Network


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

Hotspot


Dive into the research topics where Alan H. Bryce is active.

Publication


Featured researches published by Alan H. Bryce.


Lancet Oncology | 2015

Nivolumab versus chemotherapy in patients with advanced melanoma who progressed after anti-CTLA-4 treatment (CheckMate 037): a randomised, controlled, open-label, phase 3 trial

Jeffrey S. Weber; Sandra P. D'Angelo; David R. Minor; F. Stephen Hodi; Ralf Gutzmer; Bart Neyns; Christoph Hoeller; Nikhil I. Khushalani; Wilson H. Miller; Christopher D. Lao; Gerald P. Linette; Luc Thomas; Paul Lorigan; Kenneth F. Grossmann; Jessica C. Hassel; Michele Maio; Mario Sznol; Paolo Antonio Ascierto; Peter Mohr; Bartosz Chmielowski; Alan H. Bryce; Inge Marie Svane; Jean Jacques Grob; Angela M. Krackhardt; Christine Horak; Alexandre Lambert; Arvin Yang; James Larkin

BACKGROUND Nivolumab, a fully human IgG4 PD-1 immune checkpoint inhibitor antibody, can result in durable responses in patients with melanoma who have progressed after ipilimumab and BRAF inhibitors. We assessed the efficacy and safety of nivolumab compared with investigators choice of chemotherapy (ICC) as a second-line or later-line treatment in patients with advanced melanoma. METHODS In this randomised, controlled, open-label, phase 3 trial, we recruited patients at 90 sites in 14 countries. Eligible patients were 18 years or older, had unresectable or metastatic melanoma, and progressed after ipilimumab, or ipilimumab and a BRAF inhibitor if they were BRAF(V 600) mutation-positive. Participating investigators randomly assigned (with an interactive voice response system) patients 2:1 to receive an intravenous infusion of nivolumab 3 mg/kg every 2 weeks or ICC (dacarbazine 1000 mg/m(2) every 3 weeks or paclitaxel 175 mg/m(2) combined with carboplatin area under the curve 6 every 3 weeks) until progression or unacceptable toxic effects. We stratified randomisation by BRAF mutation status, tumour expression of PD-L1, and previous best overall response to ipilimumab. We used permuted blocks (block size of six) within each stratum. Primary endpoints were the proportion of patients who had an objective response and overall survival. Treatment was given open-label, but those doing tumour assessments were masked to treatment assignment. We assessed objective responses per-protocol after 120 patients had been treated with nivolumab and had a minimum follow-up of 24 weeks, and safety in all patients who had had at least one dose of treatment. The trial is closed and this is the first interim analysis, reporting the objective response primary endpoint. This study is registered with ClinicalTrials.gov, number NCT01721746. FINDINGS Between Dec 21, 2012, and Jan 10, 2014, we screened 631 patients, randomly allocating 272 patients to nivolumab and 133 to ICC. Confirmed objective responses were reported in 38 (31·7%, 95% CI 23·5-40·8) of the first 120 patients in the nivolumab group versus five (10·6%, 3·5-23·1) of 47 patients in the ICC group. Grade 3-4 adverse events related to nivolumab included increased lipase (three [1%] of 268 patients), increased alanine aminotransferase, anaemia, and fatigue (two [1%] each); for ICC, these included neutropenia (14 [14%] of 102), thrombocytopenia (six [6%]), and anaemia (five [5%]). We noted grade 3-4 drug-related serious adverse events in 12 (5%) nivolumab-treated patients and nine (9%) patients in the ICC group. No treatment-related deaths occurred. INTERPRETATION Nivolumab led to a greater proportion of patients achieving an objective response and fewer toxic effects than with alternative available chemotherapy regimens for patients with advanced melanoma that has progressed after ipilimumab or ipilimumab and a BRAF inhibitor. Nivolumab represents a new treatment option with clinically meaningful durable objective responses in a population of high unmet need. FUNDING Bristol-Myers Squibb.


PLOS Genetics | 2014

Integrated genomic characterization reveals novel, therapeutically relevant drug targets in FGFR and EGFR pathways in sporadic intrahepatic cholangiocarcinoma.

Mitesh J. Borad; Mia D. Champion; Jan B. Egan; Winnie S. Liang; Rafael Fonseca; Alan H. Bryce; Ann E. McCullough; Michael T. Barrett; Katherine S. Hunt; Maitray D. Patel; Scott W. Young; Joseph M. Collins; Alvin C. Silva; Rachel M. Condjella; Matthew S. Block; Robert R. McWilliams; Konstantinos N. Lazaridis; Eric W. Klee; Keith C. Bible; Pamela Jo Harris; Gavin R. Oliver; Jaysheel D. Bhavsar; Asha Nair; Sumit Middha; Yan W. Asmann; Jean Pierre A Kocher; Kimberly A. Schahl; Benjamin R. Kipp; Emily G. Barr Fritcher; Angela Baker

Advanced cholangiocarcinoma continues to harbor a difficult prognosis and therapeutic options have been limited. During the course of a clinical trial of whole genomic sequencing seeking druggable targets, we examined six patients with advanced cholangiocarcinoma. Integrated genome-wide and whole transcriptome sequence analyses were performed on tumors from six patients with advanced, sporadic intrahepatic cholangiocarcinoma (SIC) to identify potential therapeutically actionable events. Among the somatic events captured in our analysis, we uncovered two novel therapeutically relevant genomic contexts that when acted upon, resulted in preliminary evidence of anti-tumor activity. Genome-wide structural analysis of sequence data revealed recurrent translocation events involving the FGFR2 locus in three of six assessed patients. These observations and supporting evidence triggered the use of FGFR inhibitors in these patients. In one example, preliminary anti-tumor activity of pazopanib (in vitro FGFR2 IC50≈350 nM) was noted in a patient with an FGFR2-TACC3 fusion. After progression on pazopanib, the same patient also had stable disease on ponatinib, a pan-FGFR inhibitor (in vitro, FGFR2 IC50≈8 nM). In an independent non-FGFR2 translocation patient, exome and transcriptome analysis revealed an allele specific somatic nonsense mutation (E384X) in ERRFI1, a direct negative regulator of EGFR activation. Rapid and robust disease regression was noted in this ERRFI1 inactivated tumor when treated with erlotinib, an EGFR kinase inhibitor. FGFR2 fusions and ERRFI mutations may represent novel targets in sporadic intrahepatic cholangiocarcinoma and trials should be characterized in larger cohorts of patients with these aberrations.


European Journal of Haematology | 2008

Peripheral blood stem cell transplant for POEMS syndrome is associated with high rates of engraftment syndrome

Angela Dispenzieri; Martha Q. Lacy; Suzanne R. Hayman; Shaji Kumar; Francis Buadi; David Dingli; Mark R. Litzow; Dennis A. Gastineau; David J. Inwards; Michelle A. Elliott; Ivana N. Micallef; Stephen M. Ansell; William J. Hogan; Luis F. Porrata; Patrick A. Johnston; Bekele Afessa; Alan H. Bryce; Robert A. Kyle; Morie A. Gertz

Polyneuropathy, organomegaly, endocrinopathy, M protein and skin changes (POEMS) syndrome is a devastating syndrome, characterized by peripheral neuropathy, organomegaly, endocrinopathy, monoclonal plasma cells, skin changes, papilledema, volume overload, sclerotic bone lesions, thrombocytosis and high vascular endothelial growth factor (VEGF). High‐dose chemotherapy with autologous peripheral blood stem cell transplantation (ASCT) ultimately yields excellent clinical responses, but there can be considerable peritransplant morbidity. We have treated 30 POEMS patients with ASCT at Mayo Clinic, Rochester. During transplant period, patients had high rates of fever, diarrhea, weight gain and rash (93%, 77%, 53% and 43%, respectively). Only 13% remained outpatient, and median time to discharge from hospital was transplant day 17 (range 0–175). Splenomegaly was the baseline factor that best predicted for a complicated peritransplant course. Depending on the definition used, ∼50% of patients satisfied criteria for engraftment syndrome. Earlier and more aggressive use of corticosteroids may be associated with less complicated post‐transplant courses. Median overall survival has not been reached; the treatment‐related mortality was 3%. In addition, important clinical improvements and reductions in plasma VEGF levels can occur in the absence of significant decrease in the monoclonal protein. Unraveling the mechanisms of the syndrome both in the context of ASCT and in general are challenges for the future.


Haematologica | 2009

Translocation t(11;14) and survival of patients with light chain (AL) amyloidosis

Alan H. Bryce; Rhett P. Ketterling; Morie A. Gertz; Martha Q. Lacy; Ryan A. Knudson; Steven R. Zeldenrust; Shaji Kumar; Suzanne R. Hayman; Francis Buadi; Robert A. Kyle; Philip R. Greipp; John A. Lust; Stephen J. Russell; S. Vincent Rajkumar; Rafael Fonseca; Angela Dispenzieri

Little is known about cytogenetic abnormalities in patients with light chain (AL) amyloidosis. The findings of this study suggest that interphase FISH coupled to cytoplasmic staining of specific Ig on bone marrow cells could be useful in light chain (AL) amyloidosis, and that t(11;14) is an adverse prognostic factor in these patients. Background Light chain amyloidosis is a rare plasma cell dyscrasia. Interphase fluorescence in situ hybridization (FISH) coupled to cytoplasmic staining of specific Ig (cIg-FISH) on bone marrow plasma cells has become well established in the initial evaluation of multiple myeloma, a related disorder. Little, however, is known about cytogenetic abnormalities in patients with light chain amyloidosis. Design and Methods We reviewed 56 patients with light chain amyloidosis who had cIg-FISH performed as part of their routine clinical testing using the standard screening panel employed in multiple myeloma at our institution. Results Seventy percent of patients had abnormal cIg-FISH, with the most common abnormalities being IgH translocations [48%] – including t(11;14) [39%], and t(14;16) [2%] – and del13/del13q [30%]. No t(4;14) or deletions of 17p (p53) were observed. Patients with t(11;14) had the lowest levels of clonal plasma cells, and those with del13 had the highest. The risk of death for patients harboring the t(11;14) translocation was 2.1 (CI 1.04–6.4), which on multivariate analysis was independent of therapy. Conclusions Although preliminary, our data would suggest that cIg-FISH testing is important in patients with light chain amyloidosis and that t(11;14) is an adverse prognostic factor in these patients.


American Journal of Medical Genetics Part C-seminars in Medical Genetics | 2014

Implementing individualized medicine into the medical practice

Konstantinos N. Lazaridis; Tammy M. McAllister; Dusica Babovic-Vuksanovic; Scott A. Beck; Mitesh J. Borad; Alan H. Bryce; Asher Chanan-Khan; Matthew J. Ferber; Rafael Fonseca; Kiley J. Johnson; Eric W. Klee; Noralane M. Lindor; Jennifer B. McCormick; Robert R. McWilliams; Alexander S. Parker; Douglas L. Riegert-Johnson; Carolyn R. Rohrer Vitek; Kimberly A. Schahl; Cloann Schultz; Keith Stewart; George C. Then; Eric D. Wieben; Gianrico Farrugia

There is increasing recognition that genomic medicine as part of individualized medicine has a defined role in patient care. Rapid advances in technology and decreasing cost combine to bring genomic medicine closer to the clinical practice. There is also growing evidence that genomic‐based medicine can advance patient outcomes, tailor therapy and decrease side effects. However the challenges to integrate genomics into the workflow involved in patient care remain vast, stalling assimilation of genomic medicine into mainstream medical practice. In this review we describe the approach taken by one institution to further individualize medicine by offering, executing and interpreting whole exome sequencing on a clinical basis through an enterprise‐wide, standalone individualized medicine clinic. We present our experience designing and executing such an individualized medicine clinic, sharing lessons learned and describing early implementation outcomes.


Cancer immunology research | 2015

PD-1 and PD-L1 Expression in Renal Cell Carcinoma with Sarcomatoid Differentiation

Richard W. Joseph; Sherri Z. Millis; Estrella M. Carballido; David Bryant; Zoran Gatalica; Sandeep K. Reddy; Alan H. Bryce; Nicholas J. Vogelzang; Erik P. Castle; Thai H. Ho

Sarcomatoid renal cell cancer (RCC) is an aggressive form of RCC that responds poorly to IL2 immunotherapy. Both PD-1 and PD-L1 were found expressed in sarcomatoid RCC samples, suggesting that blockade of the PD-L1/PD-1 pathway may have immunotherapeutic potential. Monoclonal antibodies that target the programmed death-1 (PD-1)–programmed death ligand-1 (PD-L1) axis have antitumor activity against multiple cancers. The presence of sarcomatoid differentiation in renal cell carcinoma (RCC) is associated with resistance to targeted therapy and poor responses to IL2 immunotherapy. Given the aggressive nature of RCC with sarcomatoid differentiation and the exclusion of sarcomatoid histology from metastatic RCC clinical trials, less is understood regarding selection of therapies. Here, we characterized the PD-1/PD-L1 axis in RCC with sarcomatoid differentiation. We directly compared two PD-L1 antibodies and found concordance of PD-L1 positivity in 89% of tested RCCs with sarcomatoid differentiation. Coexpression of PD-L1 on neoplastic cells and the presence of PD-1–positive tumor-infiltrating lymphocytes were identified in 50% (13 of 26) of RCCs with sarcomatoid differentiation. In contrast, only 1 of 29 clear cell RCCs (3%) had concurrent expression of PD-L1 and PD-1 (P = 0.002). Our study suggests that RCC with sarcomatoid differentiation may express PD-1/PD-L1 at a higher percentage than RCC without sarcomatoid differentiation, and patients with these tumors may be good candidates for treatment with anti–PD-1/PD-L1 therapies. Cancer Immunol Res; 3(12); 1303–7. ©2015 AACR.


BMJ Open | 2016

Determining the frequency of pathogenic germline variants from exome sequencing in patients with castrate-resistant prostate cancer

Steven N. Hart; Marissa S. Ellingson; Kim Schahl; Peter T. Vedell; Rachel Carlson; Jason P. Sinnwell; Poulami Barman; Hugues Sicotte; Jeanette E. Eckel-Passow; Liguo Wang; Krishna R. Kalari; Rui Qin; Teresa M. Kruisselbrink; Rafael E. Jimenez; Alan H. Bryce; Winston Tan; Richard M. Weinshilboum; Liewei Wang; Manish Kohli

Objectives To determine the frequency of pathogenic inherited mutations in 157 select genes from patients with metastatic castrate-resistant prostate cancer (mCRPC). Design Observational. Setting Multisite US-based cohort. Participants Seventy-one adult male patients with histological confirmation of prostate cancer, and had progressive disease while on androgen deprivation therapy. Results Twelve patients (17.4%) showed evidence of carrying pathogenic or likely pathogenic germline variants in the ATM, ATR, BRCA2, FANCL, MSR1, MUTYH, RB1, TSHR and WRN genes. All but one patient opted in to receive clinically actionable results at the time of study initiation. We also found that pathogenic germline BRCA2 variants appear to be enriched in mCRPC compared to familial prostate cancers. Conclusions Pathogenic variants in cancer-susceptibility genes are frequently observed in patients with mCRPC. A substantial proportion of patients with mCRPC or their family members would derive clinical utility from mutation screening. Trial registration number NCT01953640; Results.


Molecular Cancer Therapeutics | 2015

Pilot Trial of Selecting Molecularly Guided Therapy for Patients with Non–V600 BRAF-Mutant Metastatic Melanoma: Experience of the SU2C/MRA Melanoma Dream Team

Patricia LoRusso; Scott A. Boerner; Mary Jo Pilat; Karen M. Forman; Clarice Zuccaro; Jeffrey Kiefer; Winnie S. Liang; Sally Hunsberger; Bruce G. Redman; Svetomir N. Markovic; Aleksandar Sekulic; Alan H. Bryce; Richard W. Joseph; C. Lance Cowey; Leslie A. Fecher; Jeffrey A. Sosman; Paul B. Chapman; Gary K. Schwartz; David Craig; John D. Carpten; Jeffrey M. Trent

Targeted therapies and immunotherapies have led to significant improvements in the treatment of advanced cancers, including metastatic melanoma. However, new strategies are desperately needed to overcome therapeutic resistance to these agents, as well as to identify effective treatment approaches for cancer patients that fall outside major targetable mutational subtypes (e.g., non–V600 BRAF melanoma). One such strategy is to extend the paradigm of individually tailored, molecularly targeted therapy into a broader spectrum of melanoma patients, particularly those bearing tumors without commonly recognized therapeutic targets, as well as having failed or were ineligible for immunotherapy. In this nontreatment pilot study, next-generation sequencing (NGS) technologies were utilized, including whole genome and whole transcriptome sequencing, to identify molecular aberrations in patients with non–V600 BRAF metastatic melanoma. This information was then rationally matched to an appropriate clinical treatment from a defined pharmacopeia. Five patients with advanced non–V600 BRAF metastatic melanoma were enrolled. We demonstrated successful performance of the following during a clinically relevant time period: patient tumor biopsy, quality DNA/RNA extraction, DNA/RNA-based sequencing for gene expression analysis, analysis utilizing a series of data integration methodologies, report generation, and tumor board review with formulated treatment plan. Streamlining measures were conducted based on the experiences of enrolling, collecting specimens, and analyzing the molecular signatures of patients. We demonstrated the feasibility of using NGS to identify molecular aberrations and generate an individualized treatment plan in this patient population. A randomized treatment study utilizing lessons learned from the conduct of this pilot study is currently underway. Mol Cancer Ther; 14(8); 1962–71. ©2015 AACR.


Scientific Reports | 2016

Clinical Implementation of Integrated Genomic Profiling in Patients with Advanced Cancers

Mitesh J. Borad; Jan B. Egan; Rachel M. Condjella; Winnie S. Liang; Rafael Fonseca; Nicole R. Ritacca; Ann E. McCullough; Michael T. Barrett; Katherine S. Hunt; Mia D. Champion; Maitray D. Patel; Scott W. Young; Alvin C. Silva; Thai H. Ho; Thorvardur R. Halfdanarson; Robert R. McWilliams; Konstantinos N. Lazaridis; Ramesh K. Ramanathan; Angela Baker; Jessica Aldrich; Ahmet Kurdoglu; Tyler Izatt; Alexis Christoforides; Irene Cherni; Sara Nasser; Rebecca Reiman; Lori Cuyugan; Jacquelyn McDonald; Jonathan Adkins; Stephen D. Mastrian

DNA focused panel sequencing has been rapidly adopted to assess therapeutic targets in advanced/refractory cancer. Integrated Genomic Profiling (IGP) utilising DNA/RNA with tumour/normal comparisons in a Clinical Laboratory Improvement Amendments (CLIA) compliant setting enables a single assay to provide: therapeutic target prioritisation, novel target discovery/application and comprehensive germline assessment. A prospective study in 35 advanced/refractory cancer patients was conducted using CLIA-compliant IGP. Feasibility was assessed by estimating time to results (TTR), prioritising/assigning putative therapeutic targets, assessing drug access, ascertaining germline alterations, and assessing patient preferences/perspectives on data use/reporting. Therapeutic targets were identified using biointelligence/pathway analyses and interpreted by a Genomic Tumour Board. Seventy-five percent of cases harboured 1–3 therapeutically targetable mutations/case (median 79 mutations of potential functional significance/case). Median time to CLIA-validated results was 116 days with CLIA-validation of targets achieved in 21/22 patients. IGP directed treatment was instituted in 13 patients utilising on/off label FDA approved drugs (n = 9), clinical trials (n = 3) and single patient IND (n = 1). Preliminary clinical efficacy was noted in five patients (two partial response, three stable disease). Although barriers to broader application exist, including the need for wider availability of therapies, IGP in a CLIA-framework is feasible and valuable in selection/prioritisation of anti-cancer therapeutic targets.


International Journal of Urology | 2016

Androgen receptor splice variant 7 in castration-resistant prostate cancer: Clinical considerations.

Alan H. Bryce; Emmanuel S. Antonarakis

Constitutively‐active ligand‐independent splice variants of the androgen receptor are an adaptive response by prostate cancer cells to escape androgen deprivation therapy and novel androgen receptor‐directed treatments. Androgen receptor splice variant 7 is the most common splice variant detected in clinical biospecimens, and emerging data now suggest that the presence of tumoral androgen receptor splice variant 7 might be indicative of primary and acquired resistance to next‐generation androgen pathway inhibitors, such as abiraterone and enzalutamide. At the same time, taxane chemotherapy might retain its efficacy regardless of androgen receptor splice variant 7 status, thus suggesting the potential for a predictive biomarker guiding treatment selection in men with metastatic castration‐resistant prostate cancer. Herein, we review the preclinical data elucidating the structure and function of androgen receptor splice variant 7, we describe the existing clinical data using this biomarker in metastatic castration‐resistant prostate cancer, and we highlight potential therapeutic strategies to target androgen receptor splice variant 7‐expressing prostate cancer.

Collaboration


Dive into the Alan H. Bryce's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge