This study was designed to evaluate the safety, pharmacokinetics, and clinical activity of pazopanib combined with paclitaxel to determine the recommended phase II dose in the first-line setting in patients with advanced solid tumors. Patients were enrolled in a 3+3 dose-escalation design to determine the maximum tolerated regimen (MTR) of once daily pazopanib plus paclitaxel administered every 3 weeks at four dose levels (DL1-4). Safety, pharmacokinetics, pharmacogenetics, and disease assessments were performed. Twenty-eight patients received treatment. One patient at DL1 had dose-limiting toxicity (DLT) of elevated hepatic enzymes. After pazopanib discontinuation, liver enzyme concentrations remained high until a concurrent medication, simvastatin, was discontinued. This patient had the defective CYP2C8*3*3 genotype. At DL2, 1 patient had DLT of elevated hepatic enzymes with rash and 1 patient had DLT of rash. The MTR was paclitaxel 150 mg/m2 plus pazopanib 800 mg. The most common toxicities were alopecia, fatigue, hypertension, nausea, diarrhea, dysgeusia, neutropenia, myalgia, hair color changes, and peripheral neuropathy. Coadministration of pazopanib and paclitaxel resulted in a 38% increase in systemic exposure to paclitaxel, relative to administration of paclitaxel alone, at the MTR. Of the 28 patients treated with the combination, 10 achieved a partial response and 10 achieved stable disease of ≥12 weeks. Pazopanib 800 mg daily plus paclitaxel 150 mg/m2 every 3 weeks was the recommended phase II dose, with a manageable safety profile, and with clinical activity in both melanoma and non–small cell lung cancer that suggest further evaluation of this combination is warranted. Mol Cancer Ther; 14(2); 461–9. ©2014 AACR.

Angiogenesis remains an important pathway in tumor initiation, growth, and response to therapy. VEGF is a potent endothelial-specific angiogenic factor that is an important regulator of the angiogenic process (1, 2), and elevated VEGFR levels have been found to confer a poor prognosis in many solid tumors, including non–small cell lung cancer (NSCLC) and melanoma (3, 4).

Randomized controlled trials have demonstrated the clinical benefit of inhibition of the VEGF pathway by small-molecule multitargeted tyrosine kinase inhibitors (TKI), and a number of these agents, including sunitinib, sorafenib, pazopanib, axitinib, and vandetanib, are approved as monotherapy for a variety of advanced solid tumors (5–11). However, despite demonstration of single-agent activity, combination with standard chemotherapy regimens for these agents in other tumor types has been challenging. In some cases, combinations have not been tolerable (12, 13), whereas in other cases the combination was tolerable, but failed to deliver efficacy. For example, sorafenib had been reported to have promising activity in early-phase studies for NSCLC (14, 15). Yet, when sorafenib was added to paclitaxel and carboplatin in the first-line setting for NSCLC, the primary endpoint of improved overall survival (OS) or even progression-free survival (PFS) was not met. Furthermore, in the subgroup of patients with squamous cell histology, the sorafenib combination was associated with increased mortality (16). In contrast, bevacizumab, a monoclonal antibody to VEGF-A, when combined with paclitaxel and carboplatin, demonstrated a statistically significant survival advantage (median OS, 12.3 vs. 10.3 months) over chemotherapy alone (17) in patients with non-squamous NSCLC, whereas patients with squamous cell NSCLC did not tolerate this combination (18).

Pazopanib (Votrient) is an orally bioavailable, small molecule, competitive TKI of VEGFR (−1, −2, and −3), platelet-derived growth factor receptor (PDGFR) α-, β-, and c-Kit (19), which is approved as monotherapy at a dose of 800 mg daily for the treatment of patients with advanced renal cell carcinoma (RCC; ref. 8) and advanced soft-tissue sarcoma (STS) who have received prior chemotherapy (9). Short-term treatment with pazopanib 800 mg demonstrated single-agent activity in patients with early-stage NSCLC in the preoperative setting in a proof-of-concept study that supported further exploration of pazopanib in NSCLC (20).

Because paclitaxel is a backbone of standard chemotherapeutic regimens used in a number of malignancies, including NSCLC, we had an interest in exploring the combination of pazopanib and paclitaxel in solid tumors. In addition, preclinical evidence suggested the possibility of synergy from the combination of antiangiogenic agents with taxanes (21), and recent data suggested synergism between paclitaxel and pazopanib via inhibition of aurora A in anaplastic thyroid cancer (22). Previous studies demonstrated that pazopanib could not be readily combined with paclitaxel 175 mg/m2 and carboplatin area under the plasma drug concentration curve (AUC) 5 administered every 3 weeks at doses higher than pazopanib 200 mg (23, 24). However, it was feasible to administer pazopanib 800 mg with a weekly regimen of paclitaxel 80 mg/m2, which resulted in a 26% higher geometric mean paclitaxel AUC that was similar to the systemic exposure of a paclitaxel dose of 100 mg/m2 (25).

This study was designed to evaluate the safety of pazopanib in combination with paclitaxel administered every 3 weeks, and to determine the recommended phase II dose for this combination in the first-line setting in patients with advanced solid tumors. Because of the expected increase in exposure of paclitaxel when administered in combination with pazopanib, careful dose-escalation and real-time pharmacokinetic analyses were performed.

Patients with previously untreated advanced solid tumors for whom paclitaxel-based therapy was considered appropriate; age ≥18 years; Eastern Cooperative Oncology Group performance status (ECOG PS) 0 or 1; measurable disease per RECIST (version 1.0; ref. 26); adequate bone marrow, hepatic, and renal function; and life expectancy ≥12 weeks were eligible. Exclusion criteria included clinically significant gastrointestinal abnormalities; poorly controlled hypertension; a history of cerebrovascular accident, including transient ischemic attack, pulmonary embolism or untreated deep venous thrombosis, and cardiac dysfunction within the past 6 months; evidence of active bleeding or bleeding diathesis; recent hemoptysis; and known endobronchial lesions. Coadministration of pazopanib plus paclitaxel with strong CYP3A4 inhibitors was prohibited beginning 14 days before the first dose of study drug until the end of study treatment.

This study (Clinicaltrials.gov NCT00866528) was conducted in accordance with the Declaration of Helsinki and the International Conference on Harmonization Good Clinical Practice guidelines, and was approved by institutional review boards. All patients provided written informed consent before any study procedures were performed.

Study design and treatment

This was an open-label, multicenter, phase I study of pazopanib in combination with paclitaxel. Pazopanib (Votrient; GlaxoSmithKline) was administered orally once daily starting on day 2 of cycle 1 in combination with paclitaxel [TAXOL (paclitaxel) INJECTION, Bristol–Myers Squibb Company) administered as a 3-hour i.v. infusion on day 1 of each 3-week treatment cycle for up to 6 cycles. After the 6 cycles, single-agent pazopanib treatment continued until disease progression, unacceptable toxicities, or death. Initial doses tested were pazopanib 800 mg plus paclitaxel 135 mg/m2 and a standard 3+3 dose-escalation design was applied.

Dose-limiting toxicity (DLT) was defined as one of the following events occurring within cycle 1 (a grade 2 or higher toxicity occurring after cycle 1 that was considered dose limiting by the investigator may also have been a DLT): febrile neutropenia, grade 4 granulocytopenia (>5 days), grade 4 thrombocytopenia, grade 3 or 4 clinically significant non-hematologic toxicity [excluding transient grade 3 alanine aminotransferase (ALT) or aspartate aminotransferase (AST) elevations without elevated bilirubin, grade 3 hypertension adequately controlled with anti-hypertensive medications, asymptomatic grade 3 proteinuria that improved within 14 days in the absence of hypertension and/or renal impairment, grade 3 nausea, vomiting, or diarrhea for which adequate supportive therapy had not been instituted, and alopecia], and the inability to begin the next course of treatment within 2 weeks of scheduled dosing due to toxicity. The maximum tolerated regimen (MTR) was defined as the highest combined dose of pazopanib and paclitaxel in which ≤1 of 6 patients experienced a DLT; once the MTR was reached, 6 additional patients were treated at the MTR.

Dose modification guidelines for adverse events were prespecified. Dose interruptions or reductions of pazopanib (in 200 mg decrements) were required following potential drug-related toxicities, including hypertension, proteinuria, hepatotoxicity, bleeding events, vascular thrombosis, thrombocytopenia, and neutropenia; 400 mg was the lowest dose of pazopanib permitted. Cycle delays for paclitaxel or interruption of pazopanib treatment for up to 14 days were permitted for recovery from adverse events. Patients with hepatotoxicity were evaluated for symptoms of hypersensitivity, and investigations, including liver imaging, viral serology (hepatitis A, B, and C; Epstein-Barr virus; cytomegalovirus), antinuclear antibody (ANA), pharmacokinetics, and pharmacogenetic testing were performed to rule out other contributing causes.

Endpoints and safety and response assessments

The primary outcome measure was safety and tolerability of pazopanib in combination with paclitaxel based on the frequency and nature of DLTs, adverse events (graded according to NCI CTCAE version 3.0), vital signs, electrocardiograms, and clinical laboratory parameters. Safety assessments were performed every week until completion of cycle 2, on days 1 and 8 of subsequent cycles, and then every 4 weeks during pazopanib monotherapy treatment.

Disease assessments were performed approximately every 6 weeks for the first 18 weeks and every 8 weeks thereafter until disease progression. Tumor response was assessed according to RECIST, version 1.0 (26).

Pharmacokinetic assessments

To determine the effect of pazopanib on the pharmacokinetics of paclitaxel, blood samples (2 mL) were collected for the analysis of plasma paclitaxel concentrations on day 1, cycle 1 (paclitaxel alone) and on day 1, cycle 2 (paclitaxel plus pazopanib) pre-dose, and at the following times after the start of the paclitaxel infusion: 0.5, 1, 2, 3, 3.5, 4, 4.5, 5, 6, 8 to 10, and 24 hours. To estimate the pharmacokinetics of pazopanib in the presence of paclitaxel, additional blood samples (2 mL) were collected on day 1, cycle 2 pre-dose, and 1, 2, 4, 8, and 24 hours after the start of the paclitaxel infusion. Plasma samples were analyzed for pazopanib using a validated analytic method based on protein precipitation followed by high-performance liquid chromatography tandem mass spectrometry (HPLC/MS-MS) analysis. Plasma samples were analyzed for paclitaxel by Advion Bioanalytical Labs, using a validated analytic method based on solid-phase extraction followed by HPLC/MS-MS analysis.

To investigate observed hepatotoxicity in 1 patient, three plasma samples obtained during the course of the event were analyzed for pazopanib and were also analyzed for simvastatin and simvastatin acid using a validated analytic method based on solid-phase extraction followed by HPLC/MS-MS analysis.

Pharmacogenetic assessments

The evaluation of the association of genetic variations in host DNA with safety, tolerability, and pharmacokinetics was an exploratory objective in this study. Each patient had a 10-mL blood sample taken for analysis. DNA extraction was performed by Covance. The UGT1A1 and HFE markers were genotyped by GlaxoSmithKline Genetics, using the Third Wave Invader assay and TaqMan SNP Genotyping assays, respectively. Polymorphisms in ABCB1, ABCG2, CYP2C8, CYP3A4, and SLCO1B1 were evaluated via the Affymetrix DMET Plus Array by Expression Analysis.

Statistical analysis

Analyses were based on all patients who received at least one dose of pazopanib and one dose of paclitaxel within at least one cycle of treatment. Pharmacokinetic parameters were calculated by standard noncompartmental methods using WinNonlin Professional Edition version 5.2 (Pharsight Corporation). The objective response rate (ORR), defined as the percentage of patients achieving either a complete or partial response (CR or PR) was summarized overall and for the subgroups of patients with NSCLC and melanoma. A patient was defined as a responder if he/she sustained a CR or PR, which was confirmed after no less than 28 days. Approximately 95% confidence intervals (CI) for response rates were calculated for each treatment but no P values were calculated. Waterfall plots of the percentage of change at the maximum reduction from baseline in tumor measurement were produced separately for each tumor type.

Patient characteristics

Thirty patients were enrolled from four study sites from July 2009 to May 2011. Two patients did not receive pazopanib and were not included in the analyses. All patients had a diagnosis of either NSCLC or melanoma. Table 1 summarizes patient characteristics.

Table 1.

Patient baseline and disease characteristics

Characteristics
 Treated patients, N 28 
  Male, n (%) 15 (54) 
  Female, n (%) 13 (46) 
 Median age, y (range) 57.5 (31–80) 
 ECOG PS, n (%)  
  0 19 (68) 
  1 9 (32) 
 Baseline LDH (Melanoma, n = 17), n (%)  
  Normal 9 (53) 
  Elevated (>ULN) 8 (43) 
 Primary tumor type and histology, n (%)  
  Melanoma stage 17 (61) 
   Stage IIIc 2 (7) 
   Stage IV 15 (54) 
  Melanoma histology 17 (61) 
   Malignant melanoma NOS 12 (43) 
   Spindle cell melanoma 2 (7) 
   Other/missing 3 (11) 
  NSCLC Stage IV 11 (39) 
   Adenocarcinoma 6 (21) 
   Poorly differentiated adenocarcinoma 2 (7) 
   Squamous cell carcinoma 2 (7) 
   Papillary adenocarcinoma 1 (4) 
Characteristics
 Treated patients, N 28 
  Male, n (%) 15 (54) 
  Female, n (%) 13 (46) 
 Median age, y (range) 57.5 (31–80) 
 ECOG PS, n (%)  
  0 19 (68) 
  1 9 (32) 
 Baseline LDH (Melanoma, n = 17), n (%)  
  Normal 9 (53) 
  Elevated (>ULN) 8 (43) 
 Primary tumor type and histology, n (%)  
  Melanoma stage 17 (61) 
   Stage IIIc 2 (7) 
   Stage IV 15 (54) 
  Melanoma histology 17 (61) 
   Malignant melanoma NOS 12 (43) 
   Spindle cell melanoma 2 (7) 
   Other/missing 3 (11) 
  NSCLC Stage IV 11 (39) 
   Adenocarcinoma 6 (21) 
   Poorly differentiated adenocarcinoma 2 (7) 
   Squamous cell carcinoma 2 (7) 
   Papillary adenocarcinoma 1 (4) 

Abbreviations: LDH, lactic dehydrogenase; NOS, not otherwise specified.

Dose escalation and toxicity

Four dose levels were explored and are shown in Table 2 together with DLTs and neutrophil counts for baseline and cycle 1 nadir (day 15) values. The first DLT occurred at the lowest dose level tested (cohort 1, PAC 135/PAZ 800) leading to expansion of this cohort. Two DLTs occurred at the next (and highest) dose level tested (cohort 2, PAC 175/PAZ 800); subsequently two intermediate dose levels were explored (cohort 3, PAC 150/PAZ 800; and cohort 4, PAC 175/PAZ 400) with no further DLTs reported. Both cohorts 3 and 4 qualified for the MTR; however, cohort 3 was selected for expansion to a total of 12 patients and subsequently determined to be the recommended dose for the pazopanib and paclitaxel combination.

Table 2.

DLTs and absolute neutrophil counts by dose level

ANC median (range), GI/L
CohortDose levelPatients treated (n)Patients with DLT (n)BaselineCycle 1, day 15, nadir
Paclitaxel 135 mg/m2 + Pazopanib 800 mg 1a 4.71 (3.00–8.70) 1.45 (0.51–3.50) 
Paclitaxel 175 mg/m2 + Pazopanib 800 mg 2b 4.73 (3.80–6.10)c 0.78 (0.71–4.30)c 
Paclitaxel 150 mg/m2 + Pazopanib 800 mg 12 7.04 (2.60–11.92) 2.74 (0.90–9.60) 
Paclitaxel 175 mg/m2 + Pazopanib 400 mg 4.91 (3.17–5.60) 1.58 (0.35–4.60) 
ANC median (range), GI/L
CohortDose levelPatients treated (n)Patients with DLT (n)BaselineCycle 1, day 15, nadir
Paclitaxel 135 mg/m2 + Pazopanib 800 mg 1a 4.71 (3.00–8.70) 1.45 (0.51–3.50) 
Paclitaxel 175 mg/m2 + Pazopanib 800 mg 2b 4.73 (3.80–6.10)c 0.78 (0.71–4.30)c 
Paclitaxel 150 mg/m2 + Pazopanib 800 mg 12 7.04 (2.60–11.92) 2.74 (0.90–9.60) 
Paclitaxel 175 mg/m2 + Pazopanib 400 mg 4.91 (3.17–5.60) 1.58 (0.35–4.60) 

Abbreviations: ANC, absolute neutrophil count; PAC, paclitaxel; PAZ, pazopanib.

aDLT, grade 4 elevated hepatic enzyme (ALT and AST).

bDLTs: (i) grade 3 elevated ALT and AST with grade 2 rash; (ii) grade 3 rash.

cn = 3 patients.

The first DLT, asymptomatic grade 4 elevated hepatic enzymes, with grade 2 bilirubin elevation, occurred in cohort 1 after the administration of one dose of paclitaxel (135 mg/m2) and 21 days of pazopanib, which the patient took as 400 mg twice daily (in error) instead of 800 mg once daily. Pazopanib treatment was permanently discontinued; however, hepatic enzymes remained high until a concurrent medication, simvastatin (80 mg daily), was also discontinued (see Supplementary Table S1). Three blood samples drawn during the course of this event revealed plasma pazopanib concentrations within the expected range. However, plasma concentrations of simvastatin and simvastatin acid approximately 7 hours after the last dose of pazopanib and 16.5 hours after the last dose of simvastatin were 14.7 ng/mL (approximately 4-fold greater than expected) and 20.1 ng/mL (approximately 10-fold greater than expected), respectively (27), indicating that a drug–drug interaction between pazopanib and simvastatin was present in this patient (see Supplementary Table S2). This patient received one dose of paclitaxel in the absence of pazopanib, with pazopanib treatment starting no sooner than 24 hours after the administration of paclitaxel. Paclitaxel pharmacokinetic samples drawn over the 24 hours following the administration of paclitaxel (as a 3-hour infusion) demonstrated exposures in the expected range for paclitaxel 135 mg/m2 (peak plasma concentration at 3 hours was 3,205 ng/mL; ref. 28). Viral serology and ANA tests were negative. Pharmacogenetic analysis of a blood sample revealed wild-type genotypes for UGT1A1, HFE, CYP3A4, ABCG2 (BCRP), and SLCO1B1 (OATP1B1), and a heterozygous ABCB1 (Pgp) genotype that are expected to have normal to nearly normal activity for these enzymes and transporter proteins; however, the patient did have a homozygous defective CYP2C8*3*3 genotype, predicted to have a poor metabolizer status for CYP2C8. After resolution of this event, the patient subsequently resumed treatment with paclitaxel and simvastatin outside of the study, with no further reported hepatic enzyme elevations.

In cohort 2 (PAC 175/PAZ 800), one DLT included grade 3 increases in ALT and AST with eosinophilia and subsequent development of grade 2 pruritic rash, which led to permanent discontinuation of pazopanib. No pharmacokinetic or pharmacogenetic blood samples were available for this patient. No abnormalities were detected by transabdominal ultrasound, and viral serology and ANA tests were negative. A second DLT in this cohort was grade 3 maculapapular rash. Pazopanib treatment was interrupted, but later successfully reintroduced at a lower dose (600 mg daily). Of note, there were no DLTs of neutropenia in this study; however, the lowest median neutrophil count in cycle 1 was observed in cohort 2 (Table 2).

In cohort 3 (PAC 150/PAZ 800), two patients had grade 3 ALT elevations during the second cycle of treatment that did not meet DLT criteria. In both patients, plasma pazopanib concentrations were within the expected range of concentrations for an 800-mg dose of pazopanib; viral serology and ANA tests were negative. In one patient, treatment with pazopanib was interrupted and later restarted for cycle 3 of treatment at a reduced dose (400 mg); after rechallenge there was a mild and transient grade 2 ALT elevation. There was no elevation of bilirubin. No pharmacogenetic testing was performed. In the second patient, pazopanib treatment was discontinued. Pharmacogenetic analysis of a blood sample revealed wild-type genotypes for UGT1A1, HFE, ABCB1, CYP3A4, CYP2C8, and SLCO1B1; however, the patient was homozygous for the ABCG2 *2/*2 genotype, associated with functional impairment of the transporter.

All 28 patients who received at least one dose of both study drugs reported at least one adverse event regardless of causality during the study. Overall, the most frequently reported treatment-emergent adverse events (any grade occurring in ≥30% of patients) were alopecia (86%), fatigue (82%), hypertension (71%), nausea (68%), diarrhea (61%), vomiting (54%), dysgeusia (50%), myalgia (50%), rash (50%), neutropenia (46%), arthralgia (43%), decreased appetite (43%), hair color changes (43%), headache (43%), peripheral neuropathy (43%), pain in extremity (39%), constipation (32%), dizziness (32%), and hepatic enzyme increased (32%); most were grade 1 or 2 in severity (Table 3). At the MTR (PAC 150/PAZ 800), the incidences were fatigue (92%), alopecia (83%), nausea (75%), rash (75%), hypertension (67%), diarrhea (67%), dysgeusia (58%), myalgia (50%), vomiting (50%), neutropenia (42%), headache (42%), peripheral neuropathy (33%), arthralgia (33%), decreased appetite (33%), and hair color changes (33%). Overall, the most frequently reported treatment-emergent grade 3 and 4 adverse events were neutropenia (6 and 3 patients, respectively) and hepatic enzyme elevations (4 and 1 patient, respectively). There were no embolic or thrombotic events and no severe (grade 3 or above) hemorrhagic events reported in the study. Permanent discontinuation of study treatment due to adverse events occurred in 5 patients; three patients due to elevations in liver enzymes, one patient due to subcutaneous (paravertebral) abscess, and one patient due to tachycardia (for this patient paclitaxel was discontinued but not pazopanib).

Table 3.

Treatment-emergent adverse events reported in at least 30% of patients that had at least one occurrence of grade 3 or 4 severity

Paclitaxel 135 mg/m2 + Pazopanib 800 mg (n = 6)Paclitaxel 175 mg/m2 + Pazopanib 800 mg (n = 4)Paclitaxel 150 mg/m2 + Pazopanib 800 mg (n = 12)Paclitaxel 175 mg/m2 + Pazopanib 400 mg (n = 6)
Adverse eventaAll gradesGrade 3Grade 4Grade 3Grade 4Grade 3Grade 4Grade 3Grade 4
Any event 28 (100) 4 (67) 1 (17) 2 (50) 2 (50) 9 (75) 4 (67) 1 (17) 
Fatigue 23 (82) 1 (8) 
Hypertension 20 (71) 1 (8) 1 (17) 
Diarrhea 17 (61) 1 (17) 
Rashb 14 (50) 1 (25) 
Neutropenia 13 (46) 2 (33) 1 (25) 2 (50) 2 (17) 1 (17) 1 (17) 
Headache 12 (43) 1 (8) 1 (17) 
Peripheral neuropathy 12 (43) 1 (8) 
Pain in extremity 11 (39) 1 (17) 
Hepatic enzyme increasedc 9 (32) 1 (17) 1 (17) 1 (25) 2 (17) 
Paclitaxel 135 mg/m2 + Pazopanib 800 mg (n = 6)Paclitaxel 175 mg/m2 + Pazopanib 800 mg (n = 4)Paclitaxel 150 mg/m2 + Pazopanib 800 mg (n = 12)Paclitaxel 175 mg/m2 + Pazopanib 400 mg (n = 6)
Adverse eventaAll gradesGrade 3Grade 4Grade 3Grade 4Grade 3Grade 4Grade 3Grade 4
Any event 28 (100) 4 (67) 1 (17) 2 (50) 2 (50) 9 (75) 4 (67) 1 (17) 
Fatigue 23 (82) 1 (8) 
Hypertension 20 (71) 1 (8) 1 (17) 
Diarrhea 17 (61) 1 (17) 
Rashb 14 (50) 1 (25) 
Neutropenia 13 (46) 2 (33) 1 (25) 2 (50) 2 (17) 1 (17) 1 (17) 
Headache 12 (43) 1 (8) 1 (17) 
Peripheral neuropathy 12 (43) 1 (8) 
Pain in extremity 11 (39) 1 (17) 
Hepatic enzyme increasedc 9 (32) 1 (17) 1 (17) 1 (25) 2 (17) 

aThe following treatment-emergent adverse events reported in that at least 30% of patients did not have events with grade 3 or 4 severity: alopecia, nausea, vomiting, dysgeusia, myalgia, arthraliga, decreased appetite, hair color changes, constipation, and dizziness.

bRash also includes: rash erythematous, rash macular, rash pruritic, and exfoliative rash.

cHepatic enzyme increased also includes elevated ALT.

Treatment exposure

The median number of cycles of paclitaxel administered was 5 to 6 cycles across all cohorts, and the median dose of paclitaxel and pazopanib administered was the planned dose for each cohort, with the exception of cohort 2 in which the median pazopanib dose was lower (737 mg) than the intended dose (800 mg; see Supplementary Table S3). The median duration of pazopanib treatment was 3.8, 5.3, 6.9, and 5.0 months for cohorts 1, 2, 3, and 4, respectively. One patient with ocular melanoma continued to receive treatment with 800 mg pazopanib monotherapy for 24 months after completion of his six cycles of paclitaxel plus pazopanib; his overall treatment duration was 28 months (Fig. 1).

Figure 1.

Duration of pazopanib treatment by cohort and patient. Cohort 1, paclitaxel 135 mg/m2 + pazopanib 800 mg; cohort 2, paclitaxel 175 mg/m2 + pazopanib 800 mg; cohort 3, paclitaxel 150 mg/m2 + pazopanib 800 mg; cohort 4, paclitaxel 175 mg/m2 + pazopanib 400 mg. M, melanoma; L, NSCLC.

Figure 1.

Duration of pazopanib treatment by cohort and patient. Cohort 1, paclitaxel 135 mg/m2 + pazopanib 800 mg; cohort 2, paclitaxel 175 mg/m2 + pazopanib 800 mg; cohort 3, paclitaxel 150 mg/m2 + pazopanib 800 mg; cohort 4, paclitaxel 175 mg/m2 + pazopanib 400 mg. M, melanoma; L, NSCLC.

Close modal

Pharmacokinetics

Coadministration of pazopanib and paclitaxel resulted in an increase in systemic exposure to paclitaxel relative to administration of paclitaxel alone by approximately 38% at the MTR (Fig. 2). There was no dose- or concentration-dependent effect of pazopanib 400 or 800 mg once daily on the systemic disposition of paclitaxel (see Supplementary Table S4).

Figure 2.

Median plasma paclitaxel concentration–time profiles after administration of paclitaxel 150 mg/m2 alone (cycle 1, day 1) and in combination with pazopanib 800 mg (cycle 2, day 1).

Figure 2.

Median plasma paclitaxel concentration–time profiles after administration of paclitaxel 150 mg/m2 alone (cycle 1, day 1) and in combination with pazopanib 800 mg (cycle 2, day 1).

Close modal

There were no consistent changes in systemic exposure to pazopanib as measured by maximum concentration (Cmax), AUC from time of dose to 24 hours [AUC(0–24)], or concentration at 24 hours following administration (C24) across the paclitaxel dose levels explored. These results indicate that there was no concentration-dependent effect of paclitaxel on the systemic exposure of pazopanib.

Clinical activity

Clinical activity was observed in all four cohorts, with 10 of 28 evaluable patients achieving a PR for an ORR of 36% for the total population (Fig. 3; Supplementary Table S5), and stable disease of ≥12 weeks in 10 additional patients. In the subset of patients with NSCLC, the ORR was 45% (PR in 5 of 11 patients), and stable disease of ≥12 weeks was observed in 3 of 11 patients. In the subset of patients with melanoma, the ORR was 29% (PR in 5 of 17 patients), and stable disease of ≥12 weeks in 7 additional patients.

Figure 3.

The investigator-assessed maximum percentage of reduction from baseline in tumor measurement. A, NSCLC; B, melanoma. C1, paclitaxel 135 mg/m2 + pazopanib 800 mg; C2, paclitaxel 175 mg/m2 + pazopanib 800 mg; C3, paclitaxel 150 mg/m2 + pazopanib 800 mg; C4, paclitaxel 175 mg/m2 + pazopanib 400 mg.

Figure 3.

The investigator-assessed maximum percentage of reduction from baseline in tumor measurement. A, NSCLC; B, melanoma. C1, paclitaxel 135 mg/m2 + pazopanib 800 mg; C2, paclitaxel 175 mg/m2 + pazopanib 800 mg; C3, paclitaxel 150 mg/m2 + pazopanib 800 mg; C4, paclitaxel 175 mg/m2 + pazopanib 400 mg.

Close modal

This phase I study was conducted to evaluate the safety of pazopanib in combination with paclitaxel administered every 3 weeks and to determine the recommended phase II dose in the first-line setting in patients with advanced solid tumors. Secondary objectives included an evaluation of the pharmacokinetics of each agent and of the clinical activity of this combination.

This study demonstrated that pazopanib could be safely combined with paclitaxel administered once every 3 weeks with either full-dose pazopanib (800 mg) in combination with paclitaxel 150 mg/m2 (cohort 3) or with a lower dose of pazopanib (400 mg) in combination with a higher dose of paclitaxel 175 mg/m2 (cohort 4). Minor differences in the tolerability profile and treatment exposures led to a decision to expand the pazopanib 800 mg and paclitaxel 150 mg/m2 dose level and declare this as the recommended phase II dose for patients with advanced solid tumors. The observed pharmacokinetic interaction between pazopanib and paclitaxel supported the selection of full-dose pazopanib with a dose of paclitaxel that provides clinically relevant paclitaxel exposure.

Coadministration of paclitaxel 150 mg/m2 and pazopanib 800 mg once daily resulted in a 34% and 37% increase in paclitaxel AUC extrapolated to infinity and Cmax, respectively, relative to administration of paclitaxel alone. Consequently, although higher doses of paclitaxel (up to 225 mg/m2) may typically be administered every 3 weeks to patients with solid tumors (29, 30), the presence of pazopanib increased the exposure of a 150 mg/m2 dose of paclitaxel to a level that would be expected from a dose of 200 mg/m2 (31). A drug–drug interaction was expected, because paclitaxel is a substrate for CYP2C8 and CYP3A4, and pazopanib is a weak inhibitor of CYP3A4 and CYP2C8. The extent of the interaction observed in this study appears similar to that previously reported for pazopanib in combination with paclitaxel administered on a weekly schedule (25), and in combination with paclitaxel and carboplatin (23) administered with various doses of pazopanib. Collectively, these studies suggest that coadministration of pazopanib results in a consistent increase in systemic exposure to paclitaxel regardless of the paclitaxel dose regimen and pazopanib doses investigated.

The most common treatment-emergent toxicities observed for the combination of pazopanib and paclitaxel in this study were expected on the basis of the known safety profile of each individual agent and included alopecia, fatigue, hypertension, nausea, diarrhea, vomiting, dysgeusia, myalgia, rash, and neutropenia. Although events were mostly grade 1 or 2 in severity, the incidence of some events, including fatigue, nausea, hypertension, dysgeusia, and rash was somewhat higher than expected. In this study, a large proportion of patients (67%) entered with baseline hypertension; it is unclear whether this influenced the incidence of hypertension reported. Previous studies of the combination of pazopanib with weekly paclitaxel have also reported high incidences of fatigue and nausea (25, 32); therefore, it is possible that these events could represent potential synergistic toxicities for the combination of pazopanib and paclitaxel. Of note, hematologic toxicity was not dose limiting in this study; this contrasts with the experience with the combination of pazopanib with paclitaxel 175 mg/m2 and carboplatin AUC 5, in which myelosuppression was the DLT for the combination (23, 24). DLTs reported in this study included rash and hepatotoxicity.

Hepatotoxicity is an expected event for pazopanib, manifested by increases in serum transaminases (ALT and AST) and bilirubin, with the majority (92.5%) of transaminase elevations occurring within the first 18 weeks of treatment (33). In clinical trials in RCC and STS, approximately 18% of patients experienced elevations of ALT greater than 3× the upper limit of normal (ULN), 5% experienced elevations of ALT greater than 8 × ULN, and 2% had concurrent elevations in ALT >3 × ULN and bilirubin >2 × ULN in the absence of significant alkaline phosphatase >3 × ULN (33). Although an expected event for pazopanib, efforts were made to explore potential underlying causes of the hepatotoxicity events observed in the 4 patients who experienced grade 3 or 4 transaminase elevations. Pharmacogenetic analyses included an evaluation of genetic markers in the HFE and UGT1A1 genes, which have been associated with ALT and bilirubin elevation, respectively, in patients with RCC treated with pazopanib (34, 35); functional polymorphisms for enzymes involved in the metabolism of pazopanib and paclitaxel (CYP3A4 and CYP2C8); and transport proteins for which pazopanib and/or paclitaxel are substrates or inhibitors (ABCB1, ABCG2, and OATP1B1). In all 3 patients who provided a sample for pharmacokinetic and pharmacogenetic analysis, plasma concentrations of pazopanib were in the expected range, and in 2 patients, functional polymorphisms were detected that may have contributed to the observed liver event; one patient did not have pharmacogenetic testing performed. One patient had a functional impairment of the ABCG2 transporter protein (ABCG2 *2/*2 genotype). It is possible that the presence of this genotype potentially contributed to the observed liver event; however, no clear mechanism was identified. The second patient was homozygous for the defective CYP2C8*3*3 genotype, which is predicted to have a poor metabolizer status for CYP2C8. Of note, this patient was taking simvastatin concurrently with pazopanib. ALT levels remained elevated in this patient until simvastatin was discontinued, suggesting a key role of simvastatin administration in the occurrence of elevated ALT. Simvastatin is mainly metabolized by CYP3A to the active metabolite simvastatin acid, which is metabolized further by CYP3A4 and CYP2C8 for elimination and transported into hepatocytes by OATP1B1 (36). The poor metabolizer status for CYP2C8 coupled with potential inhibition of this enzyme and of OATP1B1 by pazopanib may explain the observed drug–drug interaction resulting in increased exposure to both simvastatin (4-fold) and simvastatin acid (10-fold) in this patient. Both pazopanib and simvastatin have been associated with transaminase elevations and may each have contributed to the observed hepatocellular injury; however, the individual contribution of each agent could not be determined.

This anecdotal observation of hepatotoxicity in conjunction with concurrent simvastatin treatment resulted in the conduct of a meta-analysis to evaluate the effects of concomitant pazopanib and statin use on the incidence of elevated ALT using data from 11 pazopanib clinical studies (37). The meta-analysis showed that the incidence of ALT elevation (≥3 × ULN) was 27% in patients receiving both pazopanib and simvastatin, which was significantly higher than the incidence of 14% in patients who did not receive statins (P = 0.04). Furthermore, the analysis demonstrated that the ABCG2 (BCRP) 421C>A polymorphism may be associated with ALT elevation in patients taking pazopanib and simvastatin; patients with the variant 421C>A allele had a higher incidence of ALT>3 × ULN (5/7, 71%) compared with those with the wild-type genotype (2/21, 10%; OR, 19.6; 95% CI, 1.9–231.6; P = 0.004). This polymorphism was not associated with ALT elevations in patients treated with pazopanib without concurrent use of statins. This analysis resulted in a label change for pazopanib describing an increased risk of ALT elevations with concomitant use of pazopanib and simvastatin. Both the case history from this study and the subsequent meta-analysis highlight the importance of signal detection, and pharmacokinetic and pharmacogenetic characterization of serious adverse events in early-phase trials to better understand potential mechanisms of injury and inform management of patients.

With the potential for synergism between antiangiogenic agents and taxanes (21, 22), the combination of pazopanib and paclitaxel offered the possibility of developing a platinum-sparing combination for first-line treatment of patients with metastatic NSCLC and other solid tumors. Although the study was open to all solid tumors, participating sites enrolled patients with only two tumor types in this study: melanoma and NSCLC. Patients with NSCLC and melanoma appeared to derive benefit from the therapy. Clinical activity was noted in all four cohorts with a best ORR of 36%. Partial responses occurred in 10 of 28 patients and stable disease >12 weeks in 10 of 28 patients.

Paclitaxel has been used in the first-line treatment of metastatic melanoma both as a single agent and in combination with carboplatin, with reported response rates of approximately 12% to 16% (38–40) and 16% to 20%, respectively (30, 41, 42). Because VEGF expression has been shown to play a role in metastatic melanoma (43), studies to explore the combination of carboplatin, paclitaxel, and antiangiogenic agents such as sorafenib and bevacizumab have been performed; however, these studies have not shown improved efficacy compared with the carboplatin and paclitaxel doublet (30, 42). In this study, the response rate for melanoma was 29% (5 of 17 patients with PR) with an additional 41% (7 of 17 patients) demonstrating SD of >12 weeks, and one patient diagnosed with ocular melanoma maintained antitumor response for more than 100 weeks. In addition, an ongoing phase II study of pazopanib in combination with a weekly regimen of paclitaxel for the first-line treatment of unresectable melanoma has reported data from a planned interim analysis in 20 patients and demonstrated a response rate of 42% (32); this study will continue to accrue 60 patients with a primary endpoint of 6-month PFS. These studies suggest that the combination of pazopanib and paclitaxel is active in melanoma and warrant further investigation.

In this study, the response rate for NSCLC was 45% (5 of 11 patients with PR) with an additional 27% (3 of 11 patients) demonstrating SD of >12 weeks. The clinical activity observed in patients with NSCLC in this study suggests further investigation of the combination of pazopanib and paclitaxel is warranted. Recent data reported with docetaxel and nintedanib in the second-line setting for advanced NSCLC demonstrated a significant improvement of both PFS and OS for the combination treatment compared with single-agent docetaxel (44). In this setting, combination treatment with docetaxel and nintedanib continued until disease progression; this approach may be worth considering in future studies of the combination of pazopanib and paclitaxel. In our phase 1 trial, 50% of patients completed the 6 cycles of combination therapy and were continued on single-agent pazopanib for a range of 2 to 24 months.

Since this study was initiated, treatment paradigms in both melanoma and NSCLC have undergone a rapid transformation. In melanoma, the emergence of newer targeted agents, including vemurafenib, dabrafenib, and trametinib, have resulted in higher response rates and improved clinical outcomes, especially for patients with V600 mutation–positive tumors (45). This study did not prospectively test for the status of V600 mutations in the melanoma subset of patients. With the availability of an approved test for this marker, future studies should incorporate evaluation of this marker. In addition, emerging data from studies in both melanoma and NSCLC with immune therapies, including ipilimumab, the PD-1 checkpoint inhibitors, and combinations of these agents, suggest that the landscape of treatment in the first-line metastatic setting for each of these tumor types will continue to change (45, 46). In the face of these changes, future studies of the combination of pazopanib and paclitaxel will need to carefully evaluate the appropriate setting for use of this combination.

In summary, this study identified pazopanib 800 mg daily and paclitaxel 150 mg/m2 administered every 3 weeks as the recommended phase II dose for patients with advanced solid tumors. The toxicity profile was consistent with the safety profile of each individual agent and was manageable; although the incidence of some toxicities may be higher than expected from single-agent treatment. In the presence of pazopanib, exposure to paclitaxel increased by 38%; however, there was no dose- or concentration-dependent effect of pazopanib 400 or 800 mg once daily on the systemic disposition of paclitaxel. Clinical activity of this combination in both melanoma and NSCLC suggests that further evaluation of this combination is warranted.

M. O'Brien is an advisory board member for GlaxoSmithKline. A.B. Suttle has ownership interest in GlaxoSmithKline. C.-F. Xu has ownership interest in GSK stocks. L. Ottesen has ownership interest in GSK stocks. M.A. Villalona-Calero reports receiving a commercial research grant from GSK. No potential conflicts of interest were disclosed by the other authors.

Conception and design: R. Plummer, R. Salgia, E.M. Paul, A.B. Suttle, L.H. Ottesen, M.A. Villalona-Calero

Development of methodology: E.M. Paul

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): K.L. Kendra, R. Plummer, R. Salgia, M.E.R. O'Brien, C.-F. Xu, L.H. Ottesen, M.A. Villalona-Calero

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): K.L. Kendra, R. Plummer, R. Salgia, M.E.R. O'Brien, E.M. Paul, A.B. Suttle, N. Compton, C.-F. Xu, L.H. Ottesen, M.A. Villalona-Calero

Writing, review, and/or revision of the manuscript: K.L. Kendra, R. Plummer, M.E.R. O'Brien, E.M. Paul, A.B. Suttle, N. Compton, C.-F. Xu, M.A. Villalona-Calero

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): K.L. Kendra, R. Salgia, M.E.R. O'Brien, E.M. Paul, M.A. Villalona-Calero

Study supervision: K.L. Kendra, R. Plummer, M.E.R. O'Brien, E.M. Paul, M.A. Villalona-Calero

The authors thank Terry Paul of Paul Medical Writing, Inc., Raleigh, North Carolina, for writing and editorial assistance (medical writing services were funded by GlaxoSmithKline), and Jennifer Thurmond for her assistance with the clinical trial patients.

This study was funded by GlaxoSmithKline.

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.
Folkman
J
. 
What is the evidence that tumors are angiogenesis dependent?
J Natl Cancer Inst
1990
;
82
:
4
6
.
2.
Majeti
BK
,
Lee
JH
,
Simmons
BH
,
Shojaei
F
. 
VEGF is an important mediator of tumor angiogenesis in malignant lesions in a genetically engineered mouse model of lung adenocarcinoma
.
BMC Cancer
2013
;
13
:
213
.
3.
Jantus-Lewintre
E
,
Sanmartín
E
,
Sirera
R
,
Blasco
A
,
Sanchez
JJ
,
Tarón
M
, et al
Combined VEGF-A and VEGFR-2 concentrations in plasma: diagnostic and prognostic implications in patients with advanced NSCLC
.
Lung Cancer
2011
;
74
:
326
31
.
4.
Tas
F
,
Duranyildiz
D
,
Oguz
H
,
Camlica
H
,
Yasasever
V
,
Topuz
E
. 
Circulating serum levels of angiogenic factors and vascular endothelial growth factor receptors 1 and 2 in melanoma patients
.
Melanoma Res
2006
;
16
:
405
11
.
5.
Motzer
RJ
,
Hutson
TE
,
Tomczak
P
,
Michaelson
MD
,
Bukowski
RM
,
Rixe
O
, et al
Sunitinib versus interferon alfa in metastatic renal-cell carcinoma
.
N Engl J Med
2007
;
356
:
115
24
.
6.
Demetri
GD
,
van Oosterom
AT
,
Garrett
CR
,
Blackstein
ME
,
Shah
MH
,
Verweij
J
, et al
Efficacy and safety of sunitinib in patients with advanced gastrointestinal stromal tumour after failure of imatinib: a randomised controlled trial
.
Lancet
2006
;
368
:
1329
38
.
7.
Escudier
B
,
Eisen
T
,
Stadler
WM
,
Szczylik
C
,
Oudard
S
,
Siebels
M
, et al
Sorafenib in advanced clear-cell renal-cell carcinoma
.
N Engl J Med
2007
;
356
:
125
34
.
8.
Sternberg
CN
,
Davis
ID
,
Mardiak
J
,
Szczylik
C
,
Lee
E
,
Wagstaff
J
, et al
Pazopanib in locally advanced or metastatic renal cell carcinoma: results of a randomized phase III trial
.
J Clin Oncol
2010
;
28
:
1061
8
.
9.
van der Graaf
WTA
,
Blay
JY
,
Chawla
SP
,
Kim
DW
,
Bui-Nguyen
B
,
Casali
PG
, et al
Pazopanib in metastatic soft tissue sarcoma (PALETTE): a randomized, double-blind, placebo controlled phase 3 trial
.
Lancet
2012
;
379
:
1879
86
.
10.
Rini
BI
,
Escudier
B
,
Tomczak
P
,
Kaprin
A
,
Szczylik
C
,
Hutson
TE
, et al
Comparative effectiveness of axitinib versus sorafenib in advanced renal cell carcinoma (AXIS): a randomized phase 3 trial
.
Lancet
2011
;
378
:
1931
9
.
11.
Wells
SA
,
Robinson
BG
,
Gagel
RF
,
Dralle
H
,
Fagin
JA
,
Santoro
M
, et al
Vandetanib in patients with locally advanced or metastatic medullary thyroid cancer: a randomized, double-blind phase III trial
.
J Clin Oncol
2011
;
30
:
134
41
.
12.
Goss
GD
,
Arnold
A
,
Shepherd
FA
,
Dediu
M
,
Ciuleanu
TE
,
Fenton
D
, et al
Randomized, double-blind trial of carboplatin and paclitaxel with either daily oral cediranib or placebo in advanced non–small cell lung cancer: NCIC clinical trials group BR24 study
.
J Clin Oncol
2010
;
28
:
49
55
.
13.
Scagliotti
GV
,
Felip
E
,
Besse
B
,
von Pawel
J
,
Mellemgaard
A
,
Reck
M
, et al
An open-label, multicenter, randomized, phase II study of pazopanib in combination with pemetrexed in first-line treatment of patients with advanced-stage non–small cell lung cancer
.
J Thorac Oncol
2013
;
8
:
1529
37
.
14.
Schiller
JH
,
Lee
JW
,
Hanna
N
,
Traynor
AM
,
Carbone
DP
. 
A randomized discontinuation phase II study of sorafenib versus placebo in patients with non–small cell lung cancer who have failed at least two prior chemotherapy regimens. E2501.
J Clin Oncol
26
, 
2008
(
suppl; abstr 8014
).
15.
Schiller
H
,
Flaherty
KT
,
Redlinger
M
,
Binger
K
,
Eun
J
,
Petrenciuc
O
, et al
Sorafenib combined with carboplatin/paclitaxel for advanced non–small cell lung cancer: a phase I subset analysis
.
J Clin Oncol
24
, 
2006
(
suppl; abstr 7194
).
16.
Scagliotti
G
,
Novello
S
,
von Pawel
J
,
Reck
M
,
Pereira
JR
,
Thomas
M
, et al
Phase III study of carboplatin and paclitaxel alone or with sorafenib in advanced non–small cell lung cancer
.
J Clin Oncol
2010
;
28
:
1835
42
.
17.
Sandler
A
,
Gray
R
,
Perry
MC
,
Brahmer
J
,
Schiller
JH
,
Dowlati
A
, et al
Paclitaxel-carboplatin alone or with bevacizumab for non–small cell lung cancer
.
N Engl J Med
2006
;
355
:
2542
50
.
18.
Johnson
DH
,
Fehrenbacher
L
,
Novotny
WF
,
Herbst
RS
,
Nemunaitis
JJ
,
Jablons
DM
, et al
Randomized phase II trial comparing bevacizumab plus carboplatin and paclitaxel with carboplatin and paclitaxel alone in previously untreated locally advanced or metastatic non–small cell lung cancer
.
J Clin Oncol
2004
;
22
:
2184
91
.
19.
Kumar
R
,
Knick
VB
,
Rudolph
SK
,
Johnson
JH
,
Crosby
RM
,
Crouthamel
MC
, et al
Pharmacokinetic-pharmacodynamic correlation from mouse to human with pazopanib, a multi-kinase angiogenesis inhibitor with potent antitumor and antiangiogenic activity
.
Mol Cancer Ther
2007
;
6
:
2012
21
.
20.
Altorki
N
,
Lane
ME
,
Bauer
T
,
Lee
PC
,
Guarino
MJ
,
Pass
H
, et al
Phase II proof-of-concept study of pazopanib monotherapy in treatment-naive patients with stage I/II resectable non–small cell lung cancer
.
J Clin Oncol
2010
;
28
:
3131
7
.
21.
Bocci
G
,
Di Paolo
A
,
Danesi
R
. 
The pharmacological bases of the antiangiogenic activity of paclitaxel
.
Angiogenesis
2013
;
16
:
481
92
.
22.
Isham
CR
,
Bossou
AR
,
Negron
V
,
Fisher
KE
,
Kumar
R
,
Marlow
L
, et al
Pazopanib enhances paclitaxel-induced mitotic catastrophe in anaplastic thyroid cancer
.
Sci Transl Med
2013
;
5
:
166ra3
.
23.
Burris
HA
 III
,
Dowlati
A
,
Moss
RA
,
Infante
JR
,
Jones
SF
,
Spigel
DR
, et al
Phase I study of pazopanib in combination with paclitaxel and carboplatin given every 21 days in patients with advanced solid tumors
.
Mol Cancer Ther
2012
;
11
:
1820
8
.
24.
Du Bois
A
,
Vergote
I
,
Wimberger
P
,
Ray-Coquard
I
,
Harter
P
,
Curtis
LB
, et al
Open-label feasibility study of pazopanib, carboplatin, and paclitaxel in women with newly diagnosed, untreated, gynaecologic tumours: a phase I/II trial of the AGO study group
.
Br J Cancer
2012
;
106
:
629
32
.
25.
Tan
AR
,
Dowlati
A
,
Jones
SF
,
Infante
JR
,
Nishioka
J
,
Fang
L
, et al
Phase I study of pazopanib in combination with weekly paclitaxel in patients with advanced solid tumors
.
Oncologist
2010
;
15
:
1253
61
.
26.
Therasse
P
,
Arbuck
SG
,
Eisenhauer
EA
,
Wanders
J
,
Kaplan
RS
,
Rubinstein
L
, et al
New guidelines to evaluate the response to treatment in solid tumours
.
J Natl Cancer Inst
2000
;
92
:
205
16
.
27.
McKenney
JM
,
Swearingen
D
,
Di Spirito
M
,
Doyle
R
,
Pantaleon
C
,
Kling
D
, et al
Study of the pharmacokinetic interaction between simvastatin and prescription omega-3-acid ethyl esters
.
J Clin Pharmacol
2006
;
46
:
785
91
.
28.
Mross
K
,
Häring
B
,
Holländer
N
,
Mielke
S
,
Behringer
D
,
Massing
U
, et al
Comparison of 1-hour and 3-hours paclitaxel infusion pharmacokinetics: results from a randomized trial
.
Onkologie
2002
;
25
:
503
8
.
29.
Belani
CP
,
Ramalingam
S
,
Perry
MC
,
LaRocca
RV
,
Rinaldi
D
,
Gable
PS
, et al
Randomized, phase III study of weekly paclitaxel in combination with carboplatin versus standard every-3-weeks administration of carboplatin and paclitaxel for patients with previously untreated advanced non–small cell lung cancer
.
J Clin Oncol
2008
;
26
:
468
73
.
30.
Flaherty
KT
,
Lee
SJ
,
Zhao
F
,
Schuchter
LM
,
Flaherty
L
,
Kefford
R
, et al
Phase III trial of carboplatin and paclitaxel with or without sorafenib in metastatic melanoma
.
J Clin Oncol
2013
;
31
:
373
9
.
31.
Yamamoto
R
,
Kaneuchi
M
,
Nishiya
M
,
Todo
Y
,
Takeda
M
,
Okamoto
K
, et al
Clinical trial and pharmacokinetic study of combination paclitaxel and carboplatin in patients with epithelial ovarian cancer
.
Cancer Chemother Pharmacol
2002
;
50
:
137
42
.
32.
Fruehauf
JP
,
Alger
B
,
Parmakhtiar
B
,
Jakowatz
JG
,
Bettis
C
,
Chuang
T
, et al
A phase II single arm study of pazopanib and paclitaxel as first-line treatment for unresectable stage III and stage IV melanoma: interim analysis
.
J Clin Oncol
30
, 
2012
(
suppl 1; abstr 8524
).
33.
Goodman
V
,
Wang
Q
,
Pandite
L
,
Watkins
PB
. 
Incidence and management of hepatic toxicity in pazopanib-treated patients
.
Ann. Oncol
2010
;
21
Suppl 8
;
viii282
.
34.
Xu
CF
,
Reck
BH
,
Goodman
VL
,
Xue
Z
,
Huang
L
,
Barnes
MR
, et al
Association of the hemochromatosis gene with pazopanib-induced transaminase elevation in renal cell carcinoma
.
J Hepatol
2011
;
54
:
1237
43
.
35.
Xu
CF
,
Reck
BH
,
Xue
Z
,
Huang
L
,
Baker
KL
,
Chen
M
, et al
Pazopanib-induced hyperbilirubinemia is associated with Gilbert's syndrome UGT1A1 polymorphism
.
Br J Cancer
2010
;
102
:
1371
7
.
36.
Neuvonen
PJ
,
Backman
JT
,
Niemi
M
. 
Pharmacokinetic comparison of the potential over-the-counter statins simvastatin, lovastatin, fluvastatin and pravastatin
.
Clin Pharmacokinet
2008
;
47
:
463
74
.
37.
Xu
CF
,
Xue
Z
,
Bing
N
,
King
KS
,
McCann
LA
,
deSouza
PL
, et al
Concomitant use of pazopanib and simvastatin increases the risk of transaminase elevations in patients with cancer
.
Ann Oncol
2012
;
23
:
2470
1
.
38.
Legha
SS
,
Ring
S
,
Papadopoulos
N
,
Raber
M
,
Benjamin
RS
. 
A phase II trial of taxol in metastatic melanoma
.
Cancer
1990
;
65
:
2478
81
.
39.
Einzig
AI
,
Hochster
H
,
Wiernik
PH
,
Trump
DL
,
Dutcher
JP
,
Garowski
E
, et al
A phase II study of taxol in patients with malignant melanoma
.
Investig New Drugs
1991
;
9
:
59
64
.
40.
Bedikian
AY
,
Plager
C
,
Papadopoulos
N
,
Eton
O
,
Ellerhorst
J
,
Smith
T
. 
Phase II evaluation of paclitaxel by short intravenous infusion in metastatic melanoma
.
Melanoma Res
2004
;
14
:
63
6
.
41.
Hodi
FS
,
Soiffer
RJ
,
Clark
J
,
Finkelstein
DM
,
Haluska
FG
. 
Phase II study of paclitaxel and carboplatin for malignant melanoma
.
Am J Clin Oncol
2002
;
25
:
283
6
.
42.
Kim
KB
,
Sosman
JA
,
Fruehauf
JP
,
Linette
GP
,
Markovic
SN
,
McDermott
DF
, et al
BEAM: a randomized phase II study evaluating the activity of bevacizumab in combination with carboplatin plus paclitaxel in patients with previously untreated advanced melanoma
.
J Clin Oncol
2012
;
30
:
34
41
.
43.
Graells
J
,
Vinyals
A
,
Figueras
A
,
Llorens
A
,
Moreno
A
,
Marcoval
J
, et al
Overproduction of VEGF165 concomitantly expressed with its receptors promotes growth and survival of melanoma cells through MAPK and PI3K signaling
.
J Invest Dermatol
2004
;
123
:
1151
61
.
44.
Reck
M
,
Kaiser
R
,
Mellemgaard
A
,
Douillard
JY
,
Orlov
S
,
Krzakowski
M
, et al
Docetaxel plus nintedanib versus docetaxel plus placebo in patients with previously treated non–small cell lung cancer (LUME-Lung 1): a phase 3, double-blind, randomized controlled trial
.
Lancet Oncol
2014
;
15
:
143
55
.
45.
Salama
AKS
. 
Evolving pharmacotherapies for the treatment of metastatic melanoma
.
Clin Med Insights Oncol
2013
;
7
:
137
49
.
46.
Savas
P
,
Hughes
B
,
Solomon
B
. 
Targeted therapy in lung cancer: IPASS and beyond, keeping abreast of the explosion of targeted therapies for lung cancer
.
J Thorac Dis
2013
;
5
Suppl 5
:
S579
92
.