Network


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

Hotspot


Dive into the research topics where Amanda Blackford is active.

Publication


Featured researches published by Amanda Blackford.


Nature Immunology | 2005

Egr-2 and Egr-3 are negative regulators of T cell activation.

Meredith Safford; Samuel L. Collins; Michael A. Lutz; Amy Allen; Ching Tai Huang; Jeanne Kowalski; Amanda Blackford; Maureen R. Horton; Charles G. Drake; Ronald H. Schwartz; Jonathan D. Powell

T cell receptor engagement in the absence of proper accessory signals leads to T cell anergy. E3 ligases are involved in maintaining the anergic state. However, the specific molecules responsible for the induction of anergy have yet to be elucidated. Using microarray analysis we have identified here early growth response gene 2 (Egr-2) and Egr-3 as key negative regulators of T cell activation. Overexpression of Egr2 and Egr3 was associated with an increase in the E3 ubiquitin ligase Cbl-b and inhibition of T cell activation. Conversely, T cells from Egr3−/− mice had lower expression of Cbl-b and were resistant to in vivo peptide-induced tolerance. These data support the idea that Egr-2 and Egr-3 are involved in promoting a T cell receptor–induced negative regulatory genetic program.


Cancer Research | 2008

RNAi-Mediated Silencing of Nuclear Factor Erythroid-2–Related Factor 2 Gene Expression in Non–Small Cell Lung Cancer Inhibits Tumor Growth and Increases Efficacy of Chemotherapy

Anju Singh; Swetlana Boldin-Adamsky; Rajesh K. Thimmulappa; Srikanta Kumar Rath; Hagit Ashush; Jonathan Coulter; Amanda Blackford; Steven N. Goodman; Fred Bunz; Walter H. Watson; Edward Gabrielson; Elena Feinstein; Shyam Biswal

Nuclear factor erythroid-2-related factor 2 (Nrf2) is a redox-sensitive transcription factor that regulates the expression of electrophile and xenobiotic detoxification enzymes and efflux proteins, which confer cytoprotection against oxidative stress and apoptosis in normal cells. Loss of function mutations in the Nrf2 inhibitor, Kelch-like ECH-associated protein (Keap1), results in constitutive activation of Nrf2 function in non-small cell lung cancer. In this study, we show that constitutive activation of Nrf2 in lung cancer cells promotes tumorigenicity and contributes to chemoresistance by up-regulation of glutathione, thioredoxin, and the drug efflux pathways involved in detoxification of electrophiles and broad spectrum of drugs. RNAi-mediated reduction of Nrf2 expression in lung cancer cells induces generation of reactive oxygen species, suppresses tumor growth, and results in increased sensitivity to chemotherapeutic drug-induced cell death in vitro and in vivo. Inhibiting Nrf2 expression using naked siRNA duplexes in combination with carboplatin significantly inhibits tumor growth in a subcutaneous model of lung cancer. Thus, targeting Nrf2 activity in lung cancers, particularly those with Keap1 mutations, could be a promising strategy to inhibit tumor growth and circumvent chemoresistance.


Nature Genetics | 2013

Integrated genomic analyses identify ARID1A and ARID1B alterations in the childhood cancer neuroblastoma

Mark Sausen; Rebecca J. Leary; Siân Jones; Jian Wu; C. Patrick Reynolds; Xueyuan Liu; Amanda Blackford; Giovanni Parmigiani; Luis A. Diaz; Nickolas Papadopoulos; Bert Vogelstein; Kenneth W. Kinzler; Victor E. Velculescu; Michael D. Hogarty

Neuroblastomas are tumors of peripheral sympathetic neurons and are the most common solid tumor in children. To determine the genetic basis for neuroblastoma, we performed whole-genome sequencing (6 cases), exome sequencing (16 cases), genome-wide rearrangement analyses (32 cases) and targeted analyses of specific genomic loci (40 cases) using massively parallel sequencing. On average, each tumor had 19 somatic alterations in coding genes (range of 3–70). Among genes not previously known to be involved in neuroblastoma, chromosomal deletions and sequence alterations of the chromatin-remodeling genes ARID1A and ARID1B were identified in 8 of 71 tumors (11%) and were associated with early treatment failure and decreased survival. Using tumor-specific structural alterations, we developed an approach to identify rearranged DNA fragments in sera, providing personalized biomarkers for minimal residual disease detection and monitoring. These results highlight the dysregulation of chromatin remodeling in pediatric tumorigenesis and provide new approaches for the management of patients with neuroblastoma.


Clinical Cancer Research | 2009

SMAD4 Gene Mutations Are Associated with Poor Prognosis in Pancreatic Cancer

Amanda Blackford; Oscar K. Serrano; Christopher L. Wolfgang; Giovanni Parmigiani; Siân Jones; Xiaosong Zhang; D. Williams Parsons; Jimmy Lin; Rebecca J. Leary; James R. Eshleman; Michael Goggins; Elizabeth M. Jaffee; Christine A. Iacobuzio-Donahue; Anirban Maitra; John L. Cameron; Kelly Olino; Richard D. Schulick; Jordan M. Winter; Joseph M. Herman; Daniel A. Laheru; Alison P. Klein; Bert Vogelstein; Kenneth W. Kinzler; Victor E. Velculescu; Ralph H. Hruban

Purpose: Recently, the majority of protein coding genes were sequenced in a collection of pancreatic cancers, providing an unprecedented opportunity to identify genetic markers of prognosis for patients with adenocarcinoma of the pancreas. Experimental Design: We previously sequenced more than 750 million base pairs of DNA from 23,219 transcripts in a series of 24 adenocarcinomas of the pancreas. In addition, 39 genes that were mutated in more than one of these 24 cancers were sequenced in a separate panel of 90 well-characterized adenocarcinomas of the pancreas. Of these 114 patients, 89 underwent pancreaticoduodenectomy, and the somatic mutations in these cancers were correlated with patient outcome. Results: When adjusted for age, lymph node status, margin status, and tumor size, SMAD4 gene inactivation was significantly associated with shorter overall survival (hazard ratio, 1.92; 95% confidence interval, 1.20-3.05; P = 0.006). Patients with SMAD4 gene inactivation survived a median of 11.5 months, compared with 14.2 months for patients without SMAD4 inactivation. By contrast, mutations in CDKN2A or TP53 or the presence of multiple (≥4) mutations or homozygous deletions among the 39 most frequently mutated genes were not associated with survival. Conclusions:SMAD4 gene inactivation is associated with poorer prognosis in patients with surgically resected adenocarcinoma of the pancreas.


Gastroenterology | 2015

A Combination of Molecular Markers and Clinical Features Improve the Classification of Pancreatic Cysts

Simeon Springer; Yuxuan Wang; Marco Dal Molin; David L. Masica; Yuchen Jiao; Isaac Kinde; Amanda Blackford; Siva P. Raman; Christopher L. Wolfgang; Tyler Tomita; Noushin Niknafs; Christopher Douville; Janine Ptak; Lisa Dobbyn; Peter J. Allen; David S. Klimstra; Mark A. Schattner; C. Max Schmidt; Michele T. Yip-Schneider; Oscar W. Cummings; Randall E. Brand; Herbert J. Zeh; Aatur D. Singhi; Aldo Scarpa; Roberto Salvia; Giuseppe Malleo; Giuseppe Zamboni; Massimo Falconi; Jin Young Jang; Sun Whe Kim

BACKGROUND & AIMS The management of pancreatic cysts poses challenges to both patients and their physicians. We investigated whether a combination of molecular markers and clinical information could improve the classification of pancreatic cysts and management of patients. METHODS We performed a multi-center, retrospective study of 130 patients with resected pancreatic cystic neoplasms (12 serous cystadenomas, 10 solid pseudopapillary neoplasms, 12 mucinous cystic neoplasms, and 96 intraductal papillary mucinous neoplasms). Cyst fluid was analyzed to identify subtle mutations in genes known to be mutated in pancreatic cysts (BRAF, CDKN2A, CTNNB1, GNAS, KRAS, NRAS, PIK3CA, RNF43, SMAD4, TP53, and VHL); to identify loss of heterozygozity at CDKN2A, RNF43, SMAD4, TP53, and VHL tumor suppressor loci; and to identify aneuploidy. The analyses were performed using specialized technologies for implementing and interpreting massively parallel sequencing data acquisition. An algorithm was used to select markers that could classify cyst type and grade. The accuracy of the molecular markers was compared with that of clinical markers and a combination of molecular and clinical markers. RESULTS We identified molecular markers and clinical features that classified cyst type with 90%-100% sensitivity and 92%-98% specificity. The molecular marker panel correctly identified 67 of the 74 patients who did not require surgery and could, therefore, reduce the number of unnecessary operations by 91%. CONCLUSIONS We identified a panel of molecular markers and clinical features that show promise for the accurate classification of cystic neoplasms of the pancreas and identification of cysts that require surgery.


Journal of Clinical Oncology | 2008

Preventive Care for Colorectal Cancer Survivors: A 5-Year Longitudinal Study

Claire F. Snyder; Craig C. Earle; Robert J. Herbert; Bridget A. Neville; Amanda Blackford; Kevin D. Frick

PURPOSE To explore the mix of physician specialties that long-term survivors visit and how the mix relates to preventive care. PATIENTS AND METHODS Using the Surveillance, Epidemiology, and End Results-Medicare database, we conducted a retrospective, longitudinal study of stage I to III Medicare fee-for-service colorectal cancer patients diagnosed in 1997. We examined physician visits and preventive care each year for 5 years, starting 366 days postdiagnosis, and how preventive service receipt related to the physician mix seen: primary care provider (PCP) only, oncologist only, both, or neither. RESULTS A total of 1,541 patients met the eligibility criteria (mean age, 76; 43% male; 85% white). During 5 years, PCP visits increased from a mean of 4.2 to 4.7 (P < .0001), and oncology visits decreased from 1.3 to 0.5 (P < .0001). Survivor care by PCPs only increased from 44% to 62%, whereas shared care by PCPs and oncologists dropped from 37% to 21% (P < .0001). Survivors who saw both PCPs and oncologists were most likely to receive influenza vaccination, mammograms, and cervical cancer screening; survivors who saw PCPs only were most likely to receive cholesterol screening and bone densitometry. Higher socioeconomic status was associated with increased influenza vaccination, mammograms, and cervical cancer screening (P < .05). Over time, there was a decrease in mammography and cervical cancer screening and an increase in influenza vaccination (P < .05). CONCLUSION As oncologists become less involved in survivor care, cancer-related screening decreases significantly. These results support the need for survivorship care plans that explicitly outline the roles of PCPs and oncologists in sharing care for cancer survivors, and how these roles may change over time.


The Journal of Nuclear Medicine | 2015

18F-DCFBC PET/CT for PSMA-Based Detection and Characterization of Primary Prostate Cancer

Steven P. Rowe; Kenneth L. Gage; Sheila Faraj; Katarzyna J. Macura; Toby C. Cornish; Nilda Gonzalez-Roibon; Gunes Guner; Enrico Munari; Alan W. Partin; Christian P. Pavlovich; Misop Han; H. Ballentine Carter; Trinity J. Bivalacqua; Amanda Blackford; Daniel P. Holt; Robert F. Dannals; George J. Netto; Martin Lodge; Ronnie C. Mease; Martin G. Pomper; Steve Cho

We previously demonstrated the ability to detect metastatic prostate cancer using N-[N-[(S)-1,3-dicarboxypropyl]carbamoyl]-4-18F-fluorobenzyl-l-cysteine (18F-DCFBC), a low-molecular-weight radiotracer that targets the prostate-specific membrane antigen (PSMA). PSMA has been shown to be associated with higher Gleason grade and more aggressive disease. An imaging biomarker able to detect clinically significant high-grade primary prostate cancer reliably would address an unmet clinical need by allowing for risk-adapted patient management. Methods: We enrolled 13 patients with primary prostate cancer who were imaged with 18F-DCFBC PET before scheduled prostatectomy, with 12 of these patients also undergoing pelvic prostate MR imaging. Prostate 18F-DCFBC PET was correlated with MR imaging and histologic and immunohistochemical analysis on a prostate-segment (12 regions) and dominant-lesion basis. There were no incidental extraprostatic findings on PET suggestive of metastatic disease. Results: MR imaging was more sensitive than 18F-DCFBC PET for detection of primary prostate cancer on a per-segment (sensitivities of up to 0.17 and 0.39 for PET and MR imaging, respectively) and per-dominant-lesion analysis (sensitivities of 0.46 and 0.92 for PET and MR imaging, respectively). However, 18F-DCFBC PET was more specific than MR imaging by per-segment analysis (specificities of 0.96 and 0.89 for PET and MR imaging for corresponding sensitivity, respectively) and specific for detection of high-grade lesions (Gleason 8 and 9) greater than 1.0 mL in size (4/4 of these patients positive by PET). 18F-DCFBC uptake in tumors was positively correlated with Gleason score (ρ = 0.64; PSMA expression, ρ = 0.47; and prostate-specific antigen, ρ = 0.52). There was significantly lower 18F-DCFBC uptake in benign prostatic hypertrophy than primary tumors (median maximum standardized uptake value, 2.2 vs. 3.5; P = 0.004). Conclusion: Although the sensitivity of 18F-DCFBC for primary prostate cancer was less than MR imaging, 18F-DCFBC PET was able to detect the more clinically significant high-grade and larger-volume tumors (Gleason score 8 and 9) with higher specificity than MR imaging. In particular, there was relatively low 18F-DCFBC PET uptake in benign prostatic hypertrophy lesions, compared with cancer in the prostate, which may allow for more specific detection of primary prostate cancer by 18F-DCFBC PET. This study demonstrates the utility of PSMA-based PET, which may be used in conjunction with MR imaging to identify clinically significant prostate cancer.


Cancer Research | 2009

Genetic mutations associated with Cigarette smoking in pancreatic cancer

Amanda Blackford; Giovanni Parmigiani; Thomas W. Kensler; Christopher L. Wolfgang; Siân Jones; Xiaosong Zhang; D. Willams Parsons; Jimmy Lin; Rebecca J. Leary; James R. Eshleman; Michael Goggins; Elizabeth M. Jaffee; Christine A. Iacobuzio-Donahue; Anirban Maitra; Alison P. Klein; John L. Cameron; Kelly Olino; Richard D. Schulick; Jordan M. Winter; Bert Vogelstein; Victor E. Velculescu; Kenneth W. Kinzler; Ralph H. Hruban

Cigarette smoking doubles the risk of pancreatic cancer, and smoking accounts for 20% to 25% of pancreatic cancers. The recent sequencing of the pancreatic cancer genome provides an unprecedented opportunity to identify mutational patterns associated with smoking. We previously sequenced >750 million bp DNA from 23,219 transcripts in 24 adenocarcinomas of the pancreas (discovery screen). In this previous study, the 39 genes that were mutated more than once in the discovery screen were sequenced in an additional 90 adenocarcinomas of the pancreas (validation screen). Here, we compared the somatic mutations in the cancers obtained from individuals who ever smoked cigarettes (n = 64) to the somatic mutations in the cancers obtained from individuals who never smoked cigarettes (n = 50). When adjusted for age and gender, analyses of the discovery screen revealed significantly more nonsynonymous mutations in the carcinomas obtained from ever smokers (mean, 53.1 mutations per tumor; SD, 27.9) than in the carcinomas obtained from never smokers (mean, 38.5; SD, 11.1; P = 0.04). The difference between smokers and nonsmokers was not driven by mutations in known driver genes in pancreatic cancer (KRAS, TP53, CDKN2A/p16, and SMAD4), but instead was predominantly observed in genes mutated at lower frequency. No differences were observed in mutations in carcinomas from the head versus tail of the gland. Pancreatic carcinomas from cigarette smokers harbor more mutations than do carcinomas from never smokers. The types and patterns of these mutations provide insight into the mechanisms by which cigarette smoking causes pancreatic cancer.


Nature Genetics | 2015

Common variation at 2p13.3, 3q29, 7p13 and 17q25.1 associated with susceptibility to pancreatic cancer.

Erica J. Childs; Evelina Mocci; Daniele Campa; Paige M. Bracci; Steven Gallinger; Michael Goggins; Donghui Li; Rachel E. Neale; Sara H. Olson; Ghislaine Scelo; Laufey Amundadottir; William R. Bamlet; Maarten F. Bijlsma; Amanda Blackford; Michael Borges; Paul Brennan; Hermann Brenner; H. Bas Bueno-de-Mesquita; Federico Canzian; Gabriele Capurso; Giulia Martina Cavestro; Kari G. Chaffee; Stephen J. Chanock; Sean P. Cleary; Michelle Cotterchio; Lenka Foretova; Charles S. Fuchs; Niccola Funel; Maria Gazouli; Manal Hassan

Pancreatic cancer is the fourth leading cause of cancer death in the developed world. Both inherited high-penetrance mutations in BRCA2 (ref. 2), ATM, PALB2 (ref. 4), BRCA1 (ref. 5), STK11 (ref. 6), CDKN2A and mismatch-repair genes and low-penetrance loci are associated with increased risk. To identify new risk loci, we performed a genome-wide association study on 9,925 pancreatic cancer cases and 11,569 controls, including 4,164 newly genotyped cases and 3,792 controls in 9 studies from North America, Central Europe and Australia. We identified three newly associated regions: 17q25.1 (LINC00673, rs11655237, odds ratio (OR) = 1.26, 95% confidence interval (CI) = 1.19–1.34, P = 1.42 × 10−14), 7p13 (SUGCT, rs17688601, OR = 0.88, 95% CI = 0.84–0.92, P = 1.41 × 10−8) and 3q29 (TP63, rs9854771, OR = 0.89, 95% CI = 0.85–0.93, P = 2.35 × 10−8). We detected significant association at 2p13.3 (ETAA1, rs1486134, OR = 1.14, 95% CI = 1.09–1.19, P = 3.36 × 10−9), a region with previous suggestive evidence in Han Chinese. We replicated previously reported associations at 9q34.2 (ABO), 13q22.1 (KLF5), 5p15.33 (TERT and CLPTM1), 13q12.2 (PDX1), 1q32.1 (NR5A2), 7q32.3 (LINC-PINT), 16q23.1 (BCAR1) and 22q12.1 (ZNRF3). Our study identifies new loci associated with pancreatic cancer risk.


Oncologist | 2013

Repurposing Itraconazole as a Treatment for Advanced Prostate Cancer: A Noncomparative Randomized Phase II Trial in Men With Metastatic Castration-Resistant Prostate Cancer

Emmanuel S. Antonarakis; Elisabeth I. Heath; David C. Smith; Dana E. Rathkopf; Amanda Blackford; Daniel C. Danila; Serina King; Anja Frost; A. Seun Ajiboye; Ming Zhao; Janet Mendonca; Sushant Kachhap; Michelle A. Rudek; Michael A. Carducci

BACKGROUND The antifungal drug itraconazole inhibits angiogenesis and Hedgehog signaling and delays tumor growth in murine prostate cancer xenograft models. We conducted a noncomparative, randomized, phase II study evaluating the antitumor efficacy of two doses of oral itraconazole in men with metastatic prostate cancer. PATIENTS AND METHODS We randomly assigned 46 men with chemotherapy-naïve metastatic castration-resistant prostate cancer (CRPC) to receive low-dose (200 mg/day) or high-dose (600 mg/day) itraconazole until disease progression or unacceptable toxicity. The primary endpoint was the prostate-specific antigen (PSA) progression-free survival (PPFS) rate at 24 weeks; a 45% success rate in either arm was prespecified as constituting clinical significance. Secondary endpoints included the progression-free survival (PFS) rate and PSA response rate (Prostate Cancer Working Group criteria). Exploratory outcomes included circulating tumor cell (CTC) enumeration, serum androgen measurements, as well as pharmacokinetic and pharmacodynamic analyses. RESULTS The high-dose arm enrolled to completion (n = 29), but the low-dose arm closed early (n = 17) because of a prespecified futility rule. The PPFS rates at 24 weeks were 11.8% in the low-dose arm and 48.0% in the high-dose arm. The median PFS times were 11.9 weeks and 35.9 weeks, respectively. PSA response rates were 0% and 14.3%, respectively. In addition, itraconazole had favorable effects on CTC counts, and it suppressed Hedgehog signaling in skin biopsy samples. Itraconazole did not reduce serum testosterone or dehydroepiandrostenedione sulfate levels. Common toxicities included fatigue, nausea, anorexia, rash, and a syndrome of hypokalemia, hypertension, and edema. CONCLUSION High-dose itraconazole (600 mg/day) has modest antitumor activity in men with metastatic CRPC that is not mediated by testosterone suppression.

Collaboration


Dive into the Amanda Blackford's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Judith E. Karp

Johns Hopkins University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ralph H. Hruban

Johns Hopkins University School of Medicine

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Joseph M. Herman

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Christopher L. Wolfgang

Johns Hopkins University School of Medicine

View shared research outputs
Top Co-Authors

Avatar

Kevin D. Frick

Johns Hopkins University

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge