Network


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

Hotspot


Dive into the research topics where Uthra Balaji is active.

Publication


Featured researches published by Uthra Balaji.


Nature Communications | 2015

Whole-exome sequencing of pancreatic cancer defines genetic diversity and therapeutic targets.

Agnieszka K. Witkiewicz; Elizabeth McMillan; Uthra Balaji; GuemHee Baek; Wan Chi Lin; John C. Mansour; Mehri Mollaee; Kay Uwe Wagner; Prasad Koduru; Adam C. Yopp; Michael A. Choti; Charles J. Yeo; Peter McCue; Michael A. White; Erik S. Knudsen

Pancreatic ductal adenocarcinoma (PDA) has a dismal prognosis and insights into both disease etiology and targeted intervention are needed. A total of 109 micro-dissected PDA cases were subjected to whole-exome sequencing. Microdissection enriches tumour cellularity and enhances mutation calling. Here we show that environmental stress and alterations in DNA repair genes associate with distinct mutation spectra. Copy number alterations target multiple tumour suppressive/oncogenic loci; however, amplification of MYC is uniquely associated with poor outcome and adenosquamous subtype. We identify multiple novel mutated genes in PDA, with select genes harbouring prognostic significance. RBM10 mutations associate with longer survival in spite of histological features of aggressive disease. KRAS mutations are observed in >90% of cases, but codon Q61 alleles are selectively associated with improved survival. Oncogenic BRAF mutations are mutually exclusive with KRAS and define sensitivity to vemurafenib in PDA models. High-frequency alterations in Wnt signalling, chromatin remodelling, Hedgehog signalling, DNA repair and cell cycle processes are observed. Together, these data delineate new genetic diversity of PDA and provide insights into prognostic determinants and therapeutic targets.


Cell Reports | 2016

Metabolic Reprogramming of Pancreatic Cancer Mediated by CDK4/6 Inhibition Elicits Unique Vulnerabilities.

Jorge Franco; Uthra Balaji; Elizaveta Freinkman; Agnieszka K. Witkiewicz; Erik S. Knudsen

Due to loss of p16ink4a in pancreatic ductal adenocarcinoma (PDA), pharmacological suppression of CDK4/6 could represent a potent target for treatment. In PDA models, CDK4/6 inhibition had a variable effect on cell cycle but yielded accumulation of ATP and mitochondria. Pharmacological CDK4/6 inhibitors induce cyclin D1 protein levels; however, RB activation was required and sufficient for mitochondrial accumulation. CDK4/6 inhibition stimulated glycolytic and oxidative metabolism and was associated with an increase in mTORC1 activity. MTOR and MEK inhibitors potently cooperate with CDK4/6 inhibition in eliciting cell-cycle exit. However, MTOR inhibition fully suppressed metabolism and yielded apoptosis and suppression of tumor growth in xenograft models. The metabolic state mediated by CDK4/6 inhibition increases mitochondrial number and reactive oxygen species (ROS). Concordantly, the suppression of ROS scavenging or BCL2 antagonists cooperated with CDK4/6 inhibition. Together, these data define the impact of therapeutics on PDA metabolism and provide strategies for converting cytostatic response to tumor cell killing.


Clinical Cancer Research | 2017

Stratification of Pancreatic Ductal Adenocarcinoma: Combinatorial Genetic, Stromal, and Immunologic Markers

Erik S. Knudsen; Paris Vail; Uthra Balaji; Hoai Ngo; Ihab W. Botros; Vladimir Makarov; Nadeem Riaz; Vinod P. Balachandran; Steven D. Leach; Debrah Thompson; Timothy A. Chan; Agnieszka K. Witkiewicz

Purpose: Pancreatic ductal adenocarcinoma (PDAC) is associated with an immunosuppressive milieu that supports immune system evasion and disease progression. Here, we interrogated genetic, stromal, and immunologic features of PDAC to delineate impact on prognosis and means to more effectively employ immunotherapy. Experimental Design: A cohort of 109 PDAC cases annotated for overall survival was utilized as a primary discovery cohort. Gene expression analysis defined immunologic subtypes of PDAC that were confirmed in the Cancer Genome Atlas dataset. Stromal and metabolic characteristics of PDAC cases were evaluated by histologic analysis and immunostaining. Enumeration of lymphocytes, as well as staining for CD8, FOXP3, CD68, CD163, PDL1, and CTLA4 characterized immune infiltrate. Neoantigens were determined by analysis of whole-exome sequencing data. Random-forest clustering was employed to define multimarker subtypes, with univariate and multivariate analyses interrogating prognostic significance. Results: PDAC cases exhibited distinct stromal phenotypes that were associated with prognosis, glycolytic and hypoxic biomarkers, and immune infiltrate composition. Immune infiltrate was diverse among PDAC cases and enrichment for M2 macrophages and select immune checkpoints regulators were specifically associated with survival. Composite analysis with neoantigen burden, immunologic, and stromal features defined novel subtypes of PDAC that could have bearing on sensitivity to immunologic therapy approaches. In addition, a subtype with low levels of neoantigens and minimal lymphocyte infiltrate was associated with improved overall survival. Conclusions: The mutational burden of PDAC is associated with distinct immunosuppressive mechanisms that are conditioned by the tumor stromal environment. The defined subtypes have significance for utilizing immunotherapy in the treatment of PDAC. Clin Cancer Res; 23(15); 4429–40. ©2017 AACR.


Clinical Cancer Research | 2016

Immunologic and Metabolic Features of Pancreatic Ductal Adenocarcinoma Define Prognostic Subtypes of Disease.

Jack Hutcheson; Uthra Balaji; Matthew R. Porembka; Megan B. Wachsmann; Peter McCue; Erik S. Knudsen; Agnieszka K. Witkiewicz

Purpose: Pancreatic ductal adenocarcinoma (PDA) is associated with an immunosuppressive microenvironment that supports the growth of the malignancy as well as immune system evasion. Here we examine markers of immunosuppression in PDA within the context of the glycolytic tumor microenvironment, their interrelationship with tumor biology and association with overall survival. Experimental Design: We utilized tissue microarrays consisting of 223 PDA patients annotated for clinical stage, tumor size, lymph node involvement, and survival. Expression of CD163, FoxP3, PD-L1, and MCT4 was assessed by IHC and statistical associations were evaluated by univariate and multivariate analysis. Multimarker subtypes were defined by random forest analysis. Mechanistic interactions were evaluated using PDA cell lines and models for myeloid differentiation. Results: PDA exhibits discrete expression of CD163, FoxP3, and PD-L1 with modest individual significance. However, combined low expression of these markers was associated with improved prognosis (P = 0.02). PDA tumor cells altered macrophage phenotype and function, which supported enhanced invasiveness in cell-based models. Lactate efflux mediated by MCT4 was associated with, and required for, the selective conversion of myeloid cells. Correspondingly, MCT4 expression correlated with immune markers in PDA cases, and increased the significance of prognostic subtypes (P = 0.002). Conclusions: There exists a complex interplay between PDA tumor cells and the host immune system wherein immunosuppression is associated with negative outcome. MCT4 expression, representative of the glycolytic state of PDA, contributes to the phenotypic conversion of myeloid cells. Thus, metabolic status of PDA tumors is an important determinant of the immunosuppressive environment. Clin Cancer Res; 22(14); 3606–17. ©2016 AACR.


Hepatology | 2015

Astrocyte elevated gene-1 and c-Myc cooperate to promote hepatocarcinogenesis in mice

Jyoti Srivastava; Ayesha Siddiq; Rachel Gredler; Xue Ning Shen; Devaraja Rajasekaran; Chadia L. Robertson; Mark A. Subler; Jolene J. Windle; Catherine I. Dumur; Nitai D. Mukhopadhyay; Dawn Garcia; Zhao Lai; Yidong Chen; Uthra Balaji; Paul B. Fisher; Devanand Sarkar

Astrocyte elevated gene‐1 (AEG‐1) and c‐Myc are overexpressed in human hepatocellular carcinoma (HCC) functioning as oncogenes. AEG‐1 is transcriptionally regulated by c‐Myc, and AEG‐1 itself induces c‐Myc by activating the Wnt/β‐catenin–signaling pathway. We now document the cooperation of AEG‐1 and c‐Myc in promoting hepatocarcinogenesis by analyzing hepatocyte‐specific transgenic mice expressing either AEG‐1 (albumin [Alb]/AEG‐1), c‐Myc (Alb/c‐Myc), or both (Alb/AEG‐1/c‐Myc). Wild‐type and Alb/AEG‐1 mice did not develop spontaneous HCC. Alb/c‐Myc mice developed spontaneous HCC without distant metastasis, whereas Alb/AEG‐1/c‐Myc mice developed highly aggressive HCC with frank metastasis to the lungs. Induction of carcinogenesis by N‐nitrosodiethylamine significantly accelerated the kinetics of tumor formation in all groups. However, in Alb/AEG‐1/c‐Myc, the effect was markedly pronounced with lung metastasis. In vitro analysis showed that Alb/AEG‐1/c‐Myc hepatocytes acquired increased proliferation and transformative potential with sustained activation of prosurvival and epithelial‐mesenchymal transition–signaling pathways. RNA‐sequencing analysis identified a unique gene signature in livers of Alb/AEG‐1/c‐Myc mice that was not observed when either AEG‐1 or c‐Myc was overexpressed. Specifically, Alb/AEG‐1/c‐Myc mice overexpressed maternally imprinted noncoding RNAs (ncRNAs), such as Rian, Meg‐3, and Mirg, which are implicated in hepatocarcinogenesis. Knocking down these ncRNAs significantly inhibited proliferation and invasion by Alb/AEG‐1/c‐Myc hepatocytes. Conclusion: Our studies reveal a novel cooperative oncogenic effect of AEG‐1 and c‐Myc that might explain the mechanism of aggressive HCC. Alb/AEG‐1/c‐Myc mice provide a useful model to understand the molecular mechanism of cooperation between these two oncogenes and other molecules involved in hepatocarcinogenesis. This model might also be of use for evaluating novel therapeutic strategies targeting HCC. (Hepatology 2015;61:915–929)


Hepatology | 2014

Retinoblastoma protein potentiates the innate immune response in hepatocytes: Significance for hepatocellular carcinoma

Jack Hutcheson; Ryan J. Bourgo; Uthra Balaji; Adam Ertel; Agnieszka K. Witkiewicz; Erik S. Knudsen

Cancers mediated by viral etiology must exhibit deregulated cellular proliferation and evade immune recognition. The role of the retinoblastoma tumor suppressor (RB) pathway, which is lost at relatively high frequency in hepatocellular carcinoma (HCC), has recently been expanded to include the regulation of innate immune responsiveness. In this study we investigated the coordinate impact of RB‐loss on cell cycle control and immune function in the liver. We found that RB depletion in hepatoma cells resulted in a compromised immunological response to multiple stimuli and reduced the potential of these cells to recruit myeloid cells. Viral‐mediated liver‐specific RB deletion in vivo led to the induction of genes associated with proliferation and cell cycle entry as well as the significant attenuation of genes associated with immune function, as evidenced by decreases in cytokine and chemokine expression, leukocyte recruitment, and hepatic inflammation. To determine if these changes in gene expression were instructive in human disease, we compared our liver‐specific RB‐loss gene signature to existing profiles of HCC and found that this signature was associated with disease progression and confers a worse prognosis. Conclusion: Our data confirm that RB participates in the regulation of innate immunity in liver parenchymal cells both in vitro and in vivo and to our knowledge describes the first gene signature associated with HCC that includes both immunoregulatory and proliferative genes and that can also be attributed to the alteration of a single gene in vitro. (Hepatology 2014;60:1231–1240)


Gut | 2018

Pancreatic cancer cell lines as patient-derived avatars: genetic characterisation and functional utility

Erik S. Knudsen; Uthra Balaji; Brian K. Mannakee; Paris Vail; Cody Eslinger; Christopher Moxom; John C. Mansour; Agnieszka K. Witkiewicz

Objective Pancreatic ductal adenocarcinoma (PDAC) is a therapy recalcitrant disease with the worst survival rate of common solid tumours. Preclinical models that accurately reflect the genetic and biological diversity of PDAC will be important for delineating features of tumour biology and therapeutic vulnerabilities. Design 27 primary PDAC tumours were employed for genetic analysis and development of tumour models. Tumour tissue was used for derivation of xenografts and cell lines. Exome sequencing was performed on the originating tumour and developed models. RNA sequencing, histological and functional analyses were employed to determine the relationship of the patient-derived models to clinical presentation of PDAC. Results The cohort employed captured the genetic diversity of PDAC. From most cases, both cell lines and xenograft models were developed. Exome sequencing confirmed preservation of the primary tumour mutations in developed cell lines, which remained stable with extended passaging. The level of genetic conservation in the cell lines was comparable to that observed with patient-derived xenograft (PDX) models. Unlike historically established PDAC cancer cell lines, patient-derived models recapitulated the histological architecture of the primary tumour and exhibited metastatic spread similar to that observed clinically. Detailed genetic analyses of tumours and derived models revealed features of ex vivo evolution and the clonal architecture of PDAC. Functional analysis was used to elucidate therapeutic vulnerabilities of relevance to treatment of PDAC. Conclusions These data illustrate that with the appropriate methods it is possible to develop cell lines that maintain genetic features of PDAC. Such models serve as important substrates for analysing the significance of genetic variants and create a unique biorepository of annotated cell lines and xenografts that were established simultaneously from same primary tumour. These models can be used to infer genetic and empirically determined therapeutic sensitivities that would be germane to the patient.


Oncotarget | 2016

Unique metabolic features of pancreatic cancer stroma: relevance to the tumor compartment, prognosis, and invasive potential.

Erik S. Knudsen; Uthra Balaji; Elizaveta Freinkman; Peter McCue; Agnieszka K. Witkiewicz

Pancreatic ductal adenocarcinoma (PDAC) has a dismal prognosis. The aggressiveness and therapeutic recalcitrance of this malignancy has been attributed to multiple factors including the influence of an active desmoplastic stroma. How the stromal microenvironment of PDAC contributes to the fatal nature of this disease is not well defined. In the analysis of clinical specimens, we observed diverse expression of the hypoxic marker carbonic anhydrase IX and the lactate transporter MCT4 in the stromal compartment. These stromal features were associated with the epithelial to mesenchymal phenotype in PDAC tumor cells, and with shorter patient survival. Cultured cancer-associated fibroblasts (CAFs) derived from primary PDAC exhibited a high basal level of hypoxia inducible factor 1a (HIF1α) that was both required and sufficient to modulate the expression of MCT4. This event was associated with increased transcription and protein synthesis of HIF1α in CAFs relative to PDAC cell lines, while surprisingly the protein turnover rate was equivalent. CAFs utilized glucose predominantly for glycolytic intermediates, whereas glutamine was the preferred metabolite for the TCA cycle. Unlike PDAC cell lines, CAFs were resistant to glucose withdrawal but sensitive to glutamine depletion. Consistent with the lack of reliance on glucose, CAFs could survive the acute depletion of MCT4. In co-culture and xenograft studies CAFs stimulated the invasive potential and metastatic spread of PDAC cell lines through a mechanism dependent on HIF1α and MCT4. Together, these data indicate that stromal metabolic features influence PDAC tumor cells to promote invasiveness and metastatic potential and associate with poor outcome in patients with PDAC.


Clinical Cancer Research | 2014

Systematically defining single gene determinants of response to neoadjuvant chemotherapy reveals specific biomarkers

Agnieszka K. Witkiewicz; Uthra Balaji; Erik S. Knudsen

Purpose: We sought to systematically define determinants of the response to neoadjuvant chemotherapy to elucidate predictive biomarkers for breast cancer. Experimental Design: An unbiased systematic analysis was performed in multiple independent datasets to define genes predictive of complete pathologic response (pCR) following treatment with neoadjuvant chemotherapy. These genes were interrogated across estrogen receptor (ER)–positive and ER-negative breast cancer and those in common across three different treatment regimens were analyzed for optimal predictive power. Subsequent validation was performed on independent cohorts by gene expression and IHC analyses. Results: Genes that were highly associated with the response to neoadjuvant chemotherapy in breast cancer were readily defined using a computational method ranking individual genes by their respective ROC. Such predictive genes of the response to taxane-associated therapies were strongly enriched for cell-cycle control processes in both ER-positive and ER-negative breast cancer and correlated with pCR. However, other genes that were specifically associated with residual disease were also identified under other treatment conditions. Using the intersection between treatment groups, nine genes were identified that harbored strong predictive power in multiple contexts and validation cohort. In particular, the nuclear oncogene DEK was strongly associated with pCR, whereas the cell surface protein BCAM was strongly associated with residual disease. By IHC staining, these markers exhibited potent predictive power that remained significant in multivariate analysis. Conclusion: Systematic computational approaches can define key genes that will be able to predict the response to chemotherapy across multiple treatment modalities yielding a small collection of biomarkers that can be readily deployed by IHC analyses. Clin Cancer Res; 20(18); 4837–48. ©2014 AACR.


Bioinformatics | 2018

Sensitive and specific post-call filtering of genetic variants in xenograft and primary tumors

Brian K. Mannakee; Uthra Balaji; Agnieszka K. Witkiewicz; Ryan N. Gutenkunst; Erik S. Knudsen

Motivation: Tumor genome sequencing offers great promise for guiding research and therapy, but spurious variant calls can arise from multiple sources. Mouse contamination can generate many spurious calls when sequencing patient‐derived xenografts. Paralogous genome sequences can also generate spurious calls when sequencing any tumor. We developed a BLAST‐based algorithm, Mouse And Paralog EXterminator (MAPEX), to identify and filter out spurious calls from both these sources. Results: When calling variants from xenografts, MAPEX has similar sensitivity and specificity to more complex algorithms. When applied to any tumor, MAPEX also automatically flags calls that potentially arise from paralogous sequences. Our implementation, mapexr, runs quickly and easily on a desktop computer. MAPEX is thus a useful addition to almost any pipeline for calling genetic variants in tumors. Availability and implementation: The mapexr package for R is available at https://github.com/bmannakee/mapexr under the MIT license. Contact: [email protected] or [email protected] or [email protected] Supplementary information: Supplementary data are available at Bioinformatics online.

Collaboration


Dive into the Uthra Balaji's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Peter McCue

Thomas Jefferson University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Cody Eslinger

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

Elizabeth McMillan

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

Elizaveta Freinkman

Massachusetts Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Jack Hutcheson

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

John C. Mansour

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

Jorge Franco

University of Texas Southwestern Medical Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge