Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Benjamin C. Creelan is active.

Publication


Featured researches published by Benjamin C. Creelan.


The New England Journal of Medicine | 2017

First-Line Nivolumab in Stage IV or Recurrent Non–Small-Cell Lung Cancer

David P. Carbone; Martin Reck; Luis Paz-Ares; Benjamin C. Creelan; Leora Horn; Martin Steins; Enriqueta Felip; Michel M. van den Heuvel; Tudor-Eliade Ciuleanu; Firas Benyamine Badin; Neal Ready; T. Jeroen N. Hiltermann; Suresh R. Nair; Rosalyn A. Juergens; Solange Peters; Elisa Minenza; John Wrangle; Delvys Rodriguez-Abreu; Hossein Borghaei; George R. Blumenschein; Liza C. Villaruz; Libor Havel; J. Krejčí; Jesus Corral Jaime; Han Chang; William J. Geese; Prabhu Bhagavatheeswaran; Allen C. Chen; Mark A. Socinski

BACKGROUND Nivolumab has been associated with longer overall survival than docetaxel among patients with previously treated non–small‐cell lung cancer (NSCLC). In an open‐label phase 3 trial, we compared first‐line nivolumab with chemotherapy in patients with programmed death ligand 1 (PD‐L1)–positive NSCLC. METHODS We randomly assigned, in a 1:1 ratio, patients with untreated stage IV or recurrent NSCLC and a PD‐L1 tumor‐expression level of 1% or more to receive nivolumab (administered intravenously at a dose of 3 mg per kilogram of body weight once every 2 weeks) or platinum‐based chemotherapy (administered once every 3 weeks for up to six cycles). Patients receiving chemotherapy could cross over to receive nivolumab at the time of disease progression. The primary end point was progression‐free survival, as assessed by means of blinded independent central review, among patients with a PD‐L1 expression level of 5% or more. RESULTS Among the 423 patients with a PD‐L1 expression level of 5% or more, the median progression‐free survival was 4.2 months with nivolumab versus 5.9 months with chemotherapy (hazard ratio for disease progression or death, 1.15; 95% confidence interval [CI], 0.91 to 1.45; P=0.25), and the median overall survival was 14.4 months versus 13.2 months (hazard ratio for death, 1.02; 95% CI, 0.80 to 1.30). A total of 128 of 212 patients (60%) in the chemotherapy group received nivolumab as subsequent therapy. Treatment‐related adverse events of any grade occurred in 71% of the patients who received nivolumab and in 92% of those who received chemotherapy. Treatment‐related adverse events of grade 3 or 4 occurred in 18% of the patients who received nivolumab and in 51% of those who received chemotherapy. CONCLUSIONS Nivolumab was not associated with significantly longer progression‐free survival than chemotherapy among patients with previously untreated stage IV or recurrent NSCLC with a PD‐L1 expression level of 5% or more. Overall survival was similar between groups. Nivolumab had a favorable safety profile, as compared with chemotherapy, with no new or unexpected safety signals. (Funded by Bristol‐Myers Squibb and others; CheckMate 026 ClinicalTrials.gov number, NCT02041533.)


Cancer Control | 2014

Update on immune checkpoint inhibitors in lung cancer.

Benjamin C. Creelan

BACKGROUND The immune checkpoint proteins, including the B7/CD28 receptor superfamily, have become increasingly important targets for pharmacologic blockade. Several classes of new agents have impressive clinical activity, and their eventual approval for treatment of lung cancer seems likely. METHODS This article discusses the current development of these agents, including the CTLA-4, PD-1, and PD-L1 inhibitory pathways, killer immunoglobulin receptor (KIR ) inhibition, and other checkpoint proteins. RESULTS Ipilimumab in combination with chemotherapy has exhibited encouraging results in small-cell and non-small-cell lung cancer alike. Reported phase I trials of the monoclonal antibodies nivolumab, MK-3475, MEDI4736, and MPDL3280A are demonstrating durable overall radiological response rates in the 20% to 25% range in lung cancer. This exceptional activity includes squamous lung cancers, a population historically bereft of significant therapeutic advances. Retrospective examination of tumor PD-L1 expression suggests that PD-L1 may eventually be evaluable as a predictive biomarker. Dual checkpoint blockade strategies, such as those combining anti-CTLA-4, anti-LAG-3, or anti-KIR, are being tested to increase the proportion and durability of tumor responses. Examination of acquired immune resistance and post-immunotherapy relapse strategies are underway. CONCLUSIONS These emerging antibodies hold great potential for the systemic control of epithelial cancers such as lung cancer.


Oncogene | 2016

Differential association of STK11 and TP53 with KRAS mutation-associated gene expression, proliferation and immune surveillance in lung adenocarcinoma.

Matthew B. Schabath; Eric A. Welsh; William J. Fulp; Lu Chen; Jamie K. Teer; Zachary Thompson; Brienne E. Engel; Xie M; Anders Berglund; Benjamin C. Creelan; Scott Antonia; Jhanelle E. Gray; Steven Eschrich; Dung-Tsa Chen; W D Cress; Eric B. Haura; Amer A. Beg

While mutations in the KRAS oncogene are among the most prevalent in human cancer, there are few successful treatments to target these tumors. It is also likely that heterogeneity in KRAS-mutant tumor biology significantly contributes to the response to therapy. We hypothesized that the presence of commonly co-occurring mutations in STK11 and TP53 tumor suppressors may represent a significant source of heterogeneity in KRAS-mutant tumors. To address this, we utilized a large cohort of resected tumors from 442 lung adenocarcinoma patients with data including annotation of prevalent driver mutations (KRAS and EGFR) and tumor suppressor mutations (STK11 and TP53), microarray-based gene expression and clinical covariates, including overall survival (OS). Specifically, we determined impact of STK11 and TP53 mutations on a new KRAS mutation-associated gene expression signature as well as previously defined signatures of tumor cell proliferation and immune surveillance responses. Interestingly, STK11, but not TP53 mutations, were associated with highly elevated expression of KRAS mutation-associated genes. Mutations in TP53 and STK11 also impacted tumor biology regardless of KRAS status, with TP53 strongly associated with enhanced proliferation and STK11 with suppression of immune surveillance. These findings illustrate the remarkably distinct ways through which tumor suppressor mutations may contribute to heterogeneity in KRAS-mutant tumor biology. In addition, these studies point to novel associations between gene mutations and immune surveillance that could impact the response to immunotherapy.


Cancer Research | 2017

Abstract CT082: Impact of tumor mutation burden on the efficacy of first-line nivolumab in stage iv or recurrent non-small cell lung cancer: An exploratory analysis of CheckMate 026

Solange Peters; Benjamin C. Creelan; Matthew D. Hellmann; Mark A. Socinski; Martin Reck; Prabhu Bhagavatheeswaran; Han Chang; William J. Geese; Luis Paz-Ares; David P. Carbone

Background: CheckMate 026 is a randomized trial of nivolumab monotherapy versus platinum doublet chemotherapy in patients with untreated stage IV or recurrent non-small cell lung cancer (NSCLC) and ≥1% programmed death-1 ligand 1 (PD-L1) tumor expression. Nivolumab was not associated with improved progression-free survival (PFS) versus chemotherapy in patients with ≥5% PD-L1 expression, however, nivolumab had a favorable safety profile compared with chemotherapy and overall survival (OS) was similar between treatment arms. In an exploratory analysis, we assessed whether patients with high tumor mutation burden (TMB) may derive enhanced benefit from nivolumab compared with platinum doublet chemotherapy. Methods: TMB scores for missense, somatic mutations were determined in patients with sufficient samples for whole exome sequencing of tumor and matched whole blood DNA. For the initial analyses, patients were equally divided into groups based on TMB tertile distribution. Patients in the low, medium, and high TMB tertiles had 0-99, 100-242, and ≥243 mutations, respectively. Results: Of 541 randomized patients, 312 (57.7%) had evaluable data to determine TMB. The percentage of patients with high TMB was lower in the nivolumab arm than in the chemotherapy arm (29.7% vs 39.0%). Baseline characteristics, PFS, and OS in patients evaluable for TMB were similar to all randomized patients. In patients with high TMB, PFS was improved (median PFS of 9.7 vs. 5.8 months; HR, 0.62; 95% CI, 0.38 to 1.00) and objective response rate was higher with nivolumab versus chemotherapy (46.8% vs 28.3%). OS in patients with high TMB was similar between the 2 treatment arms, which may be attributable in part to a 65% crossover rate to nivolumab in the chemotherapy arm for this subgroup. Conclusions: This is the first pivotal randomized phase 3 trial to incorporate an analysis evaluating TMB and clinical benefit with programmed death-1 inhibitor therapy. The findings of this exploratory retrospective analysis suggest that nivolumab improves ORR and PFS compared with platinum doublet chemotherapy in patients with high TMB. Citation Format: Solange Peters, Benjamin Creelan, Matthew D. Hellmann, Mark A. Socinski, Martin Reck, Prabhu Bhagavatheeswaran, Han Chang, William J. Geese, Luis Paz-Ares, David P. Carbone. Impact of tumor mutation burden on the efficacy of first-line nivolumab in stage iv or recurrent non-small cell lung cancer: An exploratory analysis of CheckMate 026 [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr CT082. doi:10.1158/1538-7445.AM2017-CT082


Cancer Cell | 2018

Genomic Features of Response to Combination Immunotherapy in Patients with Advanced Non-Small-Cell Lung Cancer

Matthew D. Hellmann; Tavi Nathanson; Hira Rizvi; Benjamin C. Creelan; Francisco Sanchez-Vega; Arun Ahuja; Ai Ni; Jacki B. Novik; Levi Mangarin; Mohsen Abu-Akeel; Cailian Liu; Jennifer Sauter; Natasha Rekhtman; Eliza Chang; Margaret K. Callahan; Jamie E. Chaft; Martin H. Voss; Megan Tenet; Xuemei Li; Kelly Covello; Andrea Renninger; Patrik Vitazka; William J. Geese; Hossein Borghaei; Charles M. Rudin; Scott Antonia; Charles Swanton; Jeff Hammerbacher; Taha Merghoub; Nicholas McGranahan

Summary Combination immune checkpoint blockade has demonstrated promising benefit in lung cancer, but predictors of response to combination therapy are unknown. Using whole-exome sequencing to examine non-small-cell lung cancer (NSCLC) treated with PD-1 plus CTLA-4 blockade, we found that high tumor mutation burden (TMB) predicted improved objective response, durable benefit, and progression-free survival. TMB was independent of PD-L1 expression and the strongest feature associated with efficacy in multivariable analysis. The low response rate in TMB low NSCLCs demonstrates that combination immunotherapy does not overcome the negative predictive impact of low TMB. This study demonstrates the association between TMB and benefit to combination immunotherapy in NSCLC. TMB should be incorporated in future trials examining PD-(L)1 with CTLA-4 blockade in NSCLC.


Cancer Control | 2014

Current Clinical Application of Genomic and Proteomic Profiling in Non-Small-Cell Lung Cancer

Tawee Tanvetyanon; Benjamin C. Creelan; Alberto Chiappori

BACKGROUND Genomic or proteomic profiling of cancer can be broadly defined as a systematic grouping of cancer based on its genetic or protein makeup. In the management of non-small-cell lung cancer (NSCLC), genomic and proteomic profiling applications have become useful in early disease detection, diagnosis, treatment, and prognostication. METHODS We reviewed the recent literature on the applications of genomic and proteomic profiling in NSCLC. Important applications were summarized into those already adopted as standard care and those still under investigation. RESULTS For genomic profiling, testing for EGFR mutation and ALK rearrangement has become routine for adenocarcinoma. Multiplex assay and malignancy-risk gene signature are both important applications in development. A test to predict outcome after treatment with an epidermal growth factor rector/tyrosine kinase inhibitor and a screening blood test for lung cancer are being investigated for use in proteomic profiling. CONCLUSIONS Genomic profiling is routine in patients with NSCLC, and proteomic profiling shows promise. Additional genomic and proteomic profiling applications may also prove to be useful contributions in the care of these patients.


Clinical Cancer Research | 2017

MET–GRB2 Signaling-Associated Complexes Correlate with Oncogenic MET Signaling and Sensitivity to MET Kinase Inhibitors

Matthew A. Smith; Thomas Licata; Aliya Lakhani; Marileila Garcia; Hans-Ulrich Schildhaus; Vincent Vuaroqueaux; Balazs Halmos; Alain C. Borczuk; Y. Ann Chen; Benjamin C. Creelan; Theresa A. Boyle; Eric B. Haura

Purpose: Targeting MET in cancer is hampered by lack of diagnostics that accurately reflect high MET signaling and dependence. We hypothesized that assays reflecting MET signaling associated protein complexes could redefine tumors dependent on MET and could add additional precision beyond genomic assessments. Experimental Design: We used biochemical approaches, cellular viability studies, and proximity ligation assays to assess MET dependence. We examined MET signaling complexes in lung cancer patient specimens (N = 406) and patient-derived xenograft (PDX) models of solid tumors (N = 308). We evaluated response to crizotinib in a MET-amplified cohort of PDX models of lung cancer (N = 6) and provide a case report of a lung cancer patient harboring a Δexon14 MET splice variant. Results: We found the interaction of MET with the adaptor protein GRB2 is necessary for oncogenic survival signaling by MET. MET-GRB2 complexes were identified only within MET-amplified PDX models and patient specimens but exhibit substantial variability. Lack of MET-GRB2 complexes was associated with lack of response to MET TKI in cell lines and PDX models. Presence of MET-GRB2 complexes can further subtype tumors with Δexon14 MET splice variants. Presence of these complexes correlated with response to crizotinib in one patient with Δexon14 MET lacking MET gene amplification. Conclusions: Proximity assays measuring MET-GRB2 signaling complexes provide novel insights into MET-mediated signaling and could complement current clinical genomics-based assay platforms. Clin Cancer Res; 23(22); 7084–96. ©2017 AACR.


Expert Review of Anticancer Therapy | 2016

The safety and efficacy of nivolumab in advanced (metastatic) non-small cell lung cancer

Tawee Tanvetyanon; Benjamin C. Creelan; Scott Antonia

ABSTRACT Introduction: Advanced non-small cell lung cancer (NSCLC) is a challenging oncological problem. Following standard initial therapy, disease progression will typically develop. Patients with relapsed or refractory disease are left with limited treatment options. The advent of nivolumab, a monoclonal antibody against Program Death-1 (PD-1), has substantially changed the outlook for such patients. Area covered: Nivolumab is the first checkpoint immunotherapeutic agent to gain regulatory approval for NSCLC. By enabling host immune-mediated cytotoxic activity against tumor cells, nivolumab induces a partial or complete tumor response in 15–20% of patients, regardless of number of previous lines of anti-cancer therapy. Nivolumab-related adverse effects are generally milder and less frequent than those observed with conventional cytotoxic chemotherapy. Although immune-related adverse events such as fatal pneumonitis have been reported with nivolumab therapy, most adverse events are reversible with a prompt immunosuppression. Studies investigating nivolumab in combination with other agents are ongoing. Expert commentary: Nivolumab represents a significant breakthrough in the treatment of advanced NSCLC. Its therapeutic role for NSCLC may soon expand to include consolidation or maintenance setting. Furthermore, several clinical trials investigating the combination of nivolumab with other immunologic or non-immunologic treatments are ongoing and these will likely result in additional roles of nivolumab in NSCLC.


Journal of Thoracic Oncology | 2018

P2.09-17 A Call to Action: Rapid Collection of Post-Mortem Lung Cancer Tissue in the Community to Enable Lung Cancer Research

Theresa A. Boyle; Gwendolyn P. Quinn; Matthew B. Schabath; Teresita Muñoz-Antonia; L. Duarte; Christie Pratt; Dung-Tsa Chen; L.S. Hair; Scott Antonia; Alberto Chiappori; Benjamin C. Creelan; Jhanelle E. Gray; Charles Williams; Eric B. Haura

panel is a 31-gene NGS multiplex assay that synchronously detects multiple oncogene fusion transcripts from formalin-fixed paraffinembedded (FFPE) tissue derived RNA (Figure 1). Method: QIAseq analysis was undertaken in 33 samples (31 NSCLC samples, 2 commercial controls). 12/31 NSCLC samples were positive controls with a known fusion genotype identified by Quantide X NGS or FISH +/RTPCR. The remaining 19/31 fusion negative NSCLC controls included 6 samples with EGFR/KRAS/NRAS mutations. Analysis required a minimum 2x 5u Î M thick FFPE scrolls with >30% neoplastic cell content. Manual RNA extraction was undertaken in all samples except n1⁄46 (ExScale automation extraction). NGS fusion breakpoints, crossing and spanning reads were calculated in QIAseq fusion-detected samples. An additional validation cohort of 40 NSCLC samples, including 20 with unknown fusion status will optimise QIAseq thresholds for fusion detection. Result: 48 QIAseq sequencing experiments was undertaken in 33 samples with 2 sequencing runs in 12 samples. QIAseq analysis detected a corresponding NGS fusion breakpoint in 14/14 (100%) positive controls including EML4-ALK (n1⁄48), CLTC-ALK (n1⁄41), CD74ROS1 (n1⁄43), CCDC6-RET (n1⁄41) and 5-fusion control (n1⁄41). QIAseq analysis was negative in 17/19 (89.4%) negative controls samples including all KRAS (n1⁄44), NRAS (n1⁄41) and EGFR (n1⁄41) mutation samples. QIAseq detected novel fusion gene CD74 Exon 6-CAMK2A Exon 2 in n1⁄41 sample subsequently confirmed on Sanger sequencing. Two separate runs detected TPM3 Exon 8-S100A7A Exon 2 fusion in n1⁄41 sample not identified with Sanger sequencing. Both fusions have uncertain clinical significance. Validation cohort results will be presented. Conclusion: QIAseq detects NSCLC oncogenic fusions with high sensitivity and specificity. Future applications include optimising use of small biopsy specimens for synchronous gene rearrangement screening and identification of novel gene fusion targets.


Journal of Thoracic Oncology | 2018

Hybrid capture-based genomic profiling of circulating tumor DNA from patients with advanced non-small cell lung cancer

Alexa B. Schrock; Allison Welsh; Jon H. Chung; Dean Pavlick; Eric H. Bernicker; Benjamin C. Creelan; Brady Forcier; Jeffrey S. Ross; Philip J. Stephens; Siraj M. Ali; Ibiayi Dagogo-Jack; Alice T. Shaw; Tianhong Li; Sai-Hong Ignatius Ou; Vincent A. Miller

Introduction: Genomic profiling informs selection of matched targeted therapies as part of routine clinical care in NSCLC. Tissue biopsy is the criterion standard; however, genomic profiling of blood‐derived circulating tumor DNA (ctDNA) has emerged as a minimally invasive alternative. Methods: Hybrid capture–based genomic profiling of 62 genes was performed on blood‐based ctDNA from 1552 patients with NSCLC. Results: Evidence of ctDNA was detected in 80% of samples, and in 86% of these cases, at least one reportable genomic alteration (GA) was detected. Frequently altered genes were tumor protein p53 gene (TP53) (59%), EGFR (25%), and KRAS (17%). Comparative analysis with a tissue genomic database (N = 21,500) showed similar frequencies of GAs per gene, although KRAS mutation and EGFR T790M were more frequent in tissue and ctDNA, respectively (both p < 0.0001), likely reflecting the use of liquid versus tissue biopsy after relapse during targeted therapy. In temporally matched ctDNA and tissue samples from 33 patients with evidence of ctDNA in their blood, 64% of GAs detected in tissue were also detected in ctDNA, including 78% of short variants (58 of 74) and 100% of rearrangements (four of four), but only 16% of amplifications (four of 25). Conclusions: Genomic profiling of ctDNA detected clinically relevant GAs in a significant subset of NSCLC cases. Most alterations detected in matched tissue were also detected in ctDNA. These results suggest the utility of ctDNA testing in advanced NSCLC as a complementary approach to tissue testing. Blood‐based ctDNA testing may be particularly useful at the time of progression during targeted therapy.

Collaboration


Dive into the Benjamin C. Creelan's collaboration.

Top Co-Authors

Avatar

Scott Antonia

University of South Florida

View shared research outputs
Top Co-Authors

Avatar

Eric B. Haura

University of South Florida

View shared research outputs
Top Co-Authors

Avatar

Jhanelle E. Gray

University of South Florida

View shared research outputs
Top Co-Authors

Avatar

Alberto Chiappori

University of South Florida

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Charles Williams

University of South Florida

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Tawee Tanvetyanon

University of South Florida

View shared research outputs
Researchain Logo
Decentralizing Knowledge