Network


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

Hotspot


Dive into the research topics where Agda Karina Eterovic is active.

Publication


Featured researches published by Agda Karina Eterovic.


Blood | 2017

Ph-like acute lymphoblastic leukemia: a high-risk subtype in adults

Nitin Jain; Kathryn G. Roberts; Elias Jabbour; Keyur P. Patel; Agda Karina Eterovic; Ken Chen; Patrick A. Zweidler-McKay; Xinyan Lu; Gloria L. Fawcett; Sa A. Wang; Sergej Konoplev; Richard C. Harvey; I-Ming Chen; Debbie Payne-Turner; Marcus B. Valentine; Deborah A. Thomas; Guillermo Garcia-Manero; Farhad Ravandi; Jorge Cortes; Steven M. Kornblau; Susan O'Brien; Sherry Pierce; Jeffrey L. Jorgensen; Kenna R. Mills Shaw; Cheryl L. Willman; Charles G. Mullighan; Hagop M. Kantarjian; Marina Konopleva

Philadelphia chromosome (Ph)-like acute lymphoblastic leukemia (ALL) is a high-risk subtype of ALL in children. There are conflicting data on the incidence and prognosis of Ph-like ALL in adults. Patients with newly diagnosed B-cell ALL (B-ALL) who received frontline chemotherapy at MD Anderson Cancer Center underwent gene expression profiling of leukemic cells. Of 148 patients, 33.1% had Ph-like, 31.1% had Ph+, and 35.8% had other B-ALL subtypes (B-other). Within the Ph-like ALL cohort, 61% had cytokine receptor-like factor 2 (CRLF2) overexpression. Patients with Ph-like ALL had significantly worse overall survival (OS), and event-free survival compared with B-other with a 5-year survival of 23% (vs 59% for B-other, P = .006). Sixty-eight percent of patients with Ph-like ALL were of Hispanic ethnicity. The following were associated with inferior OS on multivariable analysis: age (hazard ratio [HR], 3.299; P < .001), white blood cell count (HR, 1.910; P = .017), platelet count (HR, 7.437; P = .005), and Ph-like ALL (HR, 1.818; P = .03). Next-generation sequencing of the CRLF2+ group identified mutations in the JAK-STAT and Ras pathway in 85% of patients, and 20% had a CRLF2 mutation. Within the CRLF2+ group, JAK2 mutation was associated with inferior outcomes. Our findings show high frequency of Ph-like ALL in adults, an increased frequency of Ph-like ALL in adults of Hispanic ethnicity, significantly inferior outcomes of adult patients with Ph-like ALL, and significantly worse outcomes in the CRLF2+ subset of Ph-like ALL. Novel strategies are needed to improve the outcome of these patients.


Clinical Chemistry | 2015

Clinical Actionability Enhanced through Deep Targeted Sequencing of Solid Tumors

Ken Chen; Funda Meric-Bernstam; Hao Zhao; Qingxiu Zhang; Nader Ezzeddine; Lin Ya Tang; Yuan Qi; Yong Mao; Tenghui Chen; Zechen Chong; Wanding Zhou; Xiaofeng Zheng; Amber Johnson; Kenneth D. Aldape; Mark Routbort; Rajyalakshmi Luthra; Scott Kopetz; Michael A. Davies; John F. de Groot; Stacy L. Moulder; Ravi Vinod; Carol J. Farhangfar; Kenna Mills Shaw; John Mendelsohn; Gordon B. Mills; Agda Karina Eterovic

BACKGROUND Further advances of targeted cancer therapy require comprehensive in-depth profiling of somatic mutations that are present in subpopulations of tumor cells in a clinical tumor sample. However, it is unclear to what extent such intratumor heterogeneity is present and whether it may affect clinical decision-making. To study this question, we established a deep targeted sequencing platform to identify potentially actionable DNA alterations in tumor samples. METHODS We assayed 515 formalin-fixed paraffin-embedded (FFPE) tumor samples and matched germline DNA (475 patients) from 11 disease sites by capturing and sequencing all the exons in 201 cancer-related genes. Mutations, indels, and copy number data were reported. RESULTS We obtained a 1000-fold mean sequencing depth and identified 4794 nonsynonymous mutations in the samples analyzed, of which 15.2% were present at <10% allele frequency. Most of these low level mutations occurred at known oncogenic hotspots and are likely functional. Identifying low level mutations improved identification of mutations in actionable genes in 118 (24.84%) patients, among which 47 (9.8%) otherwise would have been unactionable. In addition, acquiring ultrahigh depth also ensured a low false discovery rate (<2.2%) from FFPE samples. CONCLUSIONS Our results were as accurate as a commercially available CLIA-compliant hotspot panel but allowed the detection of a higher number of mutations in actionable genes. Our study reveals the critical importance of acquiring and utilizing high sequencing depth in profiling clinical tumor samples and presents a very useful platform for implementing routine sequencing in a cancer care institution.


Cancer Letters | 2015

Gene mutations in primary tumors and corresponding patient-derived xenografts derived from non-small cell lung cancer

Chuncheng Hao; Li Wang; Shaohua Peng; Hongyu Li; Jing Hu; Xiao Huang; Wei Liu; Hui Zhang; Shuhong Wu; Apar Pataer; John V. Heymach; Agda Karina Eterovic; Qingxiu Zhang; Kenna R. Shaw; Ken Chen; Andrew Futreal; Michael Wang; Wayne L. Hofstetter; Reza J. Mehran; David C. Rice; Jack A. Roth; Boris Sepesi; Stephen G. Swisher; Ara A. Vaporciyan; Garrett L. Walsh; Faye M. Johnson; Bingliang Fang

Molecular annotated patient-derived xenograft (PDX) models are useful for the preclinical investigation of anticancer drugs and individualized anticancer therapy. We established 23 PDXs from 88 surgical specimens of lung cancer patients and determined gene mutations in these PDXs and their paired primary tumors by ultradeep exome sequencing on 202 cancer-related genes. The numbers of primary tumors with deleterious mutations in TP53, KRAS, PI3KCA, ALK, STK11, and EGFR were 43.5%, 21.7%, 17.4%, 17.4%, 13.0%, and 8.7%, respectively. Other genes with deleterious mutations in ≥3 (13.0%) primary tumors were MLL3, SETD2, ATM, ARID1A, CRIPAK, HGF, BAI3, EP300, KDR, PDGRRA and RUNX1. Of 315 mutations detected in the primary tumors, 293 (93%) were also detected in their corresponding PDXs, indicating that PDXs have the capacity to recapitulate the mutations in primary tumors. Nevertheless, a substantial number of mutations had higher allele frequencies in the PDXs than in the primary tumors, or were not detectable in the primary tumor, suggesting the possibility of tumor cell enrichment in PDXs or heterogeneity in the primary tumors. The molecularly annotated PDXs generated from this study could be useful for future translational studies.


BMC Medicine | 2016

Novel algorithmic approach predicts tumor mutation load and correlates with immunotherapy clinical outcomes using a defined gene mutation set

Jason Roszik; Lauren E. Haydu; Kenneth R. Hess; Junna Oba; Aron Joon; Alan Siroy; Tatiana Karpinets; Francesco C. Stingo; Veera Baladandayuthapani; Michael T. Tetzlaff; Jennifer A. Wargo; Ken Chen; Marie Andrée Forget; Cara Haymaker; Jie Qing Chen; Funda Meric-Bernstam; Agda Karina Eterovic; Kenna R. Shaw; Gordon B. Mills; Jeffrey E. Gershenwald; Laszlo Radvanyi; Patrick Hwu; P. Andrew Futreal; Don L. Gibbons; Alexander J. Lazar; Chantale Bernatchez; Michael A. Davies; Scott E. Woodman

BackgroundWhile clinical outcomes following immunotherapy have shown an association with tumor mutation load using whole exome sequencing (WES), its clinical applicability is currently limited by cost and bioinformatics requirements.MethodsWe developed a method to accurately derive the predicted total mutation load (PTML) within individual tumors from a small set of genes that can be used in clinical next generation sequencing (NGS) panels. PTML was derived from the actual total mutation load (ATML) of 575 distinct melanoma and lung cancer samples and validated using independent melanoma (n = 312) and lung cancer (n = 217) cohorts. The correlation of PTML status with clinical outcome, following distinct immunotherapies, was assessed using the Kaplan–Meier method.ResultsPTML (derived from 170 genes) was highly correlated with ATML in cutaneous melanoma and lung adenocarcinoma validation cohorts (R2 = 0.73 and R2 = 0.82, respectively). PTML was strongly associated with clinical outcome to ipilimumab (anti-CTLA-4, three cohorts) and adoptive T-cell therapy (1 cohort) clinical outcome in melanoma. Clinical benefit from pembrolizumab (anti-PD-1) in lung cancer was also shown to significantly correlate with PTML status (log rank P value < 0.05 in all cohorts).ConclusionsThe approach of using small NGS gene panels, already applied to guide employment of targeted therapies, may have utility in the personalized use of immunotherapy in cancer.


Human Pathology | 2014

Molecular Characterization of Gallbladder Cancer using Somatic Mutation Profiling

Milind Javle; Asif Rashid; Chaitanya Churi; Siddhartha Kar; Mingxin Zuo; Agda Karina Eterovic; Graciela M. Nogueras-Gonzalez; Filip Janku; Rachna T. Shroff; Thomas A. Aloia; Jean Nicolas Vauthey; Steven A. Curley; Gordon B. Mills; Iván Roa

Gallbladder cancer is relatively uncommon with high incidence in certain geographic locations, including Latin America, East and South Asia and Eastern Europe. Molecular characterization of this disease has been limited and targeted therapy options for advanced disease remain an open area of investigation. In the present study, surgical pathology obtained from resected gallbladder cancer cases (n=72) was examined for the presence of targetable, somatic mutations. All cases were formalin-fixed and paraffin-embedded (FFPE). Two approaches were used: a) mass spectroscopy-based profiling for 159 point (‘hot-spot’) mutations in 33 genes commonly involved in solid tumors and b) next-generation sequencing (NGS) platform that examined the complete coding sequence of in 182 cancer-related genes. Fifty-seven cases were analyzed for hotspot mutations and 15 for NGS. Fourteen hotspot mutations were identified in nine cases. Of these, KRAS mutation was significantly associated with poor survival on multivariate analysis. Other targetable mutations included PIK3CA (N=2) and ALK (N=1). On NGS, 26 mutations were noted in 15 cases. P53 and PI3 kinase pathway (STK11, RICTOR,TSC2) mutations were common. One case had FGF10 amplification while another had FGF3-TACC gene fusion, not previously described in gallbladder cancer. In conclusion, somatic mutation profiling using archival FFPE samples from gallbladder cancer is feasible. NGS, in particular may be a useful platform for identifying novel mutations for targeted therapy.


Bioinformatics | 2014

Bias from removing read duplication in ultra-deep sequencing experiments

Wanding Zhou; Tenghui Chen; Hao Zhao; Agda Karina Eterovic; Funda Meric-Bernstam; Gordon B. Mills; Ken Chen

MOTIVATION Identifying subclonal mutations and their implications requires accurate estimation of mutant allele fractions from possibly duplicated sequencing reads. Removing duplicate reads assumes that polymerase chain reaction amplification from library constructions is the primary source. The alternative-sampling coincidence from DNA fragmentation-has not been systematically investigated. RESULTS With sufficiently high-sequencing depth, sampling-induced read duplication is non-negligible, and removing duplicate reads can overcorrect read counts, causing systemic biases in variant allele fraction and copy number variation estimations. Minimal overcorrection occurs when duplicate reads are identified accounting for their mate reads, inserts are of a variety of lengths and samples are sequenced in separate batches. We investigate sampling-induced read duplication in deep sequencing data with 500× to 2000× duplicates-removed sequence coverage. We provide a quantitative solution to overcorrection and guidance for effective designs of deep sequencing platforms that facilitate accurate estimation of variant allele fraction and copy number variation. AVAILABILITY AND IMPLEMENTATION A Python implementation is freely available at https://bitbucket.org/wanding/duprecover/overview CONTACT: : [email protected], [email protected] Supplementary information: Supplementary data are available at Bioinformatics online.


Science Translational Medicine | 2017

Rational combination therapy with PARP and MEK inhibitors capitalizes on therapeutic liabilities in RAS mutant cancers

Chaoyang Sun; Yong Fang; Jun Yin; Jian Chen; Zhenlin Ju; Dong Zhang; Xiaohua Chen; Christopher P. Vellano; Kang Jin Jeong; Patrick Kwok Shing Ng; Agda Karina Eterovic; Neil H. Bhola; Yiling Lu; Shannon N. Westin; Jennifer R. Grandis; Shiaw Yih Lin; Kenneth L. Scott; Guang Peng; Joan S. Brugge; Gordon B. Mills

The combination of PARP and MEK inhibitors is effective against RAS and RAF mutant tumors. No escape for KRAS mutant tumors Alterations in one of the RAS proteins, such as KRAS, are among the most common oncogenic mutations and also among the most difficult to treat. RAS mutant tumors are usually resistant to PARP inhibitors, one of the newest classes of anticancer therapeutics, and many other chemotherapy types. However, Sun et al. have discovered that inhibition of MEK or ERK (proteins in the RAS pathway) can reverse PARP inhibitor resistance in KRAS mutant tumors. MEK and PARP inhibitors are clinically approved drugs that provide a readily translatable therapeutic combination, and the authors have demonstrated its effectiveness in mouse models of aggressive tumors such as ovarian and pancreatic cancer. Mutant RAS has remained recalcitrant to targeted therapy efforts. We demonstrate that combined treatment with poly(adenosine diphosphate–ribose) polymerase (PARP) inhibitors and mitogen-activated protein kinase (MAPK) kinase (MEK) inhibitors evokes unanticipated, synergistic cytotoxic effects in vitro and in vivo in multiple RAS mutant tumor models across tumor lineages where RAS mutations are prevalent. The effects of PARP and MEK inhibitor combinations are independent of BRCA1/2 and p53 mutation status, suggesting that the synergistic activity is likely to be generalizable. Synergistic activity of PARP and MEK inhibitor combinations in RAS mutant tumors is associated with (i) induction of BIM-mediated apoptosis, (ii) decrease in expression of components of the homologous recombination DNA repair pathway, (iii) decrease in homologous recombination DNA damage repair capacity, (iv) decrease in DNA damage checkpoint activity, (v) increase in PARP inhibitor–induced DNA damage, (vi) decrease in vascularity that could increase PARP inhibitor efficacy by inducing hypoxia, and (vii) elevated PARP1 protein, which increases trapping activity of PARP inhibitors. Mechanistically, enforced expression of FOXO3a, which is a target of the RAS/MAPK pathway, was sufficient to recapitulate the functional consequences of MEK inhibitors including synergy with PARP inhibitors. Thus, the ability of mutant RAS to suppress FOXO3a and its reversal by MEK inhibitors accounts, at least in part, for the synergy of PARP and MEK inhibitors in RAS mutant tumors. The rational combination of PARP and MEK inhibitors warrants clinical investigation in patients with RAS mutant tumors where there are few effective therapeutic options.


Pigment Cell & Melanoma Research | 2015

Genetic analysis of the 'uveal melanoma' C918 cell line reveals atypical BRAF and common KRAS mutations and single tandem repeat profile identical to the cutaneous melanoma C8161 cell line

Xiaoxing Yu; Grazia Ambrosini; Jason Roszik; Agda Karina Eterovic; Katherine Stempke-Hale; Elisabeth A. Seftor; Chandrani Chattopadhyay; Elizabeth A. Grimm; Richard D. Carvajal; Mary J.C. Hendrix; F. Stephen Hodi; Gary K. Schwartz; Scott E. Woodman

To take out a personal subscription, please click here More information about Pigment Cell & Melanoma Research at www.pigment.org Genetic analysis of the ‘uveal melanoma’ C918 cell line reveals atypical BRAF and common KRAS mutations and single tandem repeat profile identical to the cutaneous melanoma C8161 cell line Xiaoxing Yu, Grazia Ambrosini, Jason Roszik, Agda Karina Eterovic, Katherine Stempke-Hale, Elisabeth A. Seftor, Chandrani Chattopadhyay, Elizabeth Grimm, Richard D. Carvajal, Mary J. C. Hendrix, F. Stephen Hodi, Gary K. Schwartz and Scott E. Woodman


Oncotarget | 2016

The degree of intratumor mutational heterogeneity varies by primary tumor sub-site.

Levi G. Ledgerwood; Dhruv Kumar; Agda Karina Eterovic; Jo Wick; Ken Chen; Hao Zhao; Loubna Tazi; Pradip Manna; Spencer Kerley; Radhika Joshi; Lin Wang; Simion I. Chiosea; James D. Garnett; Terance T. Tsue; Jeremy Chien; Gordon B. Mills; Jennifer R. Grandis; Sufi M. Thomas

In an era where mutational profiles inform treatment options, it is critical to know the extent to which tumor biopsies represent the molecular profile of the primary and metastatic tumor. Head and neck squamous cell carcinoma (HNSCC) arise primarily in the mucosal lining of oral cavity and oropharynx. Despite aggressive therapy the 5-year survival rate is at 50%. The primary objective of this study is to characterize the degree of intratumor mutational heterogeneity in HNSCC. We used multi-region sequencing of paired primary and metastatic tumor DNA of 24 spatially distinct samples from seven patients with HNSCC of larynx, floor of the mouth (FOM) or oral tongue. Full length, in-depth sequencing of 202 genes implicated in cancer was carried out. Larynx and FOM tumors had more than 69.2% unique SNVs between the paired primary and metastatic lesions. In contrast, the oral tongue HNSCC had only 33.3% unique SNVs across multiple sites. In addition, HNSCC of the oral tongue had fewer mutations than larynx and FOM tumors. These findings were validated on the Affymetrix whole genome 6.0 array platform and were consistent with data from The Cancer Genome Atlas (TCGA). This is the first report demonstrating differences in mutational heterogeneity varying by subsite in HNSCC. The heterogeneity within laryngeal tumor specimens may lead to an underestimation of the genetic abnormalities within tumors and may foster resistance to standard treatment protocols. These findings are relevant to investigators and clinicians developing personalized cancer treatments based on identification of specific mutations in tumor biopsies.


PLOS ONE | 2015

Ability to Generate Patient-Derived Breast Cancer Xenografts Is Enhanced in Chemoresistant Disease and Predicts Poor Patient Outcomes.

Priscilla F. McAuliffe; Kurt W. Evans; Argun Akcakanat; Ken Chen; Xiaofeng Zheng; Hao Zhao; Agda Karina Eterovic; Takafumi Sangai; Ashley M. Holder; Chandeshwar Sharma; Huiqin Chen; Kim Anh Do; Emily Tarco; Mihai Gagea; Aysegul A. Sahin; Asha S. Multani; Dalliah M. Black; Elizabeth A. Mittendorf; Isabelle Bedrosian; Gordon B. Mills; Ana M. Gonzalez-Angulo; Funda Meric-Bernstam

Background Breast cancer patients who are resistant to neoadjuvant chemotherapy (NeoCT) have a poor prognosis. There is a pressing need to develop in vivo models of chemo resistant tumors to test novel therapeutics. We hypothesized that patient-derived breast cancer xenografts (BCXs) from chemo- naïve and chemotherapy-exposed tumors can provide high fidelity in vivo models for chemoresistant breast cancers. Methods Patient tumors and BCXs were characterized with short tandem repeat DNA fingerprinting, reverse phase protein arrays, molecular inversion probe arrays, and next generation sequencing. Results Forty-eight breast cancers (24 post-chemotherapy, 24 chemo-naïve) were implanted and 13 BCXs were established (27%). BCX engraftment was higher in TNBC compared to hormone-receptor positive cancer (53.8% vs. 15.6%, p = 0.02), in tumors from patients who received NeoCT (41.7% vs. 8.3%, p = 0.02), and in patients who had progressive disease on NeoCT (85.7% vs. 29.4%, p = 0.02). Twelve patients developed metastases after surgery; in five, BCXs developed before distant relapse. Patients whose tumors developed BCXs had a lower recurrence-free survival (p = 0.015) and overall survival (p<0.001). Genomic losses and gains could be detected in the BCX, and three models demonstrated a transformation to induce mouse tumors. However, overall, somatic mutation profiles including potential drivers were maintained upon implantation and serial passaging. One BCX model was cultured in vitro and re-implanted, maintaining its genomic profile. Conclusions BCXs can be established from clinically aggressive breast cancers, especially in TNBC patients with poor response to NeoCT. Future studies will determine the potential of in vivo models for identification of genotype-phenotype correlations and individualization of treatment.

Collaboration


Dive into the Agda Karina Eterovic's collaboration.

Top Co-Authors

Avatar

Gordon B. Mills

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Ken Chen

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Funda Meric-Bernstam

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Xiaofeng Zheng

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Hao Zhao

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

John Mendelsohn

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Ana M. Gonzalez-Angulo

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Argun Akcakanat

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Kurt W. Evans

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Lauren Brusco

University of Texas MD Anderson Cancer Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge