54/1.44 
CV%   11.8/30.2 17.2/26.8 
12 200, twice daily 25 0.73/1.04 5.12/6.75 
13 200, twice daily 25 0.67/2.47 ND 
14 200, twice daily 25 0.83/2.96 ND 
15 200, twice daily 25 0.47/1.42 ND 
Mean   0.68/1.97 ND 
SD   0.15/0.89 ND 
CV%   22.5/45.3 ND 
16 400 am, 200 pm 25 0.90/1.40 4.19/6.80 
17 400 am, 200 pm 25 1.30/2.19 3.17/5.39 
20 400 am, 200 pm 25 1.00/1.66 6.28/9.08 
Mean   1.07/1.75 4.55/7.09 
SD   0.21/0.40 1.59/1.86 
CV%   19.5/23.0 34.9/26.3 

Cavg, average daily sorafenib plasma concentration; Cmax, peak sorafenib plasma concentration; CV%, coefficient of variation; ND, no data available.

Clinical efficacy and molecular correlates

None of the patients had a clinical response. Ten patients had disease stabilization for at least 8 weeks; only 3 patients had disease stabilization for more than 16 weeks. The median number of cycles received per patient was 2 (range < 1–8). Overall the median PFS was 2.1 months (Fig. 1), and the median OS was 1.3 years (Supplementary Fig. S1). Among the 10 patients treated at the MTD, 2 achieved stable disease, the median PFS was 2.0 months, and the median OS was 1.2 years.

Figure 1.

Clinical and pharmacodynamic effects of treatment with sorafenib and temsirolimus. A, PFS from the start of treatment with sorafenib and temsirolimus for all patients enrolled in the trial. B, PFS from the start of treatment with sorafenib and temsirolimus for all patients with known mutation status for BRAF and NRAS. Red, activating BRAF mutation; Blue, activating NRAS mutation; Black, wild-type BRAF and NRAS genes. P value determined by log-rank testing. C, changes in S6 Ser240 phosphorylation with treatment. The expression of phosphorylated (P) and total (T) S6 protein was determined by RPPA. The ratio of P:T S6 protein in the pretreatment (black bars) and day 15 (gray) tumor biopsies in the patients with matching samples are shown. The patient mutation status, best clinical response, dose level, and temsirolimus dose are shown below each patient label. SD, stable disease D, changes in ERK phosphorylation with treatment. In addition to mutation status, best clinical response, dose level, and sorafenib dose, the sorafenib day 15 average serum concentration (Cavg) for each patient is indicated. Day 15 Cavg was not available for one patient (NA).

Figure 1.

Clinical and pharmacodynamic effects of treatment with sorafenib and temsirolimus. A, PFS from the start of treatment with sorafenib and temsirolimus for all patients enrolled in the trial. B, PFS from the start of treatment with sorafenib and temsirolimus for all patients with known mutation status for BRAF and NRAS. Red, activating BRAF mutation; Blue, activating NRAS mutation; Black, wild-type BRAF and NRAS genes. P value determined by log-rank testing. C, changes in S6 Ser240 phosphorylation with treatment. The expression of phosphorylated (P) and total (T) S6 protein was determined by RPPA. The ratio of P:T S6 protein in the pretreatment (black bars) and day 15 (gray) tumor biopsies in the patients with matching samples are shown. The patient mutation status, best clinical response, dose level, and temsirolimus dose are shown below each patient label. SD, stable disease D, changes in ERK phosphorylation with treatment. In addition to mutation status, best clinical response, dose level, and sorafenib dose, the sorafenib day 15 average serum concentration (Cavg) for each patient is indicated. Day 15 Cavg was not available for one patient (NA).

Close modal

BRAF and NRAS tumor mutation status was determined for the patients. Ten (40%) patients had activating BRAF mutations, 5 (20%) had NRAS mutations, 8 (32%) had wild-type BRAF and NRAS genes; mutation status could not be determined in 2 patients (8%). There was no significant difference in median PFS (P = 0.34, Fig. 1B) or OS (P = 0.98, Supplementary Fig. S1) by mutation status. Tumor mutation status was also not associated with the chance of achieving disease stabilization as a best response (P = 0.75, Supplementary Table S1). A review of patient records identified 9 patients with clinical testing for c-KIT gene mutations. One patient, who had wild-type BRAF and NRAS genes, was found to have a c-KIT L576P mutation. This patient had stable disease as the best response and remained on treatment for 8 cycles before progressing, which was the longest PFS of any patient in the trial. None of the other patients with c-KIT testing results had a mutation in the gene.

Ten patients with pretreatment tumor biopsies also underwent an optional tumor biopsy on day 15 of cycle 1. Four patients underwent paired core needle biopsies of lymph node metastases, and 6 underwent paired punch biopsies of cutaneous metastases. Review of the core needle biopsies showed that none of the samples contained isolates of more than 70% viable tumor of sufficient size for the planned proteomic analyses, whereas 4 of the 6 pairs of punch biopsies were sufficient for RPPA. Two of the tumors were BRAF-mutant (1 V600E, 1 V600K), 1 was NRAS-mutant (Q61K), and 1 was wild type for both genes. The NRAS mutant had stable disease as best response and received 4 cycles of treatment; the other 3 patients had progressive disease and received only 2 cycles. Comparison of the levels of the ratio of phospho:total S6 protein showed that all 4 paired samples had at least a 25% decrease, and 2 had at least 75% decrease, in P-S6 following treatment with sorafenib and temsirolimus, consistent with inhibition of mTOR by temsirolimus (Fig. 1C). In contrast, only 1 of the 4 samples showed a decrease of phospho:total ERK levels of greater than 25% (wild-type, progressive disease; Fig. 1D).

Clinical experiences in multiple cancers have shown the promise of targeted therapies. However, these experiences have also identified the frequent, and often rapid, emergence of resistance as an important challenge (29). On the basis of pattern of molecular alterations observed in melanoma and the results of preclinical studies, we carried out a phase I clinical trial to determine the efficacy and toxicity of combined treatment with sorafenib, an inhibitor of BRAF and other kinases, and temsirolimus, an mTOR inhibitor, in patients with advanced melanoma. Our study identified significant toxicity with the combination at higher doses and failed to achieve clinical responses in any of the patients. Biomarker analyses failed to identify any relationship between activating mutations in the RAS-RAF-MEK-ERK signaling pathway with PFS. Furthermore, pharmacodynamic analysis showed that the combination achieved minimal inhibition of ERK activation.

Sorafenib was the first BRAF inhibitor to undergo clinical testing in melanoma following the discovery of frequent activating BRAF mutations in this disease. Although it had shown promising results in preclinical models, only 1 of 37 metastatic melanoma patients achieved a clinical response with sorafenib monotherapy (15). Recently, the mutant-selective, second-generation BRAF inhibitors vemurafenib (also known as PLX4032 or RG7204) and GSK2118436 have shown dramatic efficacy in metastatic melanoma patients with BRAF mutations (29, 30). In the phase III trial comparing vemurafenib to dacarbazine, treatment with vemurafenib resulted in significant improvement in the clinical response rate, PFS, and OS (31). Analysis of patients treated with vemurafenib in a phase I study showed that clinical responses correlated with achievement of more than 80% inhibition of P-ERK expression in the tumor cells (17). Preclinical studies showed that sorafenib inhibits P-ERK in human melanoma cell lines with activating BRAF mutations in vitro (14). In contrast to both the vemurafenib trial and the preclinical studies, in this trial of combined treatment with sorafenib and temsirolimus quantitative analysis of P-ERK levels in 4 patients with evaluable matching pretreatment and day 15 biopsies showed less than 10% inhibition in 3 of the patients, and 46% inhibition in one. Pharmacokinetic analysis found that the maximum and average levels of sorafenib on day 15 detected in this trial (Table 5) are similar to those previously reported in sorafenib single-agent phase I trials (32). Thus, it is unlikely that the failure of sorafenib to inhibit P-ERK is due to a pharmacokinetic interaction with temsirolimus. Although the in vivo pharmacodynamic failure of sorafenib to inhibit the RAS-RAF-MEK-ERK pathway has been hypothesized to contribute to its lack of efficacy, the results of this trial provide direct evidence to support this conclusion. These findings are consistent with an analysis of matched specimens from melanoma patients treated with sorafenib that did not identify significant changes in Ki67 or cyclin D1 expression, although these are not direct pharmacodynamic readouts of RAF kinases (16). Another study reported a positive correlation between pretreatment expression levels of VEGFR2 and clinical responsiveness to treatment with sorafenib, paclitaxel, and carboplatin (33). VEGFR2 is a known target of sorafenib, and likely it is responsible for its clinical activity in renal cell carcinoma. In all 3 studies, the presence of activating BRAF mutations failed to correlate with clinical benefit from sorafenib. Taken together, the data supports that sorafenib should not be used to inhibit the RAS-RAF-MEK-ERK pathway in melanoma patients, particularly those with activating BRAF mutations.

In contrast to sorafenib, the measurement of P-S6 levels in the biopsies supports that temsirolimus was able to inhibit mTOR signaling at doses that were tolerated in this trial. This result is consistent with clinical studies with temsirolimus in other cancers (22, 23). Although mTOR inhibitors have been well tolerated as single agents, they have failed to show significant clinical activity in several solid tumors, including melanoma (21). Preclinical studies showed that inhibition of the mTORC1 complex, which occurs with rapalogs such as temsirolimus, disrupts a negative feedback loop, resulting in hyperactivation of AKT (34). Consistent with this, biopsies from patients treated with everolimus, another mTOR inhibitor, showed a decrease in P-S6, but an increase in P-AKT Ser473 (35). In our study, there was no increase in P-AKT Ser473 levels with combined treatment with temsirolimus and sorafenib (Supplementary Fig. S2). It is possible that concurrent treatment with sorafenib affected this feedback loop, as there seem to be multiple interactions between the RAS-RAF-MEK-ERK and PI3K-AKT-mTOR signaling pathways (10, 36, 37). However, a similar lack of increase in P-AKT Ser473 following single-agent temsirolimus was also recently observed in matching biopsies in head and neck cancer patients (23).

Although there was no significant association between BRAF or NRAS mutations and disease stabilization or PFS, the patient in the trial with the longest treatment duration (8 months) was found to have a c-KIT L576P mutation. Two prior reports have described clinical responses and symptomatic benefit from sorafenib in metastatic melanoma patients with c-KIT V560D (38) and c-KIT D820Y (39) mutations, respectively. Interestingly, a biopsy done after progression on sorafenib in this patient identified the continued presence of the c-KIT L576P mutation and failed to identify any secondary mutations (40). As secondary resistance to KIT inhibitors in melanoma patients with c-KIT mutations is frequent, and as yet cannot be attributed to the presence of additional mutations in the c-KIT gene, it is possible that combinatorial approaches involving inhibitors targeting other pathways may be clinically beneficial in these patients.

In conclusion, we report here that treatment with sorafenib and temsirolimus is toxic and provides minimal clinical benefit in patients with advanced melanoma. This regimen was developed to test the antitumor effects of combined inhibition of the RAS-RAF-MEK-ERK and the PI3K-AKT-mTOR signaling pathways. On the basis of our pharmacodynamic analysis, we do not believe that this regimen achieved enough inhibition of the RAS-RAF-MEK-ERK signaling pathway to test the clinical benefit of such a therapeutic approach. A number of additional potent inhibitors against each of these pathways have become available for clinical testing since the design of this trial, and, likely, will be tested in combinatorial approaches in the near future. Our results, and the studies that led to the development of clinically effective BRAF inhibitors (17), support the critical need for pharmacodynamic evaluations to appropriately interpret the results of such trials.

M.A. Davies received commercial research grants from GlaxoSmithKline, AstraZeneca, Merck, and Roche. He is a consultant and on the advisory board of GlaxoSmithKline. K.B. Kim is a consultant and on the advisory board of Bayer and Onyx.

This work was supported in part by NCI grants UO1 CA062461 and N01 CM17003, the NIH through the MD Anderson Cancer Center support grant CA016672, the P50 SPORE Career Development Grant (K.B. Kim), the ASCO Young Investigator Award (M.A. Davies), and the MD Anderson Cancer Center Physician-Scientist Program (M.A. Davies, A.J. Lazar). The MDACC RPPA Core Facility is supported by M. D. Anderson Cancer Center Core Grant NIH-NCI-CA-16672.

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.
Hocker
T
,
Tsao
H
. 
Ultraviolet radiation and melanoma: a systematic review and analysis of reported sequence variants
.
Hum Mutat
2007
;
28
:
578
88
.
2.
Bos
JL
. 
ras oncogenes in human cancer: a review
.
Cancer Res
1989
;
49
:
4682
9
.
3.
Rodriguez-Viciana
P
,
Warne
PH
,
Dhand
R
,
Vanhaesebroeck
B
,
Gout
I
,
Fry
MJ
, et al
Phosphatidylinositol-3-OH kinase as a direct target of Ras
.
Nature
1994
;
370
:
527
32
.
4.
Engelman
JA
,
Chen
L
,
Tan
X
,
Crosby
K
,
Guimaraes
AR
,
Upadhyay
R
, et al
Effective use of PI3K and MEK inhibitors to treat mutant Kras G12D and PIK3CA H1047R murine lung cancers
.
Nat Med
2008
;
14
:
1351
6
.
5.
Tsao
H
,
Goel
V
,
Wu
H
,
Yang
G
,
Haluska
FG
. 
Genetic interaction between NRAS and BRAF mutations and PTEN//MMAC1 inactivation in melanoma
.
J Investig Dermatol
2004
;
122
:
337
41
.
6.
Tsao
H
,
Zhang
X
,
Fowlkes
K
,
Haluska
FG
. 
Relative reciprocity of NRAS and PTEN/MMAC1 alterations in cutaneous melanoma cell lines
.
Cancer Res
2000
;
60
:
1800
4
.
7.
Davies
MA
,
Stemke-Hale
K
,
Lin
E
,
Tellez
C
,
Deng
W
,
Gopal
YN
, et al
Integrated molecular and clinical analysis of AKT activation in metastatic melanoma
.
Clin Cancer Res
2009
;
15
:
7538
46
.
8.
Dankort
D
,
Curley
DP
,
Cartlidge
RA
,
Nelson
B
,
Karnezis
AN
,
Damsky
WE
 Jr
, et al
Braf(V600E) cooperates with Pten loss to induce metastatic melanoma
.
Nat Genet
2009
;
41
:
544
52
.
9.
Paraiso
KH
,
Xiang
Y
,
Rebecca
VW
,
Abel
EV
,
Chen
A
,
Munko
AC
, et al
PTEN loss confers BRAF inhibitor resistance to melanoma cells through the suppression of BIM expression
.
Cancer Res
2011
;
71
:
2750
60
.
10.
Gopal
YN
,
Deng
W
,
Woodman
SE
,
Komurov
K
,
Ram
P
,
Smith
PD
, et al
Basal and treatment-induced activation of AKT mediates resistance to cell death by AZD6244 (ARRY-142886) in Braf-mutant human cutaneous melanoma cells
.
Cancer Res
2010
;
70
:
8736
47
.
11.
Villanueva
J
,
Vultur
A
,
Lee
JT
,
Somasundaram
R
,
Fukunaga-Kalabis
M
,
Cipolla
AK
, et al
Acquired resistance to BRAF inhibitors mediated by a RAF kinase switch in melanoma can be overcome by cotargeting MEK and IGF-1R/PI3K
.
Cancer Cell
2010
;
18
:
683
95
.
12.
Kim
SJ
,
Kim
JS
,
Kim
SW
,
Brantley
E
,
Yun
SJ
,
He
J
, et al
Macitentan (ACT-064992), a tissue-targeting endothelin receptor antagonist, enhances therapeutic efficacy of paclitaxel by modulating survival pathways in orthotopic models of metastatic human ovarian cancer
.
Neoplasia
2011
;
13
:
167
79
.
13.
Strumberg
D
. 
Preclinical and clinical development of the oral multikinase inhibitor sorafenib in cancer treatment
.
Drugs Today (Barc)
2005
;
41
:
773
84
.
14.
Karasarides
M
,
Chiloeches
A
,
Hayward
R
,
Niculescu-Duvaz
D
,
Scanlon
I
,
Friedlos
F
, et al
B-RAF is a therapeutic target in melanoma
.
Oncogene
2004
;
23
:
6292
8
.
15.
Eisen
T
,
Ahmad
T
,
Flaherty
KT
,
Gore
M
,
Kaye
S
,
Marais
R
, et al
Sorafenib in advanced melanoma: a phase II randomised discontinuation trial analysis
.
Br J Cancer
2006
;
95
:
581
6
.
16.
Ott
PA
,
Hamilton
A
,
Min
C
,
Safarzadeh-Amiri
S
,
Goldberg
L
,
Yoon
J
, et al
A phase II trial of sorafenib in metastatic melanoma with tissue correlates
.
PLoS One
2010
;
5
:
e15588
.
17.
Bollag
G
,
Hirth
P
,
Tsai
J
,
Zhang
J
,
Ibrahim
PN
,
Cho
H
, et al
Clinical efficacy of a RAF inhibitor needs broad target blockade in BRAF-mutant melanoma
.
Nature
2010
;
467
:
596
9
.
18.
Clark
JW
,
Eder
JP
,
Ryan
D
,
Lathia
C
,
Lenz
H-J
. 
Safety and pharmacokinetics of the dual action Raf kinase and vascular endothelial growth factor receptor inhibitor, BAY 43-9006, in patients with advanced, refractory solid tumors
.
Clin Cancer Res
2005
;
11
:
5472
80
.
19.
Lasithiotakis
KG
,
Sinnberg
TW
,
Schittek
B
,
Flaherty
KT
,
Kulms
D
,
Maczey
E
, et al
Combined inhibition of MAPK and mTOR signaling inhibits growth, induces cell death, and abrogates invasive growth of melanoma cells
.
J Invest Dermatol
2008
;
128
:
2013
23
.
20.
DiVito
KA
,
Charette
LA
,
Rimm
DL
,
Camp
RL
. 
Long-term preservation of antigenicity on tissue microarrays
.
Lab Invest
2004
;
84
:
1071
8
.
21.
Margolin
K
,
Longmate
J
,
Baratta
T
,
Synold
T
,
Christensen
S
,
Weber
J
, et al
CCI-779 in metastatic melanoma: a phase II trial of the California Cancer Consortium
.
Cancer
2005
;
104
:
1045
8
.
22.
Peralba
JM
,
deGraffenried
L
,
Friedrichs
W
,
Fulcher
L
,
Grünwald
V
,
Weiss
G
, et al
Pharmacodynamic evaluation of CCI-779, an inhibitor of mTOR, in cancer patients
.
Clin Cancer Res
2003
;
9
:
2887
92
.
23.
Fergenbaum
JH
,
Garcia-Closas
M
,
Hewitt
SM
,
Lissowska
J
,
Sakoda
LC
,
Sherman
ME
. 
Loss of antigenicity in stored sections of breast cancer tissue microarrays
.
Cancer Epidemiol Biomarkers Prev
2004
;
13
:
667
72
.
24.
Hennessy
BT
,
Lu
Y
,
Gonzalez-Angulo
AM
,
Carey
MS
,
Myhre
S
,
Ju
Z
, et al
A technical assessment of the utility of reverse phase protein arrays for the study of the functional proteome in non-microdissected human breast cancers
.
Clin Proteomics
2011
;
6
:
129
51
.
25.
Tibes
R
,
Qiu
Y
,
Lu
Y
,
Hennessy
B
,
Andreeff
M
,
Mills
GB
, et al
Reverse phase protein array: validation of a novel proteomic technology and utility for analysis of primary leukemia specimens and hematopoietic stem cells
.
Mol Cancer Ther
2006
;
5
:
2512
21
.
26.
Eisenhauer
EA
,
Therasse
P
,
Bogaerts
J
,
Schwartz
LH
,
Sargent
D
,
Ford
R
, et al
New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1)
.
Eur J Cancer
2009
;
45
:
228
47
.
27.
Davies
MA
,
Stemke-Hale
K
,
Tellez
C
,
Calderone
TL
,
Deng
W
,
Prieto
VG
, et al
A novel AKT3 mutation in melanoma tumours and cell lines
.
Br J Cancer
2008
;
99
:
1265
8
.
28.
Strumberg
D
,
Voliotis
D
,
Moeller
JG
,
Hilger
RA
,
Richly
H
,
Kredtke
S
, et al
Results of phase I pharmacokinetic and pharmacodynamic studies of the Raf kinase inhibitor BAY 43-9006 in patients with solid tumors
.
Int J Clin Pharmacol Ther
2002
;
40
:
580
1
.
29.
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
.
30.
Kefford
RF
,
Arkenau
H
,
Brown
MP
,
Milward
M
,
Infante
JR
,
Long
GV
, et al
Phase I/II study of GSK2118436, a selective inhibitor of oncogenic mutant BRAF kinase, in patients with metastatic melanoma and other solid tumors
.
J Clin Oncol
2010
;
28
:
8503
.
31.
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
.
32.
Strumberg
D
,
Clark
JW
,
Awada
A
,
Moore
MJ
,
Richly
H
,
Hendlisz
A
, et al
Safety, pharmacokinetics, and preliminary antitumor activity of sorafenib: a review of four phase I trials in patients with advanced refractory solid tumors
.
Oncologist
2007
;
12
:
426
37
.
33.
Jilaveanu
L
,
Zito
C
,
Lee
SJ
,
Nathanson
KL
,
Camp
RL
,
Rimm
DL
, et al
Expression of sorafenib targets in melanoma patients treated with carboplatin, paclitaxel and sorafenib
.
Clin Cancer Res
2009
;
15
:
1076
85
.
34.
O'Reilly
KE
,
Rojo
F
,
She
QB
,
Solit
D
,
Mills
GB
,
Smith
D
, et al
mTOR inhibition induces upstream receptor tyrosine kinase signaling and activates Akt
.
Cancer Res
2006
;
66
:
1500
8
.
35.
Tabernero
J
,
Rojo
F
,
Calvo
E
,
Burris
H
,
Judson
I
,
Hazell
K
, et al
Dose- and schedule-dependent inhibition of the mammalian target of rapamycin pathway with everolimus: a phase I tumor pharmacodynamic study in patients with advanced solid tumors
.
J Clin Oncol
2008
;
26
:
1603
10
.
36.
Lu
Y
,
Muller
M
,
Smith
D
,
Dutta
B
,
Komurov
K
,
Iadevaia
S
, et al
Kinome siRNA-phosphoproteomic screen identifies networks regulating AKT signaling
.
Oncogene
2011
;
30
:
4567
77
.
37.
Mirzoeva
OK
,
Das
D
,
Heiser
LM
,
Bhattacharya
S
,
Siwak
D
,
Gendelman
R
, et al
Basal subtype and MAPK/ERK kinase (MEK)-phosphoinositide 3-kinase feedback signaling determine susceptibility of breast cancer cells to MEK inhibition
.
Cancer Res
2009
;
69
:
565
72
.
38.
Quintas-Cardama
A
,
Lazar
AJ
,
Woodman
SE
,
Kim
K
,
Ross
M
,
Hwu
P
. 
Complete response of stage IV anal mucosal melanoma expressing KIT Val560Asp to the multikinase inhibitor sorafenib
.
Nat Clin Pract
2008
;
5
:
737
40
.
39.
Handolias
D
,
Hamilton
AL
,
Salemi
R
,
Tan
A
,
Moodie
K
,
Kerr
L
, et al
Clinical responses observed with imatinib or sorafenib in melanoma patients expressing mutations in KIT
.
Br J Cancer
2010
;
102
:
1219
23
.
40.
Woodman
SE
,
Trent
JC
,
Stemke-Hale
K
,
Lazar
AJ
,
Pricl
S
,
Pavan
GM
, et al
Activity of dasatinib against L576P KIT mutant melanoma: molecular, cellular, and clinical correlates
.
Mol Cancer Ther
2009
;
8
:
2079
85
.