Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Mark D. McKee.
Journal of Clinical Oncology | 2015
Calin Cainap; Shukui Qin; Wen-Tsung Huang; Ik Joo Chung; Hongming Pan; Ying Cheng; Masatoshi Kudo; Yoon-Koo Kang; Pei-Jer Chen; Han-Chong Toh; Vera Gorbunova; Ferry Eskens; Jiang Qian; Mark D. McKee; Justin L. Ricker; Dawn M. Carlson; Saied El-Nowiem
PURPOSE This open-label phase III trial evaluated efficacy and tolerability of linifanib versus sorafenib in patients with advanced hepatocellular carcinoma (HCC) without prior systemic therapy. PATIENTS AND METHODS Patients were randomly assigned in a 1:1 ratio to linifanib 17.5 mg once daily or sorafenib 400 mg twice daily. Patients were stratified by region (Outside Asia, Japan, and rest of Asia), Eastern Cooperative Oncology Group performance score (ECOG PS; 0 or 1), vascular invasion or extrahepatic spread (yes or no), and hepatitis B virus (HBV) infection (yes or no). The primary end point of the study was overall survival (OS). Secondary end points were time to progression (TTP) and objective response rate (ORR) per RECIST v1.1. RESULTS We randomly assigned 1,035 patients (median age, 60 years; Asian, 66.6%; ECOG PS 0, 65.2%; HBV, 49.1%; vascular invasion or extrahepatic spread, 70.1%). Median OS was 9.1 months on the linifanib arm (95% CI, 8.1 to 10.2) and 9.8 months on the sorafenib arm (95% CI, 8.3 to 11.0; hazard ratio [HR], 1.046; 95% CI, 0.896 to 1.221). For prespecified stratification subgroups, OS HRs ranged from 0.793 to 1.119 and the 95% CI contained 1.0. Median TTP was 5.4 months on the linifanib arm (95% CI, 4.2 to 5.6) and 4.0 months on the sorafenib arm (95% CI, 2.8 to 4.2; HR, 0.759; 95% CI, 0.643 to 0.895; P = .001). Best response rate was 13.0% on the linifanib arm versus 6.9% on the sorafenib arm. Grade 3/4 adverse events (AEs); serious AEs; and AEs leading to discontinuation, dose interruption, and reduction were more frequent with linifanib (all P < .001). CONCLUSION Linifanib and sorafenib had similar OS in advanced HCC. Predefined superiority and noninferiority OS boundaries were not met for linifanib and the study failed to meet the primary end point. TTP and ORR favored linifanib; safety results favored sorafenib.
Journal of Clinical Oncology | 2015
Suresh S. Ramalingam; Mikhail Shtivelband; Ross A. Soo; Carlos H. Barrios; A. Makhson; José Getúlio Martins Segalla; Kenneth B. Pittman; Petr Kolman; Jose R. Pereira; Gordan Srkalovic; Chandra P. Belani; Rita Axelrod; Taofeek K. Owonikoko; Qin Qin; Jiang Qian; Evelyn McKeegan; Viswanath Devanarayan; Mark D. McKee; Justin L. Ricker; Dawn M. Carlson; Vera Gorbunova
PURPOSE Linifanib, a potent, selective inhibitor of vascular endothelial growth factor (VEGF) and platelet-derived growth factor (PDGF) receptors, has single-agent activity in non-small-cell lung cancer (NSCLC). We evaluated linifanib with carboplatin and paclitaxel as first-line therapy of advanced nonsquamous NSCLC. PATIENTS AND METHODS Patients with stage IIIB/IV nonsquamous NSCLC were randomly assigned to 3-week cycles of carboplatin (area under the curve 6) and paclitaxel (200 mg/m(2)) with daily placebo (arm A), linifanib 7.5 mg (arm B), or linifanib 12.5 mg (arm C). The primary end point was progression-free survival (PFS); secondary efficacy end points included overall survival (OS) and objective response rate. RESULTS One hundred thirty-eight patients were randomly assigned (median age, 61 years; 57% men; 84% smokers). Median PFS times were 5.4 months (95% CI, 4.2 to 5.7 months) in arm A (n = 47), 8.3 months (95% CI, 4.2 to 10.8 months) in arm B (n = 44), and 7.3 months (95% CI, 4.6 to 10.8 months) in arm C (n = 47). Hazard ratios (HRs) for PFS were 0.51 for arm B versus A (P = .022) and 0.64 for arm C versus A (P = .118). Median OS times were 11.3, 11.4, and 13.0 months in arms A, B, and C, respectively. HRs for OS were 1.08 for arm B versus A (P = .779) and 0.88 for arm C versus A (P = .650). Both linifanib doses were associated with increased toxicity, including a higher incidence of adverse events known to be associated with VEGF/PDGF inhibition. Baseline plasma carcinoembryonic antigen/cytokeratin 19 fragments biomarker signature was associated with PFS improvement and a trend toward OS improvement with linifanib 12.5 mg. CONCLUSION Addition of linifanib to chemotherapy significantly improved PFS (arm B), with a modest trend for survival benefit (arm C) and increased toxicity reflective of known VEGF/PDGF inhibitory effects.
Lancet Oncology | 2018
Sibylle Loibl; Joyce O'Shaughnessy; Michael Untch; William M. Sikov; Hope S. Rugo; Mark D. McKee; Jens Huober; Mehra Golshan; Gunter von Minckwitz; David Maag; Danielle Marie Sullivan; Norman Wolmark; Kristi McIntyre; Jose J Ponce Lorenzo; Otto Metzger Filho; Priya Rastogi; W. Fraser Symmans; Xuan Liu; Charles E. Geyer
BACKGROUND Although several randomised trials in patients with triple-negative breast cancer have shown that the addition of carboplatin, with or without poly(ADP-ribose) polymerase (PARP) inhibitors, to neoadjuvant chemotherapy increases the likelihood of achieving a pathological complete response, the use of these therapies in this setting has remained controversial. The BrighTNess trial was designed to assess the addition of the PARP inhibitor veliparib plus carboplatin or carboplatin alone to standard neoadjuvant chemotherapy in triple-negative breast cancer. METHODS We did a phase 3, randomised, double-blind, placebo-controlled trial (BrighTNess) across 145 sites in 15 countries. Patients aged 18 years and older with previously untreated histologically or cytologically confirmed clinical stage II-III triple-negative breast cancer, who were candidates for potentially curative surgery and had an Eastern Cooperative Oncology Group performance status of 0 or 1, were randomly assigned (2:1:1) by an interactive response technology system via permuted blocks (block size of four) within strata to receive one of three segment 1 regimens: paclitaxel (80 mg/m2 intravenously weekly for 12 doses) plus carboplatin (area under the curve 6 mg/mL per min, intravenously every 3 weeks, for four cycles) plus veliparib (50 mg orally, twice a day); paclitaxel plus carboplatin plus veliparib placebo (twice a day); or paclitaxel plus carboplatin placebo (every 3 weeks for four cycles) plus veliparib placebo. Following segment 1, all patients were assigned to segment 2 in which they received doxorubicin and cyclophosphamide every 2-3 weeks for four cycles. Randomisation for segment 1 was stratified by germline BRCA mutation status, nodal stage, and planned schedule of doxorubicin and cyclophosphamide administration. The primary endpoint was pathological complete response in breast and lymph nodes as determined by site pathologists following completion of neoadjuvant therapy. Efficacy analyses were done by intention to treat and safety analyses included all patients who received at least one dose of study treatment. These are the first results of an ongoing clinical trial; the data cutoff for the analyses presented was Dec 8, 2016. This study is registered with ClinicalTrials.gov, number NCT02032277. FINDINGS Between April 4, 2014, and March 18, 2016, 634 patients were randomly assigned: 316 to paclitaxel plus carboplatin plus veliparib, 160 to paclitaxel plus carboplatin, and 158 to paclitaxel alone. The proportion of patients who achieved a pathological complete response was higher in the paclitaxel, carboplatin, and veliparib group than in patients receiving paclitaxel alone (168 [53%] of 316 patients vs 49 [31%] of 158, p<0·0001), but not compared with patients receiving paclitaxel plus carboplatin (92 [58%] of 160 patients, p=0·36). Grade 3 or 4 toxicities, and serious adverse events were more common in patients receiving carboplatin, whereas veliparib did not substantially increase toxicity. The most common grade 3 or 4 events overall were neutropenia (352 [56%] of 628 patients), anaemia (180 [29%]), and thrombocytopenia (75 [12%]) through complete treatment, and febrile neutropenia (88 [15%] of 601 patients) during segment 2. The most common serious adverse events were febrile neutropenia (80 [13%] of 628 patients) and anaemia (20 [3%]). INTERPRETATION Although the addition of veliparib and carboplatin to paclitaxel followed by doxorubicin and cyclophosphamide improved the proportion of patients with triple-negative breast cancer who achieved a pathological complete response, the addition of veliparib to carboplatin and paclitaxel did not. Increased toxicities with the addition of carboplatin (with or without veliparib) to paclitaxel were manageable and did not substantially affect treatment delivery of paclitaxel followed by doxorubicin and cyclophosphamide. Given the consistent results with previous studies, the addition of carboplatin appears to have a favourable risk to benefit profile and might be considered as a potential component of neoadjuvant chemotherapy for patients with high-risk, triple-negative breast cancer. FUNDING AbbVie.
Clinical Cancer Research | 2017
Suresh S. Ramalingam; Normand Blais; Julien Mazieres; Martin Reck; C. Michael Jones; Erzsébet Juhász; Laszlo Urban; Sergey Orlov; Fabrice Barlesi; Ebenezer A. Kio; Ulrich Keiholz; Qin Qin; Jiang Qian; Caroline Nickner; Juliann Dziubinski; Hao Xiong; Peter Ansell; Mark D. McKee; Vincent L. Giranda; Vera Gorbunova
Purpose: PARP plays an important role in DNA repair. Veliparib, a PARP inhibitor, enhances the efficacy of platinum compounds and has been safely combined with carboplatin and paclitaxel. The primary endpoint of this phase II trial determined whether addition of veliparib to carboplatin and paclitaxel improved progression-free survival (PFS) in previously untreated patients with advanced/metastatic non–small cell lung cancer. Experimental Design: Patients were randomized 2:1 to carboplatin and paclitaxel with either veliparib or placebo. Veliparib (120 mg) or placebo was given on days 1 to 7 of each 3-week cycle, with carboplatin (AUC = 6 mg/mL/min) and paclitaxel (200 mg/m2) administered on day 3, for a maximum of 6 cycles. Results: Overall, 158 were included (median age, 63 years; male 68%, squamous histology 48%). Median PFS was 5.8 months in the veliparib group versus 4.2 months in the placebo group [HR, 0.72; 95% confidence interval (CI), 0.45–1.15; P = 0.17)]. Median overall survival (OS) was 11.7 and 9.1 months in the veliparib and placebo groups, respectively (HR, 0.80; 95% CI, 0.54–1.18; P = 0.27). In patients with squamous histology, median PFS (HR, 0.54; 95% CI, 0.26–1.12; P = 0.098) and OS (HR, 0.73; 95% CI, 0.43–1.24; P = 0.24) favored veliparib treatment. Objective response rate was similar between groups (veliparib: 32.4%; placebo: 32.1%), but duration of response favored veliparib treatment (HR, 0.47; 95% CI, 0.16–1.42; P = 0.18). Grade III/IV neutropenia, thrombocytopenia, and anemia were comparable between groups. Conclusions: Veliparib combination with carboplatin and paclitaxel was well-tolerated and demonstrated a favorable trend in PFS and OS versus chemotherapy alone. Patients with squamous histology had the best outcomes with veliparib combination. Clin Cancer Res; 23(8); 1937–44. ©2016 AACR.
Gynecologic Oncology | 2018
Heidi J. Gray; Katherine M. Bell-McGuinn; Gini F. Fleming; Mihaela C. Cristea; Hao Xiong; Danielle Marie Sullivan; Yan Luo; Mark D. McKee; Wijith Munasinghe; Lainie P. Martin
OBJECTIVE Determine the maximum tolerated dose (MTD) and recommended phase II dose (RP2D) of veliparib combined with carboplatin and gemcitabine in patients with advanced ovarian cancer and other nonhematologic malignancies. METHODS In this phase I study, patients with metastatic or unresectable solid tumors and ≤2 prior chemotherapy regimens received veliparib combined with carboplatin area under the curve (AUC) 4 on day 1 and gemcitabine 800mg/m2 on days 1 and 8 of a 21-day cycle for maximum 10cycles, followed by optional veliparib maintenance therapy. Veliparib dosing commenced twice-daily (BID) continuously on day 1 of cycle 2; granulocyte colony-stimulating factor was permitted. Dose escalation used a Bayesian continual reassessment method. Safety, tolerability, and efficacy were evaluated. RESULTS Seventy-five patients were enrolled (ovarian cancer, n=54; breast cancer, n=12). Thirty-six patients with ovarian cancer (67%) had known germline BRCA mutations. Most common treatment-related adverse events (TRAEs; ≥60%) were thrombocytopenia, neutropenia, nausea, and anemia. Most common grade 3/4 TRAEs (≥40%) were neutropenia and thrombocytopenia. Dose-limiting toxicities were thrombocytopenia and neutropenia. The MTD/RP2D was established at veliparib 250mg with carboplatin AUC 4 plus gemcitabine 800mg/m2. Responses were observed in 69% of patients with BRCA-deficient ovarian cancer (45% partial, 24% complete responses). Five patients remained on veliparib (80-310mg BID) for >34cycles. CONCLUSIONS Veliparib plus carboplatin/gemcitabine is tolerated, with a safety profile similar to carboplatin and gemcitabine alone. Combination therapy demonstrated promising preliminary antitumor activity in platinum-sensitive ovarian cancer patients with germline BRCA mutations. Trial registration ID: NCT01063816.
Lung Cancer | 2015
Evelyn McKeegan; Peter Ansell; Gerard Davis; Sabrina S. Chan; Raj Chandran; Susan Gawel; Barry L. Dowell; Anahita Bhathena; Arunava Chakravartty; Mark D. McKee; Justin L. Ricker; Dawn M. Carlson; Suresh S. Ramalingam; Viswanath Devanarayan
OBJECTIVES Linifanib, a potent and selective inhibitor of the tyrosine kinase activity of vascular endothelial growth factor and platelet-derived growth factor receptors, has clinical activity in advanced non-small cell lung cancer (NSCLC) both as monotherapy in the relapsed setting or with carboplatin and paclitaxel in the first-line setting. Though benefit was observed in unselected patient populations, identification of predictive biomarkers is critical for further development of this novel agent. MATERIALS AND METHODS Data from 4 randomized studies in relapsed NSCLC with linifanib (n=116) or other treatments (n=125) were examined in an exploratory analysis to identify a biomarker profile predictive of favorable survival. RESULTS A signature combining the established tumor markers carcinoembryonic antigen (CEA) and cytokeratin 19 fragments (CYFRA 21-1) was predictive of a favorable outcome. This signature was associated with improved survival in patients receiving linifanib monotherapy (hazard ratio [HR]=0.51 vs signature negative; p=0.002), but not in those receiving other anti-cancer treatments (p=0.716). This signature was validated on baseline plasma samples from patients enrolled in a randomized trial of daily linifanib 7.5 mg, linifanib 12.5 mg, or placebo added to first-line carboplatin and paclitaxel chemotherapy for advanced, nonsquamous NSCLC. Only linifanib-treated signature-positive patients had significant improvement in progression-free survival (PFS). Median PFS with placebo was 5.2 months versus 10.2 months (HR=0.49, p=0.049) for those receiving linifanib 7.5mg, and 8.3 months (HR=0.38, p=0.029) for linifanib 12.5 mg. Overall survival for signature-positive patients was 11.3 months with placebo, 12.5 months with linifanib 7.5mg (HR=1.02, p=0.758), and 17.4 months with linifanib 12.5 mg (HR=0.54, p=0.137). CONCLUSION This baseline plasma biomarker signature is associated with improved outcome in advanced NSCLC patients receiving linifanib. Utility of the biomarker signature in patient selection for linifanib therapy in NSCLC merits evaluation in larger, prospective trials that are powered to detect a survival benefit.
Journal of Thoracic Oncology | 2017
Martin Reck; Normand Blais; Erzsébet Juhász; Vera Gorbunova; C. Michael Jones; Laszlo Urban; Sergey Orlov; Fabrice Barlesi; Ebenezer A. Kio; Ulrich Keilholz; Qin Qin; Jiang Qian; Caroline Nickner; Juliann Dziubinski; Hao Xiong; Rajendar K. Mittapalli; Martin Dunbar; Peter Ansell; Lei He; Mark D. McKee; Vincent L. Giranda; Suresh S. Ramalingam
Introduction: Tobacco‐related NSCLC is associated with reduced survival and greater genomic instability. Veliparib, a potent poly(adenosine diphosphate–ribose) polymerase inhibitor, augments platinum‐induced DNA damage. A phase 2 trial of untreated advanced NSCLC showed a trend for improved outcomes (hazard ratio [HR] = 0.80, 95% confidence interval: 0.54–1.18, p = 0.27 for overall survival and HR = 0.72, 95% CI: 0.45–1.15, p = 0.17 for progression‐free survival) when veliparib was added to carboplatin/paclitaxel. Here we report an exploratory analysis by smoking history. Methods: Patients were randomized 2:1 to receive carboplatin/paclitaxel with veliparib, 120 mg (n = 105), or placebo (n = 53). Patients were stratified by histologic subtype and smoking history (recent smokers [n = 95], former smokers [n = 42], and never‐smokers [n = 21]). Plasma cotinine level was measured as a chemical index of smoking. Mutation status was assessed by whole exome sequencing (n = 38). Results: Smoking history, histologic subtype, age, Eastern Cooperative Oncology Group performance status, sex, and geographic region predicted veliparib benefit in univariate analyses. In multivariate analysis, history of recent smoking was most predictive for veliparib benefit. Recent smokers treated with veliparib derived significantly greater progression‐free survival and overall survival benefits (HR = 0.38 [p < 0.01] and HR = 0.43 [p < 0.01]) than former smokers (HR = 2.098 [p = 0 0208] and HR = 1.62 [p = 0.236]) and never‐smokers (HR = 1.025 [p = 0.971] and HR = 1.33 [p = 0.638]). Sequencing data revealed that mutational burden was not associated with veliparib benefit. The rate of grade 3 or 4 adverse events was higher in recent smokers with veliparib treatment; all‐grade and serious adverse events were similar in both treatment arms. Conclusions: Smoking history predicted for efficacy with a veliparib‐chemotherapy combination; toxicity was acceptable regardless of smoking history. A prespecified analysis of recent smokers is planned for ongoing phase 3 studies of veliparib in NSCLC.
British Journal of Cancer | 2018
Michael L. Maitland; Sarina Anne Piha-Paul; Gerald S. Falchook; Razelle Kurzrock; Ly M. Nguyen; Linda Janisch; Sanja Karovic; Mark D. McKee; Elizabeth Hoening; Shekman Wong; Wijith Munasinghe; Joann P. Palma; Cherrie K. Donawho; Guinan K. Lian; Peter Ansell; Mark J. Ratain; David S. Hong
BackgroundIlorasertib (ABT-348) inhibits Aurora and VEGF receptor (VEGFR) kinases. Patients with advanced solid tumours participated in a phase 1 dose-escalation trial to profile the safety, tolerability, and pharmacokinetics of ilorasertib.MethodsIlorasertib monotherapy was administered at 10–180 mg orally once daily (Arm I, n = 23), 40–340 mg orally twice daily (Arm II, n = 28), or 8–32 mg intravenously once daily (Arm III, n = 7), on days 1, 8, and 15 of each 28-day cycle.ResultsDose-limiting toxicities were predominantly related to VEGFR inhibition. The most frequent treatment-emergent adverse events ( > 30%) were: fatigue (48%), anorexia (34%), and hypertension (34%). Pharmacodynamic markers suggested that ilorasertib engaged VEGFR2 and Aurora B kinase, with the VEGFR2 effects reached at lower doses and exposures than Aurora inhibition effects. In Arm II, one basal cell carcinoma patient (40 mg twice daily (BID)) and one patient with adenocarcinoma of unknown primary site (230 mg BID) had partial responses.ConclusionsIn patients with advanced solid tumours, ilorasertib treatment resulted in evidence of engagement of the intended targets and antitumour activity, but with maximum inhibition of VEGFR family kinases occurring at lower exposures than typically required for inhibition of Aurora B in tissue.Clinical Trial Registration: NCT01110486
Cancer Medicine | 2018
Theresa L. Werner; Jasgit C. Sachdev; Elizabeth M. Swisher; Martin Gutierrez; Muaiad Kittaneh; Mark N. Stein; Hao Xiong; Martin Dunbar; Danielle Marie Sullivan; Philip Komarnitsky; Mark D. McKee; Antoinette R. Tan
The poly(ADP‐ribose) polymerase‐1/2 inhibitor veliparib is active against tumors deficient in homologous DNA damage repair. The pharmacokinetics and safety of veliparib extended‐release (ER) were evaluated in patients with advanced solid tumors. This phase I study assessed veliparib‐ER up to 800 mg once daily or 600 mg twice daily. Dose‐limiting toxicities (DLTs), recommended phase II dose (RP2D), and maximum tolerated dose (MTD) were assessed in cycle 1 and safety/tolerability during continuous administration (28‐day cycles). Seventy‐one patients (n = 53 ovarian, n = 17 breast, n = 1 prostate carcinoma) received veliparib; 50 had deleterious breast cancer susceptibility (BRCA) gene mutations. Single‐dose veliparib‐ER 200 mg (fasting) led to 58% lower peak concentration and similar area under the concentration‐time curve compared with veliparib immediate‐release (IR). Three patients experienced DLTs (grade 2: asthenia; grade 3: nausea/vomiting, seizure). RP2D and MTD for veliparib‐ER were 400 mg BID. The most frequent adverse events (AEs) were nausea (78.9%) and vomiting (50.7%). The most common grade 3/4 treatment‐related AEs were as follows: thrombocytopenia (7.0%), nausea, and anemia (4.2% each). Overall, 12 (27.3%) patients with ovarian and 10 (62.5%) patients with breast carcinoma had a partial response. Veliparib‐ER, versus veliparib‐IR, exhibited an improved pharmacokinetic profile and was well tolerated in patients with ovarian and BRCA‐mutated breast cancers.
Journal of Clinical Oncology | 2015
Calin Cainap; Mark D. McKee; Justin L. Ricker
We appreciate the opportunity to respond to the insightful comments from Bouattour et al regarding our recent report of linifanib versus sorafenib in advanced hepatocellular carcinoma (HCC). In this trial, linifanib treatment resulted in favorable time to progression, progression-free survival, and objective response rate compared with sorafenib treatment, but led to similar overall survival. Bouattour et al suggest that overall survival was not improved due to an increased rate of adverse events associated with high linifanib exposures at the 17.5-mg daily dose of linifanib. The 17.5-mg daily dose for patients with advanced HCC was chosen based on an exposure-response analysis which provided strong rationale for flat rather than weight-based linifanib dosing. Analysis of data from 266 trial patients receiving linifanib predicted adverse event rates to be similar with weight-based and fixed dosing, and predicted adverse event rates were within a tighter range for subjects of varied weights based on fixed dosing. Additionally, data from prior trials supported the use of the same dose for patients with HCC as for patients without hepatic dysfunction, because linifanib exposures in a phase II trial of patients with HCC were not increased compared with patients in other trials with other cancer types. Of note, the median weight of patients in the phase III trial of linifanib versus sorafenib for advanced HCC was 66 kg. In the median patient, 17.5 mg corresponded to a dose of 0.265 mg/kg, which is close to the recommended weight-based dose of 0.25 mg/kg determined in phase I. When flat dosing is used, it is reasonable to assume that patients with the lowest weight may be at risk for adverse events leading to early dose reductions or discontinuation, and thus be at risk for reduced efficacy. To look for evidence of this phenomenon in the phase III trial of linifanib versus sorafenib for advanced HCC, we analyzed tolerability by weight quartiles. Compared with patients in the highest weight quartile (weight 76 kg), patients in the lowest weight quartile (weight 59 kg) had similar rates of linifanib discontinuation due to adverse events (26.5 v 26.4%), and lower rates of early dose reduction (19.9 v 30.4%). Thus, we do not find evidence of a link between low weight and decreased tolerability of linifanib. Though prior studies suggested linifanib exposures were not affected by hepatic dysfunction, increased rates of grade 3/4 ascites and encephalopathy in patients who received linifanib were noted. The risk-benefit balance for patients with hepatic dysfunction was carefully considered in this trial, and toxicity management guidelines included linifanib dose modifications for patients with hepatic function changes during therapy. Nevertheless, we wondered whether linifanib treatment may have led to worse outcomes in patients with poor hepatic reserve. A posthoc analysis of the trial was performed, examining survival in among the approximately 5% of patients who entered treatment with Child-Pugh class B cirrhosis. The somewhat unexpected finding was that the median overall survival in ChildPugh class B cirrhotics was longer for patients receiving linifanib versus sorafenib (121 v 77 days), though the difference was not statistically significant (hazard ratio, 0.732; P .30). In summary, we appreciate that the dosing of antiangiogenic therapies has been an area of extensive discussion, and concern over toxicity of other antiangiogenic agents in patients with HCC was a principle motivation for the detailed analysis of linifanib dose-response that led to flat dosing. We believe it is not clear whether another trial using a different dose of linifanib for the treatment of advanced HCC would result in improved outcomes. However, data from this trial suggest that weight-based dosing or exclusion of patients with the worst hepatic function would not have led to a better result.