Network


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

Hotspot


Dive into the research topics where Guilherme Cantuaria is active.

Publication


Featured researches published by Guilherme Cantuaria.


Cell Death and Disease | 2014

Centrosome-declustering drugs mediate a two-pronged attack on interphase and mitosis in supercentrosomal cancer cells

Vaishali Pannu; Pcg Rida; Betul Celik; Ravi Chakra Turaga; Angela Ogden; Guilherme Cantuaria; Jay Gopalakrishnan; Ritu Aneja

Classical anti-mitotic drugs have failed to translate their preclinical efficacy into clinical response in human trials. Their clinical failure has challenged the notion that tumor cells divide frequently at rates comparable to those of cancer cells in vitro and in xenograft models. Given the preponderance of interphase cells in clinical tumors, we asked whether targeting amplified centrosomes, which cancer cells carefully preserve in a tightly clustered conformation throughout interphase, presents a superior chemotherapeutic strategy that sabotages interphase-specific cellular activities, such as migration. Herein we have utilized supercentrosomal N1E-115 murine neuroblastoma cells as a test-bed to study interphase centrosome declustering induced by putative declustering agents, such as Reduced-9-bromonoscapine (RedBr-Nos), Griseofulvin and PJ-34. We found tight ‘supercentrosomal’ clusters in the interphase and mitosis of ~80% of patients’ tumor cells with excess centrosomes. RedBr-Nos was the strongest declustering agent with a declustering index of 0.36 and completely dispersed interphase centrosome clusters in N1E-115 cells. Interphase centrosome declustering caused inhibition of neurite formation, impairment of cell polarization and Golgi organization, disrupted cellular protrusions and focal adhesion contacts—factors that are crucial prerequisites for directional migration. Thus our data illustrate an interphase-specific potential anti-migratory role of centrosome-declustering agents in addition to their previously acknowledged ability to induce spindle multipolarity and mitotic catastrophe. Centrosome-declustering agents counter centrosome clustering to inhibit directional cell migration in interphase cells and set up multipolar mitotic catastrophe, suggesting that disbanding the nuclear–centrosome–Golgi axis is a potential anti-metastasis strategy.


PLOS ONE | 2014

Modulation of Cytochrome P450 Metabolism and Transport across Intestinal Epithelial Barrier by Ginger Biophenolics

Rao Mukkavilli; Sushma R. Gundala; Chunhua Yang; Shashikiran Donthamsetty; Guilherme Cantuaria; Gajanan R. Jadhav; Subrahmanyam Vangala; Michelle D. Reid; Ritu Aneja

Natural and complementary therapies in conjunction with mainstream cancer care are steadily gaining popularity. Ginger extract (GE) confers significant health-promoting benefits owing to complex additive and/or synergistic interactions between its bioactive constituents. Recently, we showed that preservation of natural “milieu” confers superior anticancer activity on GE over its constituent phytochemicals, 6-gingerol (6G), 8-gingerol (8G), 10-gingerol (10G) and 6-shogaol (6S), through enterohepatic recirculation. Here we further evaluate and compare the effects of GE and its major bioactive constituents on cytochrome P450 (CYP) enzyme activity in human liver microsomes by monitoring metabolites of CYP-specific substrates using LC/MS/MS detection methods. Our data demonstrate that individual gingerols are potent inhibitors of CYP isozymes, whereas GE exhibits a much higher half-maximal inhibition value, indicating no possible herb-drug interactions. However, GEs inhibition of CYP1A2 and CYP2C8 reflects additive interactions among the constituents. In addition, studies performed to evaluate transporter-mediated intestinal efflux using Caco-2 cells revealed that GE and its phenolics are not substrates of P-glycoprotein (Pgp). Intriguingly, however, 10G and 6S were not detected in the receiver compartment, indicating possible biotransformation across the Caco-2 monolayer. These data strengthen the notion that an interplay of complex interactions among ginger phytochemicals when fed as whole extract dictates its bioactivity highlighting the importance of consuming whole foods over single agents. Our study substantiates the need for an in-depth analysis of hepatic biotransformation events and distribution profiles of GE and its active phenolics for the design of safe regimens.


Journal of Ovarian Research | 2014

KIFCI, a novel putative prognostic biomarker for ovarian adenocarcinomas: delineating protein interaction networks and signaling circuitries.

Shrikant Pawar; Shashikiran Donthamsetty; Vaishali Pannu; Padmashree C.G. Rida; Angela Ogden; Nathan J. Bowen; Remus Osan; Guilherme Cantuaria; Ritu Aneja

BackgroundAmplified centrosomes in cancers are recently garnering a lot of attention as an emerging hub of diagnostic, prognostic and therapeutic targets. Ovarian adenocarcinomas commonly harbor supernumerary centrosomes that drive chromosomal instability. A centrosome clustering molecule, KIFC1, is indispensable for the viability of extra centrosome-bearing cancer cells, and may underlie progression of ovarian cancers.MethodsCentrosome amplification in low- and high- grade serous ovarian adenocarcinomas was quantitated employing confocal imaging. KIFC1 expression was analyzed in ovarian tumors using publically-available databases. Associated grade, stage and clinical information from these databases were plotted for KIFC1 gene expression values. Furthermore, interactions and functional annotation of KIFC1 and its highly correlated genes were studied using DAVID and STRING 9.1.ResultsClinical specimens of ovarian cancers display robust centrosome amplification and deploy centrosome clustering to execute an error-prone mitosis to enable karyotypic heterogeneity that fosters tumor progression and aggressiveness. Our in silico analyses showed KIFC1 overexpression in human ovarian tumors (n = 1090) and its upregulation associated with tumor aggressiveness utilizing publically-available gene expression databases. KIFC1 expression correlated with advanced tumor grade and stage. Dichotomization of KIFC1 levels revealed a significantly lower overall survival time for patients in high KIFC1 group. Intriguingly, in a matched-cohort of primary (n = 7) and metastatic (n = 7) ovarian samples, no significant differences in KIFC1 expression were detectable, suggesting that high KIFC1 expression may serve as a marker of metastases onset. Nonetheless, KIFC1 levels in both primary and matched metastatic sites were significantly higher compared to normal tissue . Ingenuity based network prediction algorithms combined with pre-established protein interaction networks uncovered several novel cell-cycle related partner genes on the basis of interconnectivity, illuminating the centrosome clustering independent agenda of KIFC1 in ovarian tumor progression.ConclusionsOvarian cancers display amplified centrosomes, a feature of aggressive tumors. To cope up with the abnormal centrosomal load, ovarian cancer cells upregulate genes like KIFC1 that are known to induce centrosome clustering. Our data underscore KIFC1 as a putative biomarker that predicts worse prognosis, poor overall survival and may serve as a potential marker of onset of metastatic dissemination in ovarian cancer patients.


Scientific Reports | 2017

Multi-institutional study of nuclear KIFC1 as a biomarker of poor prognosis in African American women with triple-negative breast cancer

Angela Ogden; Chakravarthy Garlapati; Xiaoxian Li; Ravi Chakra Turaga; Gabriela Oprea-Ilies; Nikita Wright; Shristi Bhattarai; Karuna Mittal; Ceyda Sonmez Wetherilt; Uma Krishnamurti; Michelle D. Reid; Mildred Jones; Meenakshi V. Gupta; Remus Osan; Sonal Pattni; Ansa Riaz; Sergey Klimov; Arundhati Rao; Guilherme Cantuaria; Padmashree C.G. Rida; Ritu Aneja

Nuclear KIFC1 (nKIFC1) predicts worse outcomes in breast cancer, but its prognostic value within racially distinct triple-negative breast cancer (TNBC) patients is unknown. Thus, nKIFC1 expression was assessed by immunohistochemistry in 163 African American (AA) and 144 White TNBC tissue microarrays (TMAs) pooled from four hospitals. nKIFC1 correlated significantly with Ki67 in White TNBCs but not in AA TNBCs, suggesting that nKIFC1 is not merely a surrogate for proliferation in AA TNBCs. High nKIFC1 weighted index (WI) was associated with significantly worse overall survival (OS), progression-free survival (PFS), and distant metastasis-free survival (DMFS) (Hazard Ratios [HRs] = 3.5, 3.1, and 3.8, respectively; P = 0.01, 0.009, and 0.007, respectively) in multivariable Cox models in AA TNBCs but not White TNBCs. Furthermore, KIFC1 knockdown more severely impaired migration in AA TNBC cells than White TNBC cells. Collectively, these data suggest that nKIFC1 WI an independent biomarker of poor prognosis in AA TNBC patients, potentially due to the necessity of KIFC1 for migration in AA TNBC cells.


PLOS ONE | 2017

Distinctions in Breast Tumor Recurrence Patterns Post-Therapy among Racially Distinct Populations

Nikita Wright; Jun Xia; Guilherme Cantuaria; Sergey Klimov; Mildred Jones; Pranay Neema; Dora Il’yasova; Uma Krishnamurti; Xiaoxian Li; Michelle D. Reid; Meenakshi V. Gupta; Padmashree C.G. Rida; Remus Osan; Ritu Aneja

Background Clinical studies have revealed a higher risk of breast tumor recurrence in African-American (AA) patients compared to European-American (EA) patients, contributing to the alarming inequality in clinical outcomes among the ethnic groups. However, distinctions in recurrence patterns upon receiving hormone, radiation, and/or chemotherapy between the races remain poorly characterized. Methods We compared patterns and rates (per 1000 cancer patients per 1 year) of recurrence following each form of treatment between AA (n = 1850) and EA breast cancer patients (n = 7931) from a cohort of patients (n = 10504) treated between 2005–2015 at Northside Hospital in Atlanta, GA. Results Among patients who received any combination of adjuvant therapy, AA displayed higher overall rates of recurrence than EA (p = 0.015; HR: 1.699; CI: 1.108–2.606). Furthermore, recurrence rates were higher in AA than EA among stage I (p = 0.031; HR: 1.736; CI: 1.052–2.864) and T1 classified patients (p = 0.003; HR: 2.009; CI: 1.263–3.197). Interestingly, among patients who received neoadjuvant chemotherapy, AA displayed higher rates of local recurrence than EA (p = 0.024; HR: 7.134; CI: 1.295–39.313). Conclusion Our analysis revealed higher incidence rates of recurrence in AA compared to EA among patients that received any combination of adjuvant therapy. Moreover, our data demonstrates an increased risk of tumor recurrence in AA than EA among patients diagnosed with minimally invasive disease. This is the first clinical study to suggest that neoadjuvant chemotherapy improves breast cancer recurrence rates and patterns in AA.


Cancer and Metastasis Reviews | 2015

How to be good at being bad: centrosome amplification and mitotic propensity drive intratumoral heterogeneity

Padmashree C.G. Rida; Guilherme Cantuaria; Michelle D. Reid; Omer Kucuk; Ritu Aneja

Cancer is truly an iconic disease—a tour de force whose multiple formidable strengths can be attributed to the bewildering heterogeneity that a tumor can manifest both spatially and temporally. A Darwinian evolutionary process is believed to undergird, at least in part, the generation of this heterogeneity that contributes to poor clinical outcomes. Risk assessment in clinical oncology is currently based on a small number of clinicopathologic factors (like stage, histological grade, receptor status, and serum tumor markers) and offers limited accuracy in predicting disease course as evidenced by the prognostic heterogeneity that persists in risk segments produced by present-day models. We posit that this insufficiency stems from the exclusion of key risk contributors from such models, especially the omission of certain factors implicated in generating intratumoral heterogeneity. The extent of centrosome amplification and the mitotic propensity inherent in a tumor are two such vital factors whose contributions to poor prognosis are presently overlooked in risk prognostication. Supernumerary centrosomes occur widely in tumors and are potent drivers of chromosomal instability that fosters intratumoral heterogeneity. The mitotic propensity of a proliferating population of tumor cells reflects the cell cycling kinetics of that population. Since frequent passage through improperly regulated mitotic divisions accelerates production of diverse genotypes, the mitotic propensity inherent in a tumor serves as a powerful beacon of risk. In this review, we highlight how centrosome amplification and error-prone mitoses contribute to poor clinical outcomes and urge the need to develop these cancer-specific traits as much-needed clinically-facile prognostic biomarkers with immense potential value for individualized cancer treatment in the clinic.


Journal of Clinical Oncology | 2018

Patient-Reported Toxicity During Pelvic Intensity-Modulated Radiation Therapy: NRG Oncology–RTOG 1203

Ann H. Klopp; Anamaria R. Yeung; Snehal Deshmukh; Karen M. Gil; Lari Wenzel; Shannon N. Westin; Kent Gifford; David K. Gaffney; William Small; Spencer Thompson; D.E. Doncals; Guilherme Cantuaria; Brian Yaremko; A. Chang; Vijayananda Kundapur; Dasarahally S. Mohan; Michael Haas; Yong Bae Kim; C.L. Ferguson; Stephanie L. Pugh; Lisa A. Kachnic; Deborah Watkins Bruner

Purpose NRG Oncology/RTOG 1203 was designed to compare patient-reported acute toxicity and health-related quality of life during treatment with standard pelvic radiation or intensity-modulated radiation therapy (IMRT) in women with cervical and endometrial cancer. Methods Patients were randomly assigned to standard four-field radiation therapy (RT) or IMRT radiation treatment. The primary end point was change in patient-reported acute GI toxicity from baseline to the end of RT, measured with the bowel domain of the Expanded Prostate Cancer Index Composite (EPIC). Secondary end points included change in patient-reported urinary toxicity, change in GI toxicity measured with the Patient-Reported Outcome Common Terminology Criteria for Adverse Events, and quality of life measured with the Trial Outcome Index. Results From 2012 to 2015, 289 patients were enrolled, of whom 278 were eligible. Between baseline and end of RT, the mean EPIC bowel score declined 23.6 points in the standard RT group and 18.6 points in the IMRT group ( P = .048), the mean EPIC urinary score declined 10.4 points in the standard RT group and 5.6 points in the IMRT group ( P = .03), and the mean Trial Outcome Index score declined 12.8 points in the standard RT group and 8.8 points in the IMRT group ( P = .06). At the end of RT, 51.9% of women who received standard RT and 33.7% who received IMRT reported frequent or almost constant diarrhea ( P = .01), and more patients who received standard RT were taking antidiarrheal medications four or more times daily (20.4% v 7.8%; P = .04). Conclusion Pelvic IMRT was associated with significantly less GI and urinary toxicity than standard RT from the patients perspective.


Cancer Epidemiology, Biomarkers & Prevention | 2018

Abstract PR02: β-Catenin overexpression underlies the aggressive disease course in African American triple-negative breast cancer patients who lack androgen receptor

Karuna Mittal; Shristi Bhattarai; Sergey Klimov; Uma Krishnamurthi; Xiaoxian Li; Ceyda Sonmez Wetherilt; Mohammad A. Aleskandaran; Andrew A. Green; Emad A. Rakha; Ian O. Ellis; Guilherme Cantuaria; Guanhao Wei; Remus Osan; Meenakshi V. Gupta; Upender Manne; Padmashree C.G. Rida; Ritu Aneja

Background: Androgen receptor (AR) has emerged as a new target for treating TNBC. AR is expressed in 10-43% of TNBCs. Although there are conflicting reports in the literature about the effect of AR status on TNBC prognosis, agents targeting AR signaling (enzalutamide) are already being evaluated in AR-positive TNBCs in early-stage clinical trials. However, no study so far has evaluated the association/correlation of AR status with ethnicity in TNBCs and downstream effects of AR loss in TNBCs. Given the association of AR loss with poor prognosis in breast cancer and that the African American (AA) with TNBC suffers aggressive disease course when compared to European American (EA) TNBCs, we hypothesized that AR loss might be an underlying cause of aggressive disease course in AR-negative TNBCs. Thus, in this project we aimed to study if loss or gain of AR in AA and EA TNBCs regulates the expression of β-catenin and leads to more aggressive disease course by activating downstream canonical Wnt-beta catenin signaling. Methods: We evaluated AR expression immunohistochemically in 424 formalin-fixed, paraffin-embedded samples from TNBC patients for whom complete clinicopathologic and overall survival (OS) data were available. Samples with Results: IHC staining of AR indicated that 79.5% of AA TNBCs (n=214) and 70% of EA TNBCs (n=210) were AR negative. Loss of AR was associated with poor overall survival in adjuvant-treated high Ki67 (>14%) (HR=1.72; p=0.095) AA TNBC (n=98) when compared to EA TNBCs (n=80). These data were validated by our in silico findings, which suggested that EA TNBCs (n=81) exhibited higher levels of AR mRNA compared to AA TNBCs (n=41) (p Conclusion: This study suggests that increased expression of β-catenin coupled with AR loss in AAs may underlie the ethnic disparity in outcomes among TNBC patients and strongly supports the prognostic role of AR and β-catenin in this breast cancer subtype. Citation Format: Karuna Mittal, Shristi Bhattarai, Sergey Klimov, Uma Krishnamurthi, Xiaoxian Li, Ceyda Sonmez Wetherilt, Mohammad A. Aleskandaran, Andrew A. Green, Emad A. Rakha, Ian O. Ellis, Guilherme Cantuaria, Guanhao Wei, Remus Mihai Osan, Meenakshi V. Gupta, Upender Manne, Padmashree C.G Rida, Ritu Aneja. β-Catenin overexpression underlies the aggressive disease course in African American triple-negative breast cancer patients who lack androgen receptor [abstract]. In: Proceedings of the Tenth AACR Conference on the Science of Cancer Health Disparities in Racial/Ethnic Minorities and the Medically Underserved; 2017 Sep 25-28; Atlanta, GA. Philadelphia (PA): AACR; Cancer Epidemiol Biomarkers Prev 2018;27(7 Suppl):Abstract nr PR02.


Scientific Reports | 2017

Amplified centrosomes and mitotic index display poor concordance between patient tumors and cultured cancer cells

Karuna Mittal; Da Hoon Choi; Angela Ogden; Shashi Donthamsetty; Brian D. Melton; Meenakshi V. Gupta; Vaishali Pannu; Guilherme Cantuaria; Sooryanarayana Varambally; Michelle D. Reid; Kristin Jonsdottir; Emiel A.M. Janssen; Mohammad A. Aleskandarany; Ian O. Ellis; Emad A. Rakha; Padmashree C.G. Rida; Ritu Aneja

Centrosome aberrations (CA) and abnormal mitoses are considered beacons of malignancy. Cancer cell doubling times in patient tumors are longer than in cultures, but differences in CA between tumors and cultured cells are uncharacterized. We compare mitoses and CA in patient tumors, xenografts, and tumor cell lines. We find that mitoses are rare in patient tumors compared with xenografts and cell lines. Contrastingly, CA is more extensive in patient tumors and xenografts (~35–50% cells) than cell lines (~5–15%), although CA declines in patient-derived tumor cells over time. Intratumoral hypoxia may explain elevated CA in vivo because exposure of cultured cells to hypoxia or mimicking hypoxia pharmacologically or genetically increases CA, and HIF-1α and hypoxic gene signature expression correlate with CA and centrosomal gene signature expression in breast tumors. These results highlight the importance of utilizing low-passage-number patient-derived cell lines in studying CA to more faithfully recapitulate in vivo cellular phenotypes.


Clinical Cancer Research | 2017

Preclinical Development of a Nontoxic Oral Formulation of Monoethanolamine, a Lipid Precursor, for Prostate Cancer Treatment

Roopali Saxena; Chunhua Yang; Mukkavilli Rao; Ravi Chakra Turaga; Chakravarthy Garlapati; Sushma R. Gundala; Kimberly Myers; Ahmed Ghareeb; Shristi Bhattarai; Golnaz Kamalinia; Sangina Bristi; Dan Su; Giovanni Gadda; Padmashree C.G. Rida; Guilherme Cantuaria; Ritu Aneja

Purpose: Most currently available chemotherapeutic agents target rampant cell division in cancer cells, thereby affecting rapidly dividing normal cells resulting in toxic side-effects. This nonspecificity necessitates identification of novel cellular pathways that are reprogrammed selectively in cancer cells and can be exploited to develop pharmacologically superior and less toxic therapeutics. Despite growing awareness on dysregulation of lipid metabolism in cancer cells, targeting lipid biosynthesis is still largely uncharted territory. Herein, we report development of a novel nontoxic orally deliverable anticancer formulation of monoethanolamine (Etn) for prostate cancer by targeting the Kennedy pathway of phosphatidylethanolamine (PE) lipid biosynthesis. Experimental Design: We first evaluated gastrointestinal tract stability, drug–drug interaction liability, pharmacokinetic, and toxicokinetic properties of Etn to evaluate its suitability as a nontoxic orally deliverable agent. We next performed in vitro and in vivo experiments to investigate efficacy and mechanism of action. Results: Our data demonstrate that Etn exhibits excellent bioavailability, gastrointestinal tract stability, and no drug–drug interaction liability. Remarkably, orally fed Etn inhibited tumor growth in four weeks by approximately 67% in mice bearing human prostate cancer PC-3 xenografts without any apparent toxicity. Mechanistically, Etn exploits selective overexpression of choline kinase in cancer cells, resulting in accumulation of phosphoethanolamine (PhosE), accompanied by downregulation of HIF-1α that induces metabolic stress culminating into cell death. Conclusions: Our study provides first evidence for the superior anticancer activity of Etn, a simple lipid precursor formulation, whose nontoxicity conforms to FDA-approved standards, compelling its clinical development for prostate cancer management. Clin Cancer Res; 23(14); 3781–93. ©2017 AACR.

Collaboration


Dive into the Guilherme Cantuaria's collaboration.

Top Co-Authors

Avatar

Ritu Aneja

Georgia State University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Sergey Klimov

Georgia State University

View shared research outputs
Top Co-Authors

Avatar

Vaishali Pannu

Georgia State University

View shared research outputs
Top Co-Authors

Avatar

Remus Osan

Georgia State University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Karuna Mittal

Georgia State University

View shared research outputs
Top Co-Authors

Avatar

Angela Ogden

Georgia State University

View shared research outputs
Top Co-Authors

Avatar

Nikita Wright

Georgia State University

View shared research outputs
Researchain Logo
Decentralizing Knowledge