Network


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

Hotspot


Dive into the research topics where Alexis Christoforides is active.

Publication


Featured researches published by Alexis Christoforides.


Blood | 2012

Whole-genome sequencing of multiple myeloma from diagnosis to plasma cell leukemia reveals genomic initiating events, evolution, and clonal tides

Jan B. Egan; Chang Xin Shi; Waibhav Tembe; Alexis Christoforides; Ahmet Kurdoglu; Shripad Sinari; Sumit Middha; Yan W. Asmann; Jessica Schmidt; Esteban Braggio; Jonathan J. Keats; Rafael Fonseca; P. Leif Bergsagel; David Craig; John D. Carpten; A. Keith Stewart

The longitudinal evolution of a myeloma genome from diagnosis to plasma cell leukemia has not previously been reported. We used whole-genome sequencing (WGS) on 4 purified tumor samples and patient germline DNA drawn over a 5-year period in a t(4;14) multiple myeloma patient. Tumor samples were acquired at diagnosis, first relapse, second relapse, and end-stage secondary plasma cell leukemia (sPCL). In addition to the t(4;14), all tumor time points also shared 10 common single-nucleotide variants (SNVs) on WGS comprising shared initiating events. Interestingly, we observed genomic sequence variants that waxed and waned with time in progressive tumors, suggesting the presence of multiple independent, yet related, clones at diagnosis that rose and fell in dominance. Five newly acquired SNVs, including truncating mutations of RB1 and ZKSCAN3, were observed only in the final sPCL sample suggesting leukemic transformation events. This longitudinal WGS characterization of the natural history of a high-risk myeloma patient demonstrated tumor heterogeneity at diagnosis with shifting dominance of tumor clones over time and has also identified potential mutations contributing to myelomagenesis as well as transformation from myeloma to overt extramedullary disease such as sPCL.


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.


Molecular Cancer Therapeutics | 2013

Genome and Transcriptome Sequencing in Prospective Metastatic Triple-Negative Breast Cancer Uncovers Therapeutic Vulnerabilities

David Craig; Joyce O'Shaughnessy; Jeffrey Kiefer; Jessica Aldrich; Shripad Sinari; Tracy M. Moses; Shukmei Wong; Jennifer Dinh; Alexis Christoforides; Joanne L. Blum; Cristi L Aitelli; Cynthia Osborne; Tyler Izatt; Ahmet Kurdoglu; Angela Baker; Julie Koeman; Catalin Barbacioru; Onur Sakarya; Francisco M. De La Vega; Asim Siddiqui; Linh Hoang; Paul R. Billings; Bodour Salhia; Anthony W. Tolcher; Jeffrey M. Trent; Spyro Mousses; Daniel D. Von Hoff; John D. Carpten

Triple-negative breast cancer (TNBC) is characterized by the absence of expression of estrogen receptor, progesterone receptor, and HER-2. Thirty percent of patients recur after first-line treatment, and metastatic TNBC (mTNBC) has a poor prognosis with median survival of one year. Here, we present initial analyses of whole genome and transcriptome sequencing data from 14 prospective mTNBC. We have cataloged the collection of somatic genomic alterations in these advanced tumors, particularly those that may inform targeted therapies. Genes mutated in multiple tumors included TP53, LRP1B, HERC1, CDH5, RB1, and NF1. Notable genes involved in focal structural events were CTNNA1, PTEN, FBXW7, BRCA2, WT1, FGFR1, KRAS, HRAS, ARAF, BRAF, and PGCP. Homozygous deletion of CTNNA1 was detected in 2 of 6 African Americans. RNA sequencing revealed consistent overexpression of the FOXM1 gene when tumor gene expression was compared with nonmalignant breast samples. Using an outlier analysis of gene expression comparing one cancer with all the others, we detected expression patterns unique to each patients tumor. Integrative DNA/RNA analysis provided evidence for deregulation of mutated genes, including the monoallelic expression of TP53 mutations. Finally, molecular alterations in several cancers supported targeted therapeutic intervention on clinical trials with known inhibitors, particularly for alterations in the RAS/RAF/MEK/ERK and PI3K/AKT/mTOR pathways. In conclusion, whole genome and transcriptome profiling of mTNBC have provided insights into somatic events occurring in this difficult to treat cancer. These genomic data have guided patients to investigational treatment trials and provide hypotheses for future trials in this irremediable cancer. Mol Cancer Ther; 12(1); 104–16. ©2012 AACR.


BMC Genomics | 2013

Identification of somatic mutations in cancer through Bayesian-based analysis of sequenced genome pairs.

Alexis Christoforides; John D. Carpten; Glen J. Weiss; Michael J. Demeure; Daniel D. Von Hoff; David Craig

BackgroundThe field of cancer genomics has rapidly adopted next-generation sequencing (NGS) in order to study and characterize malignant tumors with unprecedented resolution. In particular for cancer, one is often trying to identify somatic mutations – changes specific to a tumor and not within an individual’s germline. However, false positive and false negative detections often result from lack of sufficient variant evidence, contamination of the biopsy by stromal tissue, sequencing errors, and the erroneous classification of germline variation as tumor-specific.ResultsWe have developed a generalized Bayesian analysis framework for matched tumor/normal samples with the purpose of identifying tumor-specific alterations such as single nucleotide mutations, small insertions/deletions, and structural variation. We describe our methodology, and discuss its application to other types of paired-tissue analysis such as the detection of loss of heterozygosity as well as allelic imbalance. We also demonstrate the high level of sensitivity and specificity in discovering simulated somatic mutations, for various combinations of a) genomic coverage and b) emulated heterogeneity.ConclusionWe present a Java-based implementation of our methods named Seurat, which is made available for free academic use. We have demonstrated and reported on the discovery of different types of somatic change by applying Seurat to an experimentally-derived cancer dataset using our methods; and have discussed considerations and practices regarding the accurate detection of somatic events in cancer genomes. Seurat is available at https://sites.google.com/site/seuratsomatic.


Emerging Infectious Diseases | 2011

Next-Generation Sequencing of Coccidioides immitis Isolated during Cluster Investigation

David M. Engelthaler; Tom Chiller; James A. Schupp; Joshua Colvin; Stephen M. Beckstrom-Sternberg; Elizabeth M. Driebe; Tracy Moses; Waibhav Tembe; Shripad Sinari; James S. Beckstrom-Sternberg; Alexis Christoforides; John V. Pearson; John D. Carpten; Paul Keim; Ashley Peterson; Dawn Terashita; S. Arunmozhi Balajee

Next-generation sequencing enables use of whole-genome sequence typing (WGST) as a viable and discriminatory tool for genotyping and molecular epidemiologic analysis. We used WGST to confirm the linkage of a cluster of Coccidioides immitis isolates from 3 patients who received organ transplants from a single donor who later had positive test results for coccidioidomycosis. Isolates from the 3 patients were nearly genetically identical (a total of 3 single-nucleotide polymorphisms identified among them), thereby demonstrating direct descent of the 3 isolates from an original isolate. We used WGST to demonstrate the genotypic relatedness of C. immitis isolates that were also epidemiologically linked. Thus, WGST offers unique benefits to public health for investigation of clusters considered to be linked to a single source.


PLOS ONE | 2012

Paired Tumor and Normal Whole Genome Sequencing of Metastatic Olfactory Neuroblastoma

Glen J. Weiss; Winnie S. Liang; Tyler Izatt; Shilpi Arora; Irene Cherni; Robert N. Raju; Galen Hostetter; Ahmet Kurdoglu; Alexis Christoforides; Shripad Sinari; Angela Baker; Raghu Metpally; Waibhav Tembe; Lori Phillips; Daniel D. Von Hoff; David Craig; John D. Carpten

Background Olfactory neuroblastoma (ONB) is a rare cancer of the sinonasal tract with little molecular characterization. We performed whole genome sequencing (WGS) on paired normal and tumor DNA from a patient with metastatic-ONB to identify the somatic alterations that might be drivers of tumorigenesis and/or metastatic progression. Methodology/Principal Findings Genomic DNA was isolated from fresh frozen tissue from a metastatic lesion and whole blood, followed by WGS at >30X depth, alignment and mapping, and mutation analyses. Sanger sequencing was used to confirm selected mutations. Sixty-two somatic short nucleotide variants (SNVs) and five deletions were identified inside coding regions, each causing a non-synonymous DNA sequence change. We selected seven SNVs and validated them by Sanger sequencing. In the metastatic ONB samples collected several months prior to WGS, all seven mutations were present. However, in the original surgical resection specimen (prior to evidence of metastatic disease), mutations in KDR, MYC, SIN3B, and NLRC4 genes were not present, suggesting that these were acquired with disease progression and/or as a result of post-treatment effects. Conclusions/Significance This work provides insight into the evolution of ONB cancer cells and provides a window into the more complex factors, including tumor clonality and multiple driver mutations.


BMC Medical Genomics | 2014

Whole genome sequencing reveals potential targets for therapy in patients with refractory KRAS mutated metastatic colorectal cancer

Vijayalakshmi Shanmugam; Ramesh K. Ramanathan; Nicole A. Lavender; Shripad Sinari; Manpreet Chadha; Winnie S. Liang; Ahmet Kurdoglu; Tyler Izatt; Alexis Christoforides; Hollie Benson; Lori Phillips; Angela Baker; Christopher Murray; Galen Hostetter; Daniel D. Von Hoff; David Craig; John D. Carpten

BackgroundThe outcome of patients with metastatic colorectal carcinoma (mCRC) following first line therapy is poor, with median survival of less than one year. The purpose of this study was to identify candidate therapeutically targetable somatic events in mCRC patient samples by whole genome sequencing (WGS), so as to obtain targeted treatment strategies for individual patients.MethodsFour patients were recruited, all of whom had received > 2 prior therapy regimens. Percutaneous needle biopsies of metastases were performed with whole blood collection for the extraction of constitutional DNA. One tumor was not included in this study as the quality of tumor tissue was not sufficient for further analysis. WGS was performed using Illumina paired end chemistry on HiSeq2000 sequencing systems, which yielded coverage of greater than 30X for all samples. NGS data were processed and analyzed to detect somatic genomic alterations including point mutations, indels, copy number alterations, translocations and rearrangements.ResultsAll 3 tumor samples had KRAS mutations, while 2 tumors contained mutations in the APC gene and the PIK3CA gene. Although we did not identify a TCF7L2-VTI1A translocation, we did detect a TCF7L2 mutation in one tumor. Among the other interesting mutated genes was INPPL1, an important gene involved in PI3 kinase signaling. Functional studies demonstrated that inhibition of INPPL1 reduced growth of CRC cells, suggesting that INPPL1 may promote growth in CRC.ConclusionsOur study further supports potential molecularly defined therapeutic contexts that might provide insights into treatment strategies for refractory mCRC. New insights into the role of INPPL1 in colon tumor cell growth have also been identified. Continued development of appropriate targeted agents towards specific events may be warranted to help improve outcomes in CRC.


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 Gynecological Cancer | 2014

Simultaneous characterization of somatic events and HPV-18 integration in a metastatic cervical carcinoma patient using DNA and RNA sequencing.

Winnie S. Liang; Jessica Aldrich; Sara Nasser; Ahmet Kurdoglu; Lori Phillips; Rebecca Reiman; Jacquelyn McDonald; Tyler Izatt; Alexis Christoforides; Angela Baker; C. Craig; Jan B. Egan; Dana M. Chase; John H. Farley; Alan H. Bryce; A. Keith Stewart; Mitesh J. Borad; John D. Carpten; David W. Craig; Bradley J. Monk

Objective Integration of carcinogenic human papillomaviruses (HPVs) into the host genome is a significant tumorigenic factor in specific cancers including cervical carcinoma. Although major strides have been made with respect to HPV diagnosis and prevention, identification and development of efficacious treatments for cervical cancer patients remains a goal and thus requires additional detailed characterization of both somatic events and HPV integration. Given this need, the goal of this study was to use the next generation sequencing to simultaneously evaluate somatic alterations and expression changes in a patient’s cervical squamous carcinoma lesion metastatic to the lung and to detect and analyze HPV infection in the same sample. Materials and Methods We performed tumor and normal exome, tumor and normal shallow whole-genome sequencing, and RNA sequencing of the patient’s lung metastasis. Results We generated over 1.2 billion mapped reads and identified 130 somatic point mutations and indels, 21 genic translocations, 16 coding regions demonstrating copy number changes, and over 36 genes demonstrating altered expression in the tumor (corrected P < 0.05). Sequencing also revealed the HPV type 18 (HPV-18) integration in the metastasis. Using both DNA and RNA reads, we pinpointed 3 major events indicating HPV-18 integration into an intronic region of chromosome 6p25.1 in the patient’s tumor and validated these events with Sanger sequencing. This integration site has not been reported for HPV-18. Conclusions We demonstrate that DNA and RNA sequencing can be used to concurrently characterize somatic alterations and expression changes in a biopsy and delineate HPV integration at base resolution in cervical cancer. Further sequencing will allow us to better understand the molecular basis of cervical cancer pathogenesis.


pacific symposium on biocomputing | 2014

An integrated framework for reporting clinically relevant biomarkers from paired tumor/normal genomic and transcriptomic sequencing data in support of clinical trials in personalized medicine

Sara Nasser; Ahmet A Kurdolgu; Tyler Izatt; Jessica Aldrich; Megan Russell; Alexis Christoforides; Wiabhav Tembe; Jeffery A Keifer; Jason J. Corneveaux; Sara A. Byron; Karen M. Forman; Clarice Zuccaro; Jonathan J. Keats; Patricia LoRusso; John D. Carpten; Jeffrey M. Trent; David Craig

The ability to rapidly sequence the tumor and germline DNA of an individual holds the eventual promise of revolutionizing our ability to match targeted therapies to tumors harboring the associated genetic biomarkers. Analyzing high throughput genomic data consisting of millions of base pairs and discovering alterations in clinically actionable genes in a structured and real time manner is at the crux of personalized testing. This requires a computational architecture that can monitor and track a system within a regulated environment as terabytes of data are reduced to a small number of therapeutically relevant variants, delivered as a diagnostic laboratory developed test. These high complexity assays require data structures that enable real-time and retrospective ad-hoc analysis, with a capability of updating to keep up with the rapidly changing genomic and therapeutic options, all under a regulated environment that is relevant under both CMS and FDA depending on application. We describe a flexible computational framework that uses a paired tumor/normal sample allowing for complete analysis and reporting in approximately 24 hours, providing identification of single nucleotide changes, small insertions and deletions, chromosomal rearrangements, gene fusions and gene expression with positive predictive values over 90%. In this paper we present the challenges in integrating clinical, genomic and annotation databases to provide interpreted draft reports which we utilize within ongoing clinical research protocols. We demonstrate the need to retire from existing performance measurements of accuracy and specificity and measure metrics that are meaningful to a genomic diagnostic environment. This paper presents a three-tier infrastructure that is currently being used to analyze an individual genome and provide available therapeutic options via a clinical report. Our framework utilizes a non-relational variant-centric database that is scaleable to a large amount of data and addresses the challenges and limitations of a relational database system. Our system is continuously monitored via multiple trackers each catering differently to the diversity of users involved in this process. These trackers designed in analytics web-app framework provide status updates for an individual sample accurate to a few minutes. In this paper, we also present our outcome delivery process that is designed and delivered adhering to the standards defined by various regulation agencies involved in clinical genomic testing.

Collaboration


Dive into the Alexis Christoforides's collaboration.

Top Co-Authors

Avatar

John D. Carpten

University of Southern California

View shared research outputs
Top Co-Authors

Avatar

Ahmet Kurdoglu

Translational Genomics Research Institute

View shared research outputs
Top Co-Authors

Avatar

David Craig

Translational Genomics Research Institute

View shared research outputs
Top Co-Authors

Avatar

Shripad Sinari

Translational Genomics Research Institute

View shared research outputs
Top Co-Authors

Avatar

Angela Baker

Translational Genomics Research Institute

View shared research outputs
Top Co-Authors

Avatar

Tyler Izatt

Translational Genomics Research Institute

View shared research outputs
Top Co-Authors

Avatar

Winnie S. Liang

Translational Genomics Research Institute

View shared research outputs
Top Co-Authors

Avatar

Daniel D. Von Hoff

Translational Genomics Research Institute

View shared research outputs
Top Co-Authors

Avatar

Jessica Aldrich

Translational Genomics Research Institute

View shared research outputs
Top Co-Authors

Avatar

Lori Phillips

Translational Genomics Research Institute

View shared research outputs
Researchain Logo
Decentralizing Knowledge