Network


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

Hotspot


Dive into the research topics where Brad Beattie is active.

Publication


Featured researches published by Brad Beattie.


Nature | 2016

The epichaperome is an integrated chaperome network that facilitates tumour survival

Anna Rodina; Tai Wang; Pengrong Yan; Erica DaGama Gomes; Mark Dunphy; Nagavarakishore Pillarsetty; John Koren; John F. Gerecitano; Tony Taldone; Hongliang Zong; Eloisi Caldas-Lopes; Mary L. Alpaugh; Adriana D. Corben; Matthew Riolo; Brad Beattie; Christina Pressl; Radu Ioan Peter; Chao Xu; Robert Trondl; Hardik J. Patel; Fumiko Shimizu; Alexander Bolaender; Chenghua Yang; Palak Panchal; Mohammad Farooq; Sarah Kishinevsky; Shanu Modi; Oscar Lin; Feixia Chu; Sujata Patil

Transient, multi-protein complexes are important facilitators of cellular functions. This includes the chaperome, an abundant protein family comprising chaperones, co-chaperones, adaptors, and folding enzymes—dynamic complexes of which regulate cellular homeostasis together with the protein degradation machinery. Numerous studies have addressed the role of chaperome members in isolation, yet little is known about their relationships regarding how they interact and function together in malignancy. As function is probably highly dependent on endogenous conditions found in native tumours, chaperomes have resisted investigation, mainly due to the limitations of methods needed to disrupt or engineer the cellular environment to facilitate analysis. Such limitations have led to a bottleneck in our understanding of chaperome-related disease biology and in the development of chaperome-targeted cancer treatment. Here we examined the chaperome complexes in a large set of tumour specimens. The methods used maintained the endogenous native state of tumours and we exploited this to investigate the molecular characteristics and composition of the chaperome in cancer, the molecular factors that drive chaperome networks to crosstalk in tumours, the distinguishing factors of the chaperome in tumours sensitive to pharmacologic inhibition, and the characteristics of tumours that may benefit from chaperome therapy. We find that under conditions of stress, such as malignant transformation fuelled by MYC, the chaperome becomes biochemically ‘rewired’ to form a network of stable, survival-facilitating, high-molecular-weight complexes. The chaperones heat shock protein 90 (HSP90) and heat shock cognate protein 70 (HSC70) are nucleating sites for these physically and functionally integrated complexes. The results indicate that these tightly integrated chaperome units, here termed the epichaperome, can function as a network to enhance cellular survival, irrespective of tissue of origin or genetic background. The epichaperome, present in over half of all cancers tested, has implications for diagnostics and also provides potential vulnerability as a target for drug intervention.


Journal of Clinical Investigation | 2007

Improved tumor imaging and therapy via i.v. IgG-mediated time-sequential modulation of neonatal Fc receptor.

Jaspreet Singh Jaggi; Jorge A. Carrasquillo; Surya V. Seshan; Pat Zanzonico; Erik Henke; Andrew Nagel; Jazmin Schwartz; Brad Beattie; Barry J. Kappel; Debjit Chattopadhyay; Jing Xiao; George Sgouros; Steven M. Larson; David A. Scheinberg

The long plasma half-life of IgG, while allowing for enhanced tumor uptake of tumor-targeted IgG conjugates, also results in increased background activity and normal-tissue toxicity. Therefore, successful therapeutic uses of conjugated antibodies have been limited to the highly sensitive and readily accessible hematopoietic tumors. We report a therapeutic strategy to beneficially alter the pharmacokinetics of IgG antibodies via pharmacological inhibition of the neonatal Fc receptor (FcRn) using high-dose IgG therapy. IgG-treated mice displayed enhanced blood and whole-body clearance of radioactivity, resulting in better tumor-to-blood image contrast and protection of normal tissue from radiation. Tumor uptake and the resultant therapeutic response was unaltered. Furthermore, we demonstrated the use of this approach for imaging of tumors in humans and discuss its potential applications in cancer imaging and therapy. The ability to reduce the serum persistence of conjugated IgG antibodies after their infusion can enhance their therapeutic index, resulting in improved therapeutic and diagnostic efficacy.


JAMA Oncology | 2017

Positron Emission Tomography/Computed Tomography–Based Assessments of Androgen Receptor Expression and Glycolytic Activity as a Prognostic Biomarker for Metastatic Castration-Resistant Prostate Cancer

Josef J. Fox; Somali Gavane; Estelle Blanc-Autran; Sadek A. Nehmeh; Mithat Gonen; Brad Beattie; Hebert Alberto Vargas; Heiko Schöder; John L. Humm; Samson W. Fine; Jason S. Lewis; Stephen B. Solomon; Joseph R. Osborne; Darren R. Veach; Charles L. Sawyers; Wolfgang A. Weber; Howard I. Scher; Michael J. Morris; Steven M. Larson

Importance Androgen receptor–signaling inhibitor (ARSi) drugs prolong life in metastatic castration–resistant prostate cancer (mCRPC), but such tumors eventually become resistant and progress. Comprehensive positron emission tomography/computed tomography (PET/CT) imaging using fluoro-2-D-deoxyglucose F 18 ([18F]-FDG) for glycolysis (Glyc) and fluorodihydrotestosterone F 18 ([18F]-FDHT) for androgen receptor (AR) expression determine heterogeneity of imaging phenotypes, which may be useful in distinguishing patients who will benefit from ARSi drugs from those who need alternative treatments. Objective To test the hypothesis that PET/CT-based assessments of AR expression and glycolytic activity would reveal heterogeneity affecting prognosis. Design, Setting, and Participants Between April 6, 2007, and October 4, 2012, patients with mCRPC underwent imaging with both [18F]-FDG and [18F]-FDHT at Memorial Sloan Kettering Cancer Center. The patients were naive to ARSi treatment with enzalutamide or abiraterone acetate and were referred during documented disease progression. Image-directed biopsy determined the presence or absence of prostate cancer at positive imaging sites. Interventions PET/CT imaging was performed with [18F]-FDHT and [18F]-FDG; select individual lesions were biopsied to correlate imaging phenotype with histologic findings. Main Outcomes and Measures All metabolically active lesions were interpreted as [18F]-FDHT-positive (AR1) or [18F]-FDHT-negative (AR0) and as [18F]-FDG-positive (Glyc1) or [18F]-FDG-negative (Glyc0). Correlation was performed with overall survival for both individual lesion imaging phenotype as well as patient-specific imaging phenotype. Results The mean (SD) age of the 133 patients was 68 (8.6) years. Imaging phenotypes of 2405 PET/CT-positive lesions (median, 12.0 per patient) included 1713 (71.2%) AR1Glyc1, 386 (16.0%) AR1Glyc0, and 306 (12.7%) AR0Glyc1. On multivariate analysis, each phenotype had an independent negative impact effect on survival, most pronounced for AR0Glyc1 lesions (hazard ratio [HR], 1.11; 95% CI, 1.05-1.16; P < .001), followed by AR1Glyc1 lesions (HR, 1.05; 95% CI, 1.03-1.06; P < .001) and AR1Glyc0 lesions (HR, 1.03; 95% CI, 1.00-1.05; P = .048). When sorted by lesion type, 4 patient-specific groups emerged: (1) concordant, with all AR1Glyc1 (34 patients [25.6%]); (2) AR predominant, with AR1Glyc1 and varying numbers of AR1Glyc0 (33 [24.8%]); (3) Glyc predominant, with AR1Glyc1 and varying numbers of AR0Glyc1 (40 [30.1%]); and (4) mixed, with AR1Glyc1 plus a mixture of varying numbers of AR1Glyc0 and AR0Glyc1 (26 [19.5%]). Conclusions and Relevance Heterogeneity of PET/CT imaging phenotype has clinical relevance on a lesion and individual patient level. With regard to mCRPC lesions, most express ARs, consistent with initial benefit of ARSi drugs. On a patient basis, 49% (groups 3 and 4) had at least 1 AR0Glyc1 lesion—the imaging phenotype with the most negative effect on survival, possibly due to ARSi resistance.


Nuclear Medicine Communications | 2014

How to assess background activity: introducing a histogram-based analysis as a first step for accurate one-step PET quantification.

Irene A. Burger; Hebert Alberto Vargas; Brad Beattie; Debra A. Goldman; Junting Zheng; Steven M. Larson; John L. Humm; Charles Schmidtlein

Many common PET segmentation methods for malignant lesions use surrounding background activity as a reference. To date, background has to be measured by drawing a second volume of interest (VOI) in nearby, undiseased tissue. This is time consuming as two VOIs have to be determined for each lesion. The aim of our study was to analyse whether background activity in different organs and body regions could be calculated from the tumour VOI by histogram analyses. The institutional review board waived informed consent for this retrospective study. For each of the following tumour types and areas – head and neck (neck), lung, hepatic metastasis (liver), melanoma (skin), and cervix (pelvis) – 10 consecutive patients with biopsy-proven tumours who underwent 18F-fluorodeoxyglucose-PET in January 2012 were retrospectively selected. One lesion was selected and two readers drew a cubical VOI around the lesion (VOItumour) and over the background (VOIBG). The mean value of VOIBG was compared with the mode of the histogram, using equivalence testing with an equivalence margin of ±0.5 SUV. Inter-reader agreement was analysed for the mean background, and the mode of the VOItumour histogram was assessed using the concordance correlation coefficient. For both readers, the mode of VOItumour was equivalent to the mean of VOIBG (P<0.0001 for R1 and R2). The inter-reader agreement was almost perfect, with a concordance correlation coefficient of greater than 0.92 for both the mode of VOItumour and the mean of VOIBG. Background activity determined within a tumour VOI using histogram analysis is equivalent to separately measured mean background values, with an almost perfect inter-reader agreement. This could facilitate PET quantification methods based on background values without increasing workload.


Cancer Epidemiology, Biomarkers & Prevention | 2018

Abstract B39: A pilot study of neuropsychological functions, APOE, and amyloid imaging in patients with gliomas

Denise D. Correa; Maria Kryza-Lacombe; Xiaosun Zhou; Raymond E. Baser; Brad Beattie; Zodina Beiene; Irene Orlow; John L. Humm; Lisa M. DeAngelis; Wolfgang A. Weber; Joseph R. Osborne

Patients with brain tumors treated with radiotherapy (RT) often develop cognitive dysfunction, and recent studies suggest that the apolipoprotein (APOE) ϵ-4 allele may influence cognitive outcome. The ϵ-4 allele promotes beta (β) amyloid deposition in the cortex, and preliminary evidence suggests that RT may be associated with β-amyloid deposition. In this pilot study, we assessed neuropsychological functions and β-amyloid retention using 18F-florbetaben PET in a subset of brain tumor patients who participated in our study of APOE polymorphisms and cognitive functions. Twenty glioma patients treated with conformal RT ± chemotherapy participated in the study: 6 were APOE ϵ-4 carriers and 14 were non-ϵ-4 carriers. Patients completed a neuropsychological reevaluation and brain MRI and 18F-florbetaben PET scans. The results of Wilcoxon rank sums test comparisons between prior and current assessments showed a significant decline in selective attention (Brief Test of Attention, p=0.018), and a near-significant decline in verbal learning (Hopkins Verbal Learning Test-Learning, p=0.07). Comparisons between assessments by APOE status showed a significant decline in attention and working memory (WAIS-III digits forward, p=0.028 and digits backward, p=0.032) among APOE ϵ-4 carriers. Comparisons of PET regional standard uptake value ratios (SUVRs) showed near-significant differences for the medial temporal cortex (p=0.069) and the putamen (p=0.069), with non-ϵ-4 carriers having higher SUVRs. The findings suggest that glioma patients may experience progressive worsening in attention and executive functions several years after treatment with RT ± chemotherapy, and that the APOE ϵ-4 allele may moderate cognitive decline. Note: This abstract was not presented at the conference. Citation Format: Denise Correa, Maria Kryza-Lacombe, Xiaosun Zhou, Raymond Baser, Brad Beattie, Zodina Beiene, Irene Orlow, John Humm, Lisa DeAngelis, Wolfgang Weber, Joseph Osborne. A pilot study of neuropsychological functions, APOE, and amyloid imaging in patients with gliomas [abstract]. In: Proceedings of the Tenth AACR Conference on the Science of Cancer Health Disparities in Racial/Ethnic Minorities and the Medically Underserved; 2017 Sep 25-28; Atlanta, GA. Philadelphia (PA): AACR; Cancer Epidemiol Biomarkers Prev 2018;27(7 Suppl):Abstract nr B39.


Cancer Research | 2015

Abstract 5444: Development of a noninvasive assay to determine drug concentration in tumor during hsp90 inhibitor therapy

Tony Taldone; Nagavarakishore Pillarsetty; Mark Dunphy; John F. Gerecitano; Eloisi Caldas-Lopes; Brad Beattie; Radu Ioan Peter; Yanlong Kang; Anna Rodina; Pengrong Yan; Erica DaGama Gomes; Alexander Bolaender; Christina Pressl; Blesida Punzalan; Anson Ku; Thomas Ku; Smit K. Shah; Mohammad Uddin; Mei H. Chen; Elmer Santos; Jacek Koziorowski; Adriana D. Corben; Shanu Modi; Komal Jhaveri; Oscar Lin; Efsevia Vakiani; Yelena Y. Janjigian; Pat Zanzonico; Clifford A. Hudis; Steven M. Larson

As molecularly targeted agents assume a more prominent role in anticancer therapy there is a growing need to determine in a noninvasive manner whether the target is being engaged and to what extent such drug-target binding results in desirable effects. We address this need in the context of Hsp90, a target of significant value and one in critical need for such assessment tools, by combining a novel chemical tool selective for tumor Hsp90 with PET imaging and mathematical modeling. The chemical tool is [124I]-PU-H71, the iodine-124 radiolabeled analog of the potent Hsp90 inhibitor PU-H71, which can be administered in tracer quantities for PET imaging. The resulting diagnostic, PU-PET, has been optimized and validated preclinically in mouse models of cancer and then translated to the clinic. The exquisite design of this assay is based on three essential concepts as it relates to the target (Hsp90) as well as to the PET tracer (124I-PU-H71). First, the target is “oncogenic” Hsp90 and has been shown by numerous biochemical and pharmacokinetic studies to have a strong affinity for inhibitors and a very low koff resulting in selective and prolonged retention in tumor. Secondly, the tracer incorporates a 124I in place of the naturally occurring 127I in the structure of PU-H71 and therefore there is no change in the chemical structure. This feature in a PET tracer intended as a companion diagnostic is unprecedented and ensures that the PK properties are identical to the therapeutic agent (PU-H71). Finally, the radionuclide 124I has a four-day half-life and thus is well-suited to monitor the extended tumor retention profile observed for Hsp90 inhibitors. We here demonstrate that this PET assay informs on Hsp90 targeting in individual tumors in real time and provides accurate tumor drug concentrations for at least four chemically distinct Hsp90 drugs. In contrast, we find that plasma pharmacokinetics is not predictive of intratumor parameters and therefore provides limited value in estimating target engagement. Using PU-PET we demonstrate that at least one Hsp90 inhibitor exhibits tumor targeting and retention in humans, delivering and retaining therapeutic, micromolar, concentrations at safe doses. PU-PET is currently being evaluated in Phase 0/1 (NCT01269593) clinical trials as a noninvasive companion diagnostic to determine intratumoral concentration as well as to identify those patients who would best benefit from Hsp90 inhibitor therapy. This diagnostic assay is intended to be incorporated into future Phase 2 clinical trials in order to preselect those patients who would most likely benefit from Hsp90 inhibitor treatment. Citation Format: Tony Taldone, Nagavarakishore Pillarsetty, Mark PS Dunphy, John F. Gerecitano, Eloisi Caldas-Lopes, Brad Beattie, Radu I. Peter, Yanlong Kang, Anna Rodina, Pengrong Yan, Erica M. DaGama Gomes, Alexander Bolaender, Christina Pressl, Blesida Punzalan, Anson Ku, Thomas Ku, Smit Shah, Mohammad Uddin, Mei H. Chen, Elmer Santos, Jacek Koziorowski, Adriana Corben, Shanu Modi, Komal Jhaveri, Oscar Lin, Efsevia Vakiani, Yelena Janjigian, Pat Zanzonico, Clifford Hudis, Steven M. Larson, Jason S. Lewis, Gabriela Chiosis. Development of a noninvasive assay to determine drug concentration in tumor during hsp90 inhibitor therapy. [abstract]. In: Proceedings of the 106th Annual Meeting of the American Association for Cancer Research; 2015 Apr 18-22; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2015;75(15 Suppl):Abstract nr 5444. doi:10.1158/1538-7445.AM2015-5444


The Journal of Nuclear Medicine | 2004

Tumor Localization of 16β-18F-Fluoro-5α-Dihydrotestosterone Versus 18F-FDG in Patients with Progressive, Metastatic Prostate Cancer

Steven M. Larson; Michael J. Morris; Ilonka Gunther; Brad Beattie; John L. Humm; Timothy Akhurst; Ronald D. Finn; Yusuf E. Erdi; Keith S. Pentlow; Jon Dyke; Olivia Squire; William G. Bornmann; Timothy J. McCarthy; Michael J. Welch; Howard I. Scher


International Journal of Radiation Oncology Biology Physics | 2016

Strategy of Using Intratreatment Hypoxia Imaging to Selectively and Safely Guide Radiation Dose De-escalation Concurrent With Chemotherapy for Locoregionally Advanced Human Papillomavirus-Related Oropharyngeal Carcinoma.

Nancy Y. Lee; Heiko Schöder; Brad Beattie; R.M. Lanning; Nadeem Riaz; S. McBride; Nora Katabi; Duan Li; Brett Yarusi; Susie Chan; Lindsey Mitrani; Zhigang Zhang; David G. Pfister; Eric J. Sherman; Shrujal S. Baxi; Jay O. Boyle; Luc G. T. Morris; Ian Ganly; Richard J. Wong; John L. Humm


International Journal of Radiation Oncology Biology Physics | 2016

[18F]-Fluorocholine Uptake Correlates with Pathologic Evidence of Recurrent Tumor After SRS for Brain Metastases

Zachary A. Kohutek; R.G. Blasberg; John L. Humm; L.E. Blumberg; Cameron Brennan; Viviane Tabar; P.H. Gutin; Brad Beattie; Marc K. Rosenblum; Jason T. Huse; Heiko Schöder; Kathryn Beal


International Journal of Radiation Oncology Biology Physics | 2014

Preliminary Results of a Prospective Trial of IMRT Dose De-escalation to Gross Nodal Disease in Human Papillomavirus (HPV)-Positive Oropharyngeal Carcinoma (OPC) Based on Assessment of Tumor Hypoxia Using 18F-FMISO PET Imaging

R.M. Lanning; Brad Beattie; John L. Humm; Pat Zanzonico; S. Rao; Paul B. Romesser; Richard J. Wong; Eric J. Sherman; Heiko Schöder; Nancy Y. Lee

Collaboration


Dive into the Brad Beattie's collaboration.

Top Co-Authors

Avatar

John L. Humm

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Steven M. Larson

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Heiko Schöder

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Pat Zanzonico

NewYork–Presbyterian Hospital

View shared research outputs
Top Co-Authors

Avatar

Adriana D. Corben

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Alexander Bolaender

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Andrew Nagel

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Anna Rodina

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Barry J. Kappel

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Christina Pressl

Memorial Sloan Kettering Cancer Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge