Pei He
Genentech
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Pei He.
The Lancet | 2016
Louis Fehrenbacher; Alexander I. Spira; Marcus Ballinger; Marcin Kowanetz; Johan Vansteenkiste; Julien Mazieres; Keunchil Park; David Smith; Angel Artal-Cortes; Conrad R. Lewanski; Fadi S. Braiteh; Daniel Waterkamp; Pei He; Wei Zou; Daniel S. Chen; Jing Yi; Alan Sandler; Achim Rittmeyer
BACKGROUND Outcomes are poor for patients with previously treated, advanced or metastatic non-small-cell lung cancer (NSCLC). The anti-programmed death ligand 1 (PD-L1) antibody atezolizumab is clinically active against cancer, including NSCLC, especially cancers expressing PD-L1 on tumour cells, tumour-infiltrating immune cells, or both. We assessed efficacy and safety of atezolizumab versus docetaxel in previously treated NSCLC, analysed by PD-L1 expression levels on tumour cells and tumour-infiltrating immune cells and in the intention-to-treat population. METHODS In this open-label, phase 2 randomised controlled trial, patients with NSCLC who progressed on post-platinum chemotherapy were recruited in 61 academic medical centres and community oncology practices across 13 countries in Europe and North America. Key inclusion criteria were Eastern Cooperative Oncology Group performance status 0 or 1, measurable disease by Response Evaluation Criteria In Solid Tumors version 1.1 (RECIST v1.1), and adequate haematological and end-organ function. Patients were stratified by PD-L1 tumour-infiltrating immune cell status, histology, and previous lines of therapy, and randomly assigned (1:1) by permuted block randomisation (with a block size of four) using an interactive voice or web system to receive intravenous atezolizumab 1200 mg or docetaxel 75 mg/m(2) once every 3 weeks. Baseline PD-L1 expression was scored by immunohistochemistry in tumour cells (as percentage of PD-L1-expressing tumour cells TC3≥50%, TC2≥5% and <50%, TC1≥1% and <5%, and TC0<1%) and tumour-infiltrating immune cells (as percentage of tumour area: IC3≥10%, IC2≥5% and <10%, IC1≥1% and <5%, and IC0<1%). The primary endpoint was overall survival in the intention-to-treat population and PD-L1 subgroups at 173 deaths. Biomarkers were assessed in an exploratory analysis. We assessed safety in all patients who received at least one dose of study drug. This study is registered with ClinicalTrials.gov, number NCT01903993. FINDINGS Patients were enrolled between Aug 5, 2013, and March 31, 2014. 144 patients were randomly allocated to the atezolizumab group, and 143 to the docetaxel group. 142 patients received at least one dose of atezolizumab and 135 received docetaxel. Overall survival in the intention-to-treat population was 12·6 months (95% CI 9·7-16·4) for atezolizumab versus 9·7 months (8·6-12·0) for docetaxel (hazard ratio [HR] 0·73 [95% CI 0·53-0·99]; p=0·04). Increasing improvement in overall survival was associated with increasing PD-L1 expression (TC3 or IC3 HR 0·49 [0·22-1·07; p=0·068], TC2/3 or IC2/3 HR 0·54 [0·33-0·89; p=0·014], TC1/2/3 or IC1/2/3 HR 0·59 [0·40-0·85; p=0·005], TC0 and IC0 HR 1·04 [0·62-1·75; p=0·871]). In our exploratory analysis, patients with pre-existing immunity, defined by high T-effector-interferon-γ-associated gene expression, had improved overall survival with atezolizumab. 11 (8%) patients in the atezolizumab group discontinued because of adverse events versus 30 (22%) patients in the docetaxel group. 16 (11%) patients in the atezolizumab group versus 52 (39%) patients in the docetaxel group had treatment-related grade 3-4 adverse events, and one (<1%) patient in the atezolizumab group versus three (2%) patients in the docetaxel group died from a treatment-related adverse event. INTERPRETATION Atezolizumab significantly improved survival compared with docetaxel in patients with previously treated NSCLC. Improvement correlated with PD-L1 immunohistochemistry expression on tumour cells and tumour-infiltrating immune cells, suggesting that PD-L1 expression is predictive for atezolizumab benefit. Atezolizumab was well tolerated, with a safety profile distinct from chemotherapy. FUNDING F Hoffmann-La Roche/Genentech Inc.
The Lancet | 2017
Achim Rittmeyer; Fabrice Barlesi; Daniel Waterkamp; Keunchil Park; Fortunato Ciardiello; Joachim von Pawel; Shirish M. Gadgeel; Toyoaki Hida; Dariusz M. Kowalski; Manuel Cobo Dols; Diego Cortinovis; Joseph Leach; Jonathan Polikoff; Carlos H. Barrios; Fairooz F. Kabbinavar; Osvaldo Arén Frontera; Filippo De Marinis; Hande Turna; Jongseok Lee; Marcus Ballinger; Marcin Kowanetz; Pei He; Daniel S. Chen; Alan Sandler; David R. Gandara
BACKGROUND Atezolizumab is a humanised antiprogrammed death-ligand 1 (PD-L1) monoclonal antibody that inhibits PD-L1 and programmed death-1 (PD-1) and PD-L1 and B7-1 interactions, reinvigorating anticancer immunity. We assessed its efficacy and safety versus docetaxel in previously treated patients with non-small-cell lung cancer. METHODS We did a randomised, open-label, phase 3 trial (OAK) in 194 academic or community oncology centres in 31 countries. We enrolled patients who had squamous or non-squamous non-small-cell lung cancer, were 18 years or older, had measurable disease per Response Evaluation Criteria in Solid Tumors, and had an Eastern Cooperative Oncology Group performance status of 0 or 1. Patients had received one to two previous cytotoxic chemotherapy regimens (one or more platinum based combination therapies) for stage IIIB or IV non-small-cell lung cancer. Patients with a history of autoimmune disease and those who had received previous treatments with docetaxel, CD137 agonists, anti-CTLA4, or therapies targeting the PD-L1 and PD-1 pathway were excluded. Patients were randomly assigned (1:1) to intravenously receive either atezolizumab 1200 mg or docetaxel 75 mg/m2 every 3 weeks by permuted block randomisation (block size of eight) via an interactive voice or web response system. Coprimary endpoints were overall survival in the intention-to-treat (ITT) and PD-L1-expression population TC1/2/3 or IC1/2/3 (≥1% PD-L1 on tumour cells or tumour-infiltrating immune cells). The primary efficacy analysis was done in the first 850 of 1225 enrolled patients. This study is registered with ClinicalTrials.gov, number NCT02008227. FINDINGS Between March 11, 2014, and April 29, 2015, 1225 patients were recruited. In the primary population, 425 patients were randomly assigned to receive atezolizumab and 425 patients were assigned to receive docetaxel. Overall survival was significantly longer with atezolizumab in the ITT and PD-L1-expression populations. In the ITT population, overall survival was improved with atezolizumab compared with docetaxel (median overall survival was 13·8 months [95% CI 11·8-15·7] vs 9·6 months [8·6-11·2]; hazard ratio [HR] 0·73 [95% CI 0·62-0·87], p=0·0003). Overall survival in the TC1/2/3 or IC1/2/3 population was improved with atezolizumab (n=241) compared with docetaxel (n=222; median overall survival was 15·7 months [95% CI 12·6-18·0] with atezolizumab vs 10·3 months [8·8-12·0] with docetaxel; HR 0·74 [95% CI 0·58-0·93]; p=0·0102). Patients in the PD-L1 low or undetectable subgroup (TC0 and IC0) also had improved survival with atezolizumab (median overall survival 12·6 months vs 8·9 months; HR 0·75 [95% CI 0·59-0·96]). Overall survival improvement was similar in patients with squamous (HR 0·73 [95% CI 0·54-0·98]; n=112 in the atezolizumab group and n=110 in the docetaxel group) or non-squamous (0·73 [0·60-0·89]; n=313 and n=315) histology. Fewer patients had treatment-related grade 3 or 4 adverse events with atezolizumab (90 [15%] of 609 patients) versus docetaxel (247 [43%] of 578 patients). One treatment-related death from a respiratory tract infection was reported in the docetaxel group. INTERPRETATION To our knowledge, OAK is the first randomised phase 3 study to report results of a PD-L1-targeted therapy, with atezolizumab treatment resulting in a clinically relevant improvement of overall survival versus docetaxel in previously treated non-small-cell lung cancer, regardless of PD-L1 expression or histology, with a favourable safety profile. FUNDING F. Hoffmann-La Roche Ltd, Genentech, Inc.
Journal of Thoracic Oncology | 2018
Louis Fehrenbacher; Joachim von Pawel; Keunchil Park; Achim Rittmeyer; David R. Gandara; Santiago Ponce Aix; Ji-Youn Han; Shirish M. Gadgeel; Toyoaki Hida; Diego Cortinovis; Manuel Cobo; Dariusz M. Kowalski; Filippo De Marinis; M. Gandhi; Bradford Danner; Christina Matheny; Marcin Kowanetz; Pei He; Federico Felizzi; Hina Patel; Alan Sandler; Marcus Ballinger; Fabrice Barlesi
Introduction: The efficacy and safety of atezolizumab versus the efficacy and safety of docetaxel as second‐ or third‐line treatment in patients with advanced NSCLC in the primary (n = 850) and secondary (n = 1225) efficacy populations of the randomized phase III OAK study (respectively referred to as the intention‐to‐treat [ITT] 850 [ITT850] and ITT1225) at an updated data cutoff were assessed. Methods: Patients received atezolizumab, 1200 mg, or docetaxel, 75 mg/m2, intravenously every 3 weeks until loss of clinical benefit or disease progression, respectively. The primary end point was overall survival (OS) in the ITT population and programmed death‐ligand 1–expressing subgroup. A sensitivity analysis was conducted to evaluate the impact of subsequent immunotherapy use in the docetaxel arm on the observed survival benefit with atezolizumab. Results: Atezolizumab demonstrated an OS benefit versus docetaxel in the updated ITT850 (hazard ratio [HR] = 0.75, 95% confidence interval: 0.64–0.89, p = 0.0006) and the ITT1225 (HR = 0.80, 95% confidence interval: 0.70–0.92, p = 0.0012) after minimum follow‐up times of 26 and 21 months, respectively. Improved survival with atezolizumab was observed across programmed death‐ligand 1 and histological subgroups. In the immunotherapy sensitivity analysis, the relative OS benefit with atezolizumab was slightly greater in the ITT850 (HR = 0.69) and ITT1225 (HR = 0.74) than the conventional OS estimate. Fewer patients receiving atezolizumab experienced grade 3 or 4 treatment‐related adverse events (14.9%) than did patients receiving docetaxel (42.4%); no grade 5 adverse events related to atezolizumab were observed. Conclusions: The results of the updated ITT850 and initial ITT1225 analyses were consistent with those of the primary efficacy analysis demonstrating survival benefit with atezolizumab versus with docetaxel. Atezolizumab continued to demonstrate a favorable safety profile after longer treatment exposure and follow‐up.
Journal of Thoracic Oncology | 2018
David R. Gandara; Joachim von Pawel; Julien Mazieres; Richard N. Sullivan; Åslaug Helland; Ji-Youn Han; Santiago Ponce Aix; Achim Rittmeyer; Fabrice Barlesi; Toshio Kubo; Keunchil Park; Jerome H. Goldschmidt; M. Gandhi; Cindy Yun; Wei Yu; Christina Matheny; Pei He; Alan Sandler; Marcus Ballinger; Louis Fehrenbacher
Introduction: Cancer immunotherapy may alter tumor biology such that treatment effects can extend beyond radiographic progression. In the randomized, phase III OAK study of atezolizumab (anti–programmed death‐ligand 1) versus docetaxel in advanced NSCLC, overall survival (OS) benefit with atezolizumab was observed in the overall patient population, without improvement in objective response rate (ORR) or progression‐free survival (PFS). We examine the benefit‐risk of atezolizumab treatment beyond progression (TBP). Methods: Eight hundred fifty patients included in the OAK primary efficacy analysis were evaluated. Atezolizumab was continued until loss of clinical benefit. Docetaxel was administered until Response Evaluation Criteria in Solid Tumors version 1.1 (RECIST v1.1) disease progression (PD)/unacceptable toxicity; no crossover to atezolizumab was allowed. ORR, PFS, post‐PD OS, target lesion change, and safety were evaluated. Results: In atezolizumab‐arm patients, ORR was 16% versus 14% and median PFS was 4.2 versus 2.8 months per immune‐modified RECIST versus RECIST v1.1. The median post‐PD OS was 12.7 months (95% confidence interval [CI]: 9.3–14.9) in 168 atezolizumab‐arm patients continuing TBP, 8.8 months (95% CI: 6.0–12.1) in 94 patients switching to nonprotocol therapy, and 2.2 months (95% CI: 1.9–3.4) in 70 patients receiving no further therapy. Of the atezolizumab TBP patients, 7% achieved a post‐progression response in target lesions and 49% had stable target lesions. Atezolizumab TBP was not associated with increased safety risks. Conclusions: Within the limitations of this retrospective analysis, the post‐PD efficacy and safety data from OAK are consistent with a positive benefit‐risk profile of atezolizumab TBP in patients performing well clinically at the time of PD.
Clinical Lung Cancer | 2018
Rodolfo Bordoni; Fortunato Ciardiello; Joachim von Pawel; Diego Cortinovis; Thomas Karagiannis; Marcus Ballinger; Alan Sandler; Wei Yu; Pei He; Christina Matheny; Federico Felizzi; Achim Rittmeyer
Micro‐Abstract: OAK, a phase III advanced non–small‐cell lung cancer (NSCLC) trial, compared the investigational drug atezolizumab with the standard chemotherapy agent docetaxel. The patient‐reported outcomes data revealed that atezolizumab prolonged the time to the worsening of disease‐related symptoms, such as chest pain, and maintained patients’ health‐related quality of life. These data further support the use of atezolizumab in the treatment of advanced NSCLC. Background: The randomized phase III OAK (a study of atezolizumab compared with docetaxel in participants with locally advanced or metastatic non–small‐cell lung cancer [NSCLC] who have failed platinum‐containing therapy) trial investigated the anti–programmed cell death ligand 1 (PD‐L1) antibody atezolizumab for advanced or metastatic, previously treated, NSCLC. Atezolizumab significantly improved overall survival (OS) compared with docetaxel (hazard ratio [HR], 0.73; 95% confidence interval [CI], 0.62–0.87; P = .0003; median OS, 13.8 vs. 9.6 months, respectively). Patient‐reported outcomes (PROs) were collected to evaluate disease‐related symptoms and health‐related quality of life (HRQoL) to support the finding of a survival benefit. Patients and Methods: The first 850 patients were randomized to receive atezolizumab (1200 mg every 3 weeks) or docetaxel (75 mg/m2 every 3 weeks). PROs were collected on day 1 of cycle 1, day 1 of every subsequent cycle, and at the end‐of‐treatment visit for patients who completed ≥ 1 baseline and 1 postbaseline PRO assessment. The European Organisation for the Research and Treatment of Cancer QoL questionnaire and lung cancer module were used to assess PROs. Results: Atezolizumab delayed the time to deterioration (TTD) in physical function (HR, 0.75; 95% CI, 0.58–0.98) and role function (HR, 0.79; 95% CI, 0.62–1.00) and numerically improved patients’ HRQoL from baseline compared with docetaxel. Atezolizumab also prolonged the TTD in chest pain (HR, 0.71; 95% CI, 0.49–1.05; P = .0823), although both arms showed an objective reduction relative to baseline. Overall, the patients had no clinically significant worsening in treatment‐related symptoms, although the scores favored atezolizumab. Conclusion: These PRO data support the clinical benefit of atezolizumab in patients with previously treated advanced or metastatic NSCLC. Atezolizumab prolonged the TTD of patients’ limitations in role and physical functions compared with docetaxel.
Clinical Cancer Research | 2018
Jin Y. Jin; Charles Ferté; Helen Winter; Sandhya Girish; Mark Stroh; Pei He; Marcus Ballinger; Alan Sandler; Amita Joshi; Achim Rittmeyer; David R. Gandara; Jean-Charles Soria; Rene Bruno
Purpose: Standard endpoints often poorly predict overall survival (OS) with immunotherapies. We investigated the predictive performance of model-based tumor growth inhibition (TGI) metrics using data from atezolizumab clinical trials in patients with non–small cell lung cancer. Patients and Methods: OS benefit with atezolizumab versus docetaxel was observed in both POPLAR (phase II) and OAK (phase III), although progression-free survival was similar between arms. A multivariate model linking baseline patient characteristics and on-treatment tumor growth rate constant (KG), estimated using time profiles of sum of longest diameters (RECIST 1.1) to OS, was developed using POPLAR data. The model was evaluated to predict OAK outcome based on estimated KG at TGI data cutoffs ranging from 10 to 122 weeks. Results: In POPLAR, TGI profiles in both arms crossed at 25 weeks, with more shrinkage with docetaxel and slower KG with atezolizumab. A log-normal OS model, with albumin and number of metastatic sites as independent prognostic factors and estimated KG, predicted OS HR in subpopulations of patients with varying baseline PD-L1 expression in both POPLAR and OAK: model-predicted OAK HR (95% prediction interval), 0.73 (0.63–0.85), versus 0.73 observed. The POPLAR OS model predicted greater than 97% chance of success of OAK (significant OS HR, P < 0.05) from the 40-week data cutoff onward with 50% of the total number of tumor assessments when a successful study was predicted from 70 weeks onward based on observed OS. Conclusions: KG has potential as a model-based early endpoint to inform decisions in cancer immunotherapy studies. Clin Cancer Res; 24(14); 3292–8. ©2018 AACR.
Annals of Oncology | 2016
Fabrice Barlesi; K. Park; Fortunato Ciardiello; J. von Pawel; Shirish M. Gadgeel; Toyoaki Hida; Dariusz M. Kowalski; Manuel Cobo Dols; Diego Cortinovis; Joseph Leach; Jonathan Polikoff; David R. Gandara; Carlos H. Barrios; Daniel S. Chen; Pei He; Marcin Kowanetz; Marcus Ballinger; Daniel Waterkamp; Alan Sandler; Achim Rittmeyer
Journal of Thoracic Oncology | 2017
Shirish M. Gadgeel; Fortunato Ciardiello; Achim Rittmeyer; Fabrice Barlesi; Diego Cortinovis; Carlos H. Barrios; Toyoaki Hida; Keunchil Park; Dariusz M. Kowalski; Manuel Cobo Dols; Joseph Leach; Jonathan Polikoff; Christina Matheny; Pei He; Marcin Kowanetz; Daniel Chen; Daniel Waterkamp; Marcus Ballinger; Alan Sandler; David R. Gandara; Joachim von Pawel
Journal of Clinical Oncology | 2016
David A. Smith; Johan Vansteenkiste; Louis Fehrenbacher; Keunchil Park; Julien Mazieres; Achim Rittmeyer; Angel Artal-Cortes; Conrad R. Lewanski; Fadi S. Braiteh; Jing Yi; Pei He; Wei Zou; Marcin Kowanetz; Daniel Waterkamp; Marcus Ballinger; Daniel S. Chen; Alan Sandler; Alexander I. Spira
Journal of Clinical Oncology | 2017
David R. Gandara; Joachim von Pawel; Richard N. Sullivan; Åslaug Helland; Ji-Youn Han; Santiago Ponce Aix; Achim Rittmeyer; Fabrice Barlesi; Toshio Kubo; Keunchil Park; Jerome H. Goldschmidt; M. Gandhi; Cindy Yun; Wei Yu; Christina Matheny; Pei He; Alan Sandler; Marcus Ballinger; Louis Fehrenbacher