Network


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

Hotspot


Dive into the research topics where Christine A. Iacobuzio-Donahue is active.

Publication


Featured researches published by Christine A. Iacobuzio-Donahue.


Science | 2009

Inhibition of Hedgehog Signaling Enhances Delivery of Chemotherapy in a Mouse Model of Pancreatic Cancer

Kenneth P. Olive; Michael A. Jacobetz; Christian J. Davidson; Aarthi Gopinathan; Dominick J.O. McIntyre; Davina Jean Honess; Basetti Madhu; Mae Goldgraben; Meredith E. Caldwell; David Allard; Kristopher K. Frese; Gina M. DeNicola; Christine Feig; Chelsea Combs; Stephen P. Winter; Heather Ireland-Zecchini; Stefanie Reichelt; William J. Howat; Alex R. Chang; Mousumi Dhara; Lifu Wang; Felix Rückert; Robert Grützmann; Christian Pilarsky; Kamel Izeradjene; Sunil R. Hingorani; Pearl S. Huang; Susan E. Davies; William Plunkett; Merrill J. Egorin

Its All in the Delivery Pancreatic cancer is almost universally associated with a poor prognosis, in part because the tumors are resistant to chemotherapeutic drugs. Working with a mouse tumor model that displays many features of the human disease, Olive et al. (p. 1457, published online 21 May; see the Perspective by Olson and Hanahan) found that the tumors were poorly vascularized, a factor likely to impede drug delivery. Treatment of the mice with the chemotherapeutic drug gemcitabine in combination with a drug that depletes tumor-associated stromal tissue led to an increase in tumor vasculature, enhanced delivery of gemcitabine, and a delay in disease progression. Thus, drugs targeting the tumor stroma may merit investigation as a way to enhance the efficacy of conventional chemotherapy for pancreatic cancer. Pancreatic tumors are unresponsive to chemotherapy because their limited vasculature precludes efficient drug delivery. Pancreatic ductal adenocarcinoma (PDA) is among the most lethal human cancers in part because it is insensitive to many chemotherapeutic drugs. Studying a mouse model of PDA that is refractory to the clinically used drug gemcitabine, we found that the tumors in this model were poorly perfused and poorly vascularized, properties that are shared with human PDA. We tested whether the delivery and efficacy of gemcitabine in the mice could be improved by coadministration of IPI-926, a drug that depletes tumor-associated stromal tissue by inhibition of the Hedgehog cellular signaling pathway. The combination therapy produced a transient increase in intratumoral vascular density and intratumoral concentration of gemcitabine, leading to transient stabilization of disease. Thus, inefficient drug delivery may be an important contributor to chemoresistance in pancreatic cancer.


Nature | 2010

Distant metastasis occurs late during the genetic evolution of pancreatic cancer

Shinichi Yachida; Siân Jones; Ivana Bozic; Tibor Antal; Rebecca J. Leary; Baojin Fu; Mihoko Kamiyama; Ralph H. Hruban; James R. Eshleman; Martin A. Nowak; Victor E. Velculescu; Kenneth W. Kinzler; Bert Vogelstein; Christine A. Iacobuzio-Donahue

Metastasis, the dissemination and growth of neoplastic cells in an organ distinct from that in which they originated, is the most common cause of death in cancer patients. This is particularly true for pancreatic cancers, where most patients are diagnosed with metastatic disease and few show a sustained response to chemotherapy or radiation therapy. Whether the dismal prognosis of patients with pancreatic cancer compared to patients with other types of cancer is a result of late diagnosis or early dissemination of disease to distant organs is not known. Here we rely on data generated by sequencing the genomes of seven pancreatic cancer metastases to evaluate the clonal relationships among primary and metastatic cancers. We find that clonal populations that give rise to distant metastases are represented within the primary carcinoma, but these clones are genetically evolved from the original parental, non-metastatic clone. Thus, genetic heterogeneity of metastases reflects that within the primary carcinoma. A quantitative analysis of the timing of the genetic evolution of pancreatic cancer was performed, indicating at least a decade between the occurrence of the initiating mutation and the birth of the parental, non-metastatic founder cell. At least five more years are required for the acquisition of metastatic ability and patients die an average of two years thereafter. These data provide novel insights into the genetic features underlying pancreatic cancer progression and define a broad time window of opportunity for early detection to prevent deaths from metastatic disease.


Cell | 2011

Massive genomic rearrangement acquired in a single catastrophic event during cancer development

Philip Stephens; Christopher Greenman; Beiyuan Fu; Fengtang Yang; Graham R. Bignell; Laura Mudie; Erin Pleasance; King Wai Lau; David Beare; Lucy Stebbings; Stuart McLaren; Meng-Lay Lin; David J. McBride; Ignacio Varela; Serena Nik-Zainal; Catherine Leroy; Mingming Jia; Andrew Menzies; Adam Butler; Jon Teague; Michael A. Quail; John Burton; Harold Swerdlow; Nigel P. Carter; Laura A. Morsberger; Christine A. Iacobuzio-Donahue; George A. Follows; Anthony R. Green; Adrienne M. Flanagan; Michael R. Stratton

Summary Cancer is driven by somatically acquired point mutations and chromosomal rearrangements, conventionally thought to accumulate gradually over time. Using next-generation sequencing, we characterize a phenomenon, which we term chromothripsis, whereby tens to hundreds of genomic rearrangements occur in a one-off cellular crisis. Rearrangements involving one or a few chromosomes crisscross back and forth across involved regions, generating frequent oscillations between two copy number states. These genomic hallmarks are highly improbable if rearrangements accumulate over time and instead imply that nearly all occur during a single cellular catastrophe. The stamp of chromothripsis can be seen in at least 2%–3% of all cancers, across many subtypes, and is present in ∼25% of bone cancers. We find that one, or indeed more than one, cancer-causing lesion can emerge out of the genomic crisis. This phenomenon has important implications for the origins of genomic remodeling and temporal emergence of cancer. PaperClip


Nature | 2014

A draft map of the human proteome

Min Sik Kim; Sneha M. Pinto; Derese Getnet; Raja Sekhar Nirujogi; Srikanth S. Manda; Raghothama Chaerkady; Dhanashree S. Kelkar; Ruth Isserlin; Shobhit Jain; Joji Kurian Thomas; Babylakshmi Muthusamy; Pamela Leal-Rojas; Praveen Kumar; Nandini A. Sahasrabuddhe; Lavanya Balakrishnan; Jayshree Advani; Bijesh George; Santosh Renuse; Lakshmi Dhevi N. Selvan; Arun H. Patil; Vishalakshi Nanjappa; Aneesha Radhakrishnan; Samarjeet Prasad; Tejaswini Subbannayya; Rajesh Raju; Manish Kumar; Sreelakshmi K. Sreenivasamurthy; Arivusudar Marimuthu; Gajanan Sathe; Sandip Chavan

The availability of human genome sequence has transformed biomedical research over the past decade. However, an equivalent map for the human proteome with direct measurements of proteins and peptides does not exist yet. Here we present a draft map of the human proteome using high-resolution Fourier-transform mass spectrometry. In-depth proteomic profiling of 30 histologically normal human samples, including 17 adult tissues, 7 fetal tissues and 6 purified primary haematopoietic cells, resulted in identification of proteins encoded by 17,294 genes accounting for approximately 84% of the total annotated protein-coding genes in humans. A unique and comprehensive strategy for proteogenomic analysis enabled us to discover a number of novel protein-coding regions, which includes translated pseudogenes, non-coding RNAs and upstream open reading frames. This large human proteome catalogue (available as an interactive web-based resource at http://www.humanproteomemap.org) will complement available human genome and transcriptome data to accelerate biomedical research in health and disease.


Nature | 2010

The patterns and dynamics of genomic instability in metastatic pancreatic cancer

Peter J. Campbell; Shinichi Yachida; Laura Mudie; Philip Stephens; Erin Pleasance; Lucy Stebbings; Laura A. Morsberger; Calli Latimer; Stuart McLaren; Meng-Lay Lin; David J. McBride; Ignacio Varela; Serena Nik-Zainal; Catherine Leroy; Mingming Jia; Andrew Menzies; Adam Butler; Jon Teague; Constance A. Griffin; John Burton; Harold Swerdlow; Michael A. Quail; Michael R. Stratton; Christine A. Iacobuzio-Donahue; P. Andrew Futreal

Pancreatic cancer is an aggressive malignancy with a five-year mortality of 97–98%, usually due to widespread metastatic disease. Previous studies indicate that this disease has a complex genomic landscape, with frequent copy number changes and point mutations, but genomic rearrangements have not been characterized in detail. Despite the clinical importance of metastasis, there remain fundamental questions about the clonal structures of metastatic tumours, including phylogenetic relationships among metastases, the scale of ongoing parallel evolution in metastatic and primary sites, and how the tumour disseminates. Here we harness advances in DNA sequencing to annotate genomic rearrangements in 13 patients with pancreatic cancer and explore clonal relationships among metastases. We find that pancreatic cancer acquires rearrangements indicative of telomere dysfunction and abnormal cell-cycle control, namely dysregulated G1-to-S-phase transition with intact G2–M checkpoint. These initiate amplification of cancer genes and occur predominantly in early cancer development rather than the later stages of the disease. Genomic instability frequently persists after cancer dissemination, resulting in ongoing, parallel and even convergent evolution among different metastases. We find evidence that there is genetic heterogeneity among metastasis-initiating cells, that seeding metastasis may require driver mutations beyond those required for primary tumours, and that phylogenetic trees across metastases show organ-specific branches. These data attest to the richness of genetic variation in cancer, brought about by the tandem forces of genomic instability and evolutionary selection.


Nature | 2011

Oncogene-induced Nrf2 transcription promotes ROS detoxification and tumorigenesis

Gina M. DeNicola; Florian A. Karreth; Timothy J. Humpton; Aarthi Gopinathan; Cong Wei; Kristopher K. Frese; Dipti Mangal; Kenneth H. Yu; Charles J. Yeo; Eric S. Calhoun; Francesca Scrimieri; Jordan M. Winter; Ralph H. Hruban; Christine A. Iacobuzio-Donahue; Scott E. Kern; Ian A. Blair; David A. Tuveson

Reactive oxygen species (ROS) are mutagenic and may thereby promote cancer. Normally, ROS levels are tightly controlled by an inducible antioxidant program that responds to cellular stressors and is predominantly regulated by the transcription factor Nrf2 (also known as Nfe2l2) and its repressor protein Keap1 (refs 2–5). In contrast to the acute physiological regulation of Nrf2, in neoplasia there is evidence for increased basal activation of Nrf2. Indeed, somatic mutations that disrupt the Nrf2–Keap1 interaction to stabilize Nrf2 and increase the constitutive transcription of Nrf2 target genes were recently identified, indicating that enhanced ROS detoxification and additional Nrf2 functions may in fact be pro-tumorigenic. Here, we investigated ROS metabolism in primary murine cells following the expression of endogenous oncogenic alleles of Kras, Braf and Myc, and found that ROS are actively suppressed by these oncogenes. K-RasG12D, B-RafV619E and MycERT2 each increased the transcription of Nrf2 to stably elevate the basal Nrf2 antioxidant program and thereby lower intracellular ROS and confer a more reduced intracellular environment. Oncogene-directed increased expression of Nrf2 is a new mechanism for the activation of the Nrf2 antioxidant program, and is evident in primary cells and tissues of mice expressing K-RasG12D and B-RafV619E, and in human pancreatic cancer. Furthermore, genetic targeting of the Nrf2 pathway impairs K-RasG12D-induced proliferation and tumorigenesis in vivo. Thus, the Nrf2 antioxidant and cellular detoxification program represents a previously unappreciated mediator of oncogenesis.


Nature | 2015

Whole genomes redefine the mutational landscape of pancreatic cancer

Nicola Waddell; Marina Pajic; Ann-Marie Patch; David K. Chang; Karin S. Kassahn; Peter Bailey; Amber L. Johns; David Miller; Katia Nones; Kelly Quek; Michael Quinn; Alan Robertson; Muhammad Z.H. Fadlullah; Timothy J. C. Bruxner; Angelika N. Christ; Ivon Harliwong; Senel Idrisoglu; Suzanne Manning; Craig Nourse; Ehsan Nourbakhsh; Shivangi Wani; Peter J. Wilson; Emma Markham; Nicole Cloonan; Matthew J. Anderson; J. Lynn Fink; Oliver Holmes; Stephen Kazakoff; Conrad Leonard; Felicity Newell

Pancreatic cancer remains one of the most lethal of malignancies and a major health burden. We performed whole-genome sequencing and copy number variation (CNV) analysis of 100 pancreatic ductal adenocarcinomas (PDACs). Chromosomal rearrangements leading to gene disruption were prevalent, affecting genes known to be important in pancreatic cancer (TP53, SMAD4, CDKN2A, ARID1A and ROBO2) and new candidate drivers of pancreatic carcinogenesis (KDM6A and PREX2). Patterns of structural variation (variation in chromosomal structure) classified PDACs into 4 subtypes with potential clinical utility: the subtypes were termed stable, locally rearranged, scattered and unstable. A significant proportion harboured focal amplifications, many of which contained druggable oncogenes (ERBB2, MET, FGFR1, CDK6, PIK3R3 and PIK3CA), but at low individual patient prevalence. Genomic instability co-segregated with inactivation of DNA maintenance genes (BRCA1, BRCA2 or PALB2) and a mutational signature of DNA damage repair deficiency. Of 8 patients who received platinum therapy, 4 of 5 individuals with these measures of defective DNA maintenance responded.


Journal of Clinical Oncology | 2009

DPC4 Gene Status of the Primary Carcinoma Correlates With Patterns of Failure in Patients With Pancreatic Cancer

Christine A. Iacobuzio-Donahue; Baojin Fu; Shinichi Yachida; Mingde Luo; Hisashi Abe; Clark Henderson; Felip Vilardell; Zheng Wang; Jesse Keller; Priya Banerjee; Joseph M. Herman; John L. Cameron; Charles J. Yeo; Marc K. Halushka; James R. Eshleman; Marian Raben; Alison P. Klein; Ralph H. Hruban; Manuel Hidalgo; D. Laheru

PURPOSE Contrary to the extensive data accumulated regarding pancreatic carcinogenesis, the clinical and molecular features characteristic of advanced stage (stage III and IV) disease are unknown. A comprehensive study of pancreatic cancers from patients who have succumbed to their disease has the potential to greatly expand our understanding of the most lethal stage of this disease and identify novel areas for intervention. MATERIALS AND METHODS Rapid autopsies were performed on 76 patients with documented pancreatic cancer. The histologic features of end stage disease were determined and correlated to the stage at initial diagnosis, patterns of failure (locally destructive v metastatic disease) and the status of the KRAS2, TP53, and DPC4 genes. RESULTS At autopsy, 30% of patients died with locally destructive pancreatic cancer, and 70% died with widespread metastatic disease. These divergent patterns of failure found at autopsy (locally destructive v metastatic) were unrelated to clinical stage at initial presentation, treatment history, or histopathologic features. However, Dpc4 immunolabeling status of carcinoma tissues harvested at autopsy, a sensitive marker of DPC4 genetic status, was highly correlated with the presence of widespread metastasis but not with locally destructive tumors (P = .007). CONCLUSION Pancreatic cancers are represented by distinct genetic subtypes with significantly different patterns of failure. Determinations of DPC4 status at initial diagnosis may be of value in stratifying patients into treatment regimens related to local control versus systemic therapy.


Cancer Cell | 2014

Stromal Elements Act to Restrain, Rather Than Support, Pancreatic Ductal Adenocarcinoma

Andrew D. Rhim; Paul Eliezer Oberstein; Dafydd H. Thomas; Emily T. Mirek; Carmine Palermo; Stephen A. Sastra; Erin N. Dekleva; Tyler Saunders; Claudia P. Becerra; Ian W. Tattersall; C. Benedikt Westphalen; Jan Kitajewski; Maite G. Fernandez-Barrena; Martin E. Fernandez-Zapico; Christine A. Iacobuzio-Donahue; Kenneth P. Olive; Ben Z. Stanger

Sonic hedgehog (Shh), a soluble ligand overexpressed by neoplastic cells in pancreatic ductal adenocarcinoma (PDAC), drives formation of a fibroblast-rich desmoplastic stroma. To better understand its role in malignant progression, we deleted Shh in a well-defined mouse model of PDAC. As predicted, Shh-deficient tumors had reduced stromal content. Surprisingly, such tumors were more aggressive and exhibited undifferentiated histology, increased vascularity, and heightened proliferation--features that were fully recapitulated in control mice treated with a Smoothened inhibitor. Furthermore, administration of VEGFR blocking antibody selectively improved survival of Shh-deficient tumors, indicating that Hedgehog-driven stroma suppresses tumor growth in part by restraining tumor angiogenesis. Together, these data demonstrate that some components of the tumor stroma can act to restrain tumor growth.


Cancer Cell | 2003

Notch mediates TGFα-induced changes in epithelial differentiation during pancreatic tumorigenesis

Yoshiharu Miyamoto; Anirban Maitra; Bidyut Ghosh; Ulrich Zechner; Pedram Argani; Christine A. Iacobuzio-Donahue; Virote Sriuranpong; Tatsuya Iso; Ingrid M. Meszoely; Michael S. Wolfe; Ralph H. Hruban; Douglas W. Ball; Roland M. Schmid; Steven D. Leach

Notch signaling regulates cell fate decisions in a wide variety of adult and embryonic tissues. Here we show that Notch pathway components and Notch target genes are upregulated in invasive pancreatic cancer, as well as in pancreatic cancer precursors from both mouse and human. In mouse pancreas, ectopic Notch activation results in accumulation of nestin-positive precursor cells and expansion of metaplastic ductal epithelium, previously identified as a precursor lesion for pancreatic cancer. Notch is also activated as a direct consequence of EGF receptor activation in exocrine pancreas and is required for TGF alpha-induced changes in epithelial differentiation. These findings suggest that Notch mediates the tumor-initiating effects of TG alpha by expanding a population of undifferentiated precursor cells.

Collaboration


Dive into the Christine A. Iacobuzio-Donahue's collaboration.

Top Co-Authors

Avatar

Ralph H. Hruban

Fred Hutchinson Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Anirban Maitra

Johns Hopkins University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Charles J. Yeo

Boston Children's Hospital

View shared research outputs
Top Co-Authors

Avatar

Joseph M. Herman

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Scott E. Kern

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Alvin Makohon-Moore

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge