David A. Potter
University of Minnesota
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by David A. Potter.
Clinical Cancer Research | 2006
Anjaiah Srirangam; Ranjana Mitra; Mu Wang; J. Christopher Gorski; Sunil Badve; Lee Ann Baldridge; Justin Hamilton; Hiromitsu Kishimoto; John W. Hawes; Lang Li; Christie M. Orschell; Edward F. Srour; Janice S. Blum; David B. Donner; George W. Sledge; Harikrishna Nakshatri; David A. Potter
Purpose: These studies were designed to determine whether ritonavir inhibits breast cancer in vitro and in vivo and, if so, how. Experimental Design: Ritonavir effects on breast cancer cell growth were studied in the estrogen receptor (ER)–positive lines MCF7 and T47D and in the ER-negative lines MDA-MB-436 and MDA-MB-231. Effects of ritonavir on Rb-regulated and Akt-mediated cell proliferation were studied. Ritonavir was tested for inhibition of a mammary carcinoma xenograft. Results: ER-positive estradiol-dependent lines (IC50, 12-24 μmol/L) and ER-negative (IC50, 45 μmol/L) lines exhibit ritonavir sensitivity. Ritonavir depletes ER-α levels notably in ER-positive lines. Ritonavir causes G1 arrest, depletes cyclin-dependent kinases 2, 4, and 6 and cyclin D1 but not cyclin E, and depletes phosphorylated Rb and Ser473 Akt. Ritonavir induces apoptosis independent of G1 arrest, inhibiting growth of cells that have passed the G1 checkpoint. Myristoyl-Akt, but not activated K-Ras, rescues ritonavir inhibition. Ritonavir inhibited a MDA-MB-231 xenograft and intratumoral Akt activity at a clinically attainable serum Cmax of 22 ± 8 μmol/L. Because heat shock protein 90 (Hsp90) substrates are depleted by ritonavir, ritonavir effects on Hsp90 were tested. Ritonavir binds Hsp90 (KD, 7.8 μmol/L) and partially inhibits its chaperone function. Ritonavir blocks association of Hsp90 with Akt and, with sustained exposure, notably depletes Hsp90. Stably expressed Hsp90α short hairpin RNA also depletes Hsp90, inhibiting proliferation and sensitizing breast cancer cells to low ritonavir concentrations. Conclusions: Ritonavir inhibits breast cancer growth in part by inhibiting Hsp90 substrates, including Akt. Ritonavir may be of interest for breast cancer therapeutics and its efficacy may be increased by sustained exposure or Hsp90 RNA interference.
Oncogene | 2005
Benyue Zhang; Anjaiah Srirangam; David A. Potter; Ann Roman
The E5 protein of human papillomavirus type 16 (HPV16) is a small hydrophobic protein, which localizes to the cell membrane, Golgi apparatus and endosomes. HPV16 E5 enhances the activation of the epidermal growth factor (EGFR). The activated EGFR is downregulated through the endocytic pathway, where E5 has been shown to inhibit endosomal acidification and trafficking. Ubiquitination of the activated EGFR plays a role in this downregulation. c-Cbl is a ubiquitin ligase that associates with the activated EGFR and targets it for degradation. Since E5 has been shown to form a complex with the EGFR, we tested the hypothesis that E5 affects the interaction of c-Cbl with the EGFR. We found a significant decrease of c-Cbl bound to the EGFR and of ubiquitinated EGFR in the presence of E5. E5 did not affect c-Cbl steady-state level, phosphorylation or translocation to the membrane. This novel result suggests that HPV16 E5 may, at least in part, upregulate EGFR-mediated signal transduction by inhibiting the interaction of c-Cbl with the EGFR, thereby decreasing c-Cbl-mediated degradation of the EGFR.
Journal of Biological Chemistry | 2011
Ranjana Mitra; Zhijun Guo; Monica Milani; Clementina Mesaros; Mariangellys Rodriguez; Julia Nguyen; Xianghua Luo; Duncan J. Clarke; Jatinder K. Lamba; Erin G. Schuetz; David B. Donner; Narender Puli; John R. Falck; Jorge H. Capdevila; Kalpna Gupta; Ian A. Blair; David A. Potter
CYP3A4 expression in breast cancer correlates with decreased overall survival, but the mechanisms are unknown. Cytochrome P450 gene profiling by RNAi silencing demonstrates that CYP3A or 2C8 gene expression is specifically required for growth of the breast cancer lines MCF7, T47D, and MDA-MB-231. CYP3A4 silencing blocks the cell cycle at the G2/M checkpoint and induces apoptosis in the MCF7 line, thereby inhibiting anchorage-dependent growth and survival. CYP3A4 was profiled for NADPH-dependent arachidonic acid (AA) metabolism and synthesized AA epoxygenase products (±)-8,9-, (±)-11,12-, and (±)-14,15-epoxyeicosatrienoic acid (EET) (total turnover of ∼2 pmol/pmol CYP3A4/min) but not hydroxylase products (±)-15-, (±)-19-, or 20-hydroxyeicosatetraenoic acid. Furthermore, eicosanoid profiling revealed that MCF7 cells synthesize EETs in a CYP3A4-dependent manner. The (±)-14,15-EET regioisomer selectively rescues breast cancer cells from CYP3A4 silencing in a concentration-dependent fashion and promotes mitogenesis and anchorage-dependent cloning. Stat3 (Tyr-705) phosphorylation was inhibited by CYP3A4 silencing, providing a potential mechanism for CYP3A4 involvement in breast cancer cell growth. Silencing Stat3 blocks breast cancer cell growth and abrogates (±)-14,15-EET-induced proliferation, indicating a Stat3 requirement for (±)-14,15-EET-mediated cell growth. Although silencing of CYP3A4 reduces nuclear Tyr(P)-705-Stat3, (±)-14,15-EET restores this signaling process and promotes Tyr(P)-705-Stat3 translocation to the nucleus, suggesting that (±)-14,15-EET may be involved in an autocrine/paracrine pathway driving cell growth. These studies indicate that CYP3A4 is a highly active AA epoxygenase that promotes Stat3-mediated breast cancer cell growth in part through (±)-14,15-EET biosynthesis. Furthermore, these studies indicate an essential role for Stat3 as a mediator of epoxygenase activity in breast cancer.
Journal of Biomedical Science | 2004
Vijay Narayanasamy; Snehasis Mukhopadhyay; Mathew J. Palakal; David A. Potter
Associations among biological objects such as genes, proteins, and drugs can be discovered automatically from the scientific literature. TransMiner is a system for finding associations among objects by mining the Medline database of the scientific literature. The direct associations among the objects are discovered based on the principle of co-occurrence in the form of an association graph. The principle of transitive closure is applied to the association graph to find potential transitive associations. The potential transitive associations that are indeed direct are discovered by iterative retrieval and mining of the Medline documents. Those associations that are not found explicitly in the entire Medline database are transitive associations and are the candidates for hypothesis generation. The transitive associations were ranked based on the sum of weight of terms that co-occur with both the objects. The direct and transitive associations are visualized using a graph visualization applet. TransMiner was tested by finding associations among 56 breast cancer genes and among 24 objects in the calpain signal transduction pathway. TransMiner was also used to rediscover associations between magnesium and migraine.
Lancet Oncology | 2015
Edith A. Perez; Ahmad Awada; Joyce O'Shaughnessy; Hope S. Rugo; Chris Twelves; Seock-Ah Im; Patricia Gómez-Pardo; Lee S. Schwartzberg; V. Dieras; Denise A. Yardley; David A. Potter; Audrey Mailliez; Alvaro Moreno-Aspitia; Jin Seok Ahn; Carol Zhao; Ute Hoch; Mary Tagliaferri; Alison L. Hannah; Javier Cortes
BACKGROUND New options are needed for patients with heavily pretreated breast cancer. Etirinotecan pegol is a long-acting topoisomerase-I inhibitor that prolongs exposure to, but reduces the toxicity of, SN38 (the active metabolite of irinotecan). We assessed whether etirinotecan pegol is superior to currently available treatments for patients with previously treated, locally recurrent or metastatic breast cancer. METHODS In this open-label, multicentre, randomised phase 3 study (BEACON; BrEAst Cancer Outcomes with NKTR-102), conducted at 135 sites in 11 countries, patients with locally recurrent or metastatic breast cancer previously treated with an anthracycline, a taxane, and capecitabine (and two to five previous regimens for advanced disease) were randomly assigned (1:1) centrally via an interactive response system to etirinotecan pegol (145 mg/m(2) as a 90-min intravenous infusion every 3 weeks) or single-drug treatment of physicians choice. Patients with stable brain metastases and an Eastern Cooperative Oncology Group performance status of 0-1 were eligible. Randomisation was stratified with a permuted block scheme by region, previous eribulin, and receptor status. After randomisation, patients and investigators were aware of treatment assignments. The primary endpoint was overall survival in the intention-to-treat population. This study is registered with ClinicalTrials.gov, number NCT01492101. FINDINGS Between Dec 19, 2011, and Aug 20, 2013, 852 patients were randomly assigned; 429 to etirinotecan pegol and 423 to treatment of physicians choice. There was no significant difference in overall survival between groups (median 12·4 months [95% CI 11·0-13·6] for the etirinotecan pegol group vs 10·3 months [9·0-11·3] for the treatment of physicians choice group; hazard ratio 0·87 [95% CI 0·75-1·02]; p=0·084). The safety population includes the 831 patients who received at least one dose of assigned treatment (425 assigned to etirinotecan pegol and 406 to treatment of physicians choice). Serious adverse events were recorded for 128 (30%) patients treated with etirinotecan pegol and 129 (32%) treated with treatment of physicians choice. Fewer patients in the etirinotecan pegol group had grade 3 or worse toxicity than those in the treatment of physicians choice group (204 [48%] vs 256 [63%]; p<0·0001). The most common grade 3 or worse adverse events were diarrhoea (41 [10%] in the experimental group vs five [1%] in the control group), neutropenia (41 [10%] vs 125 [31%]), and peripheral neuropathy (two [<1%] vs 15 [4%]). Three patients in the etirinotecan pegol group died of treatment-related adverse events (pneumonia, myelodysplastic syndrome, and acute renal failure) and two in the treatment of physicians choice group (neutropenic sepsis and septic shock). INTERPRETATION This trial did not demonstrate an improvement in overall survival for etirinotecan pegol compared to treatment of physicians choice in patients with heavily pre-treated advanced breast cancer. The toxicity profile noted in the etirinotecan pegol group differed from that in the control group. In view of the frequency of cross-resistance and overlapping toxicities noted with many available drugs and the need for effective drugs in highly refractory disease, etirinotecan pegol may warrant further research in some subgroups of patients. FUNDING Nektar Therapeutics.
Molecular Cancer Therapeutics | 2008
Farnaz Yamoutpour; Vidya Bodempudi; Shay E. Park; Weihong Pan; Mary Mauzy; Robert A. Kratzke; Arkadiusz Z. Dudek; David A. Potter; Richard A. Woo; Donald M. O'Rourke; Donald J. Tindall; Faris Farassati
Epidermal growth factor receptor variant III (EGFRvIII) is a constitutively active mutant form of EGFR that is expressed in 40% to 50% of gliomas and several other malignancies. Here, we describe the therapeutic effects of silencing EGFRvIII on glioma cell lines in vitro and in vivo. A small interfering RNA molecule against EGFRvIII was introduced into EGFRvIII-expressing glioma cells (U87Δ) by electroporation resulting in complete inhibition of expression of EGFRvIII as early as 48 h post-treatment. During EGFRvIII silencing, a decrease in the proliferation and invasiveness of U87Δ cells was accompanied by an increase in apoptosis (P < 0.05). Notably, EGFRvIII silencing inhibited the signal transduction machinery downstream of EGFRvIII as evidenced by decreases in the activated levels of Ras and extracellular signal-regulated kinase. A lentivirus capable of expressing anti-EGFRvIII short hairpin RNA was also able to achieve progressive silencing of EGFRvIII in U87Δ cells in addition to inhibiting cell proliferation, invasiveness, and colony formation in a significant manner (P < 0.05). Silencing EGFRvIII in U87Δ cultures with this virus reduced the expression of factors involved in epithelial-mesenchymal transition including N-cadherin, β-catenin, Snail, Slug, and paxillin but not E-cadherin. The anti-EGFRvIII lentivirus also affected the cell cycle progression of U87Δ cells with a decrease in G1 and increase in S and G2 fractions. In an in vivo model, tumor growth was completely inhibited in severe combined immunodeficient mice (n = 10) injected s.c. with U87Δ cells treated with the anti-EGFRvIII lentivirus (P = 0.005). We conclude that gene specific silencing of EGFRvIII is a promising strategy for treating cancers that contain this mutated receptor. [Mol Cancer Ther 2008;7(11):3586–97]
Clinical Cancer Research | 2011
Ranjana Mitra; Jinseon Lee; Jisuk Jo; Monica Milani; Jeanette N. McClintick; Howard J. Edenberg; Kenneth A. Kesler; Karen M. Rieger; Sunil Badve; Oscar W. Cummings; Ahmed Mohiuddin; Dafydd G. Thomas; Xianghua Luo; Beth E. Juliar; Lang Li; Clementina Mesaros; Ian A. Blair; Anjaiah Srirangam; Robert A. Kratzke; Clement J. McDonald; Jhingook Kim; David A. Potter
Purpose: This study was performed to discover prognostic genomic markers associated with postoperative outcome of stage I to III non–small cell lung cancer (NSCLC) that are reproducible between geographically distant and demographically distinct patient populations. Experimental Design: American patients (n = 27) were stratified on the basis of recurrence and microarray profiling of their tumors was performed to derive a training set of 44 genes. A larger Korean patient validation cohort (n = 138) was also stratified by recurrence and screened for these genes. Four reproducible genes were identified and used to construct genomic and clinicogenomic Cox models for both cohorts. Results: Four genomic markers, DBN1 (drebrin 1), CACNB3 (calcium channel beta 3), FLAD1 (PP591; flavin adenine dinucleotide synthetase), and CCND2 (cyclin D2), exhibited highly significant differential expression in recurrent tumors in the training set (P < 0.001). In the validation set, DBN1, FLAD1 (PP591), and CACNB3 were significant by Cox univariate analysis (P ≤ 0.035), whereas only DBN1 was significant by multivariate analysis. Genomic and clinicogenomic models for recurrence-free survival (RFS) were equally effective for risk stratification of stage I to II or I to III patients (all models P < 0.0001). For stage I to II or I to III patients, 5-year RFS of the low- and high-risk patients was approximately 70% versus 30% for both models. The genomic model for overall survival of stage I to III patients was improved by addition of pT and pN stage (P < 0.0013 vs. 0.010). Conclusion: A 4-gene prognostic model incorporating the multivariate marker DBN1 exhibits potential clinical utility for risk stratification of stage I to III NSCLC patients. Clin Cancer Res; 17(9); 2934–46. ©2011 AACR.
Oncology Nursing Forum | 2008
Janet S. Carpenter; Susan M. Rawl; Jennifer Porter; Karen Schmidt; Jennifer Tornatta; Foluso Ojewole; Paul R. Helft; David A. Potter; Christopher Sweeney; R. Brian Giesler
PURPOSE/OBJECTIVES To assess patient and provider responses to a computerized symptom assessment system. DESIGN Descriptive, longitudinal study with retrospective, longitudinal medical records review. SETTING University-based National Cancer Institute-designated outpatient cancer center. SAMPLE 80 oncology outpatients receiving chemotherapy, 8 providers, and 30 medical records. METHODS Patients completed the computerized assessment during three chemotherapy follow-up clinic appointments (times 1, 2, and 3). Patient usability was recorded via an observer checklist (ease of use) and the computer (completion time). Patient satisfaction and impact were assessed during telephone interviews two to three days after times 1 and 3 only. Provider usability and impact were assessed at the end of the study using a questionnaire and focus groups, whereas effect on provider documentation was assessed through chart audits. MAIN RESEARCH VARIABLES Patient usability (ease of use, completion time), satisfaction, and impact; provider usability and impact. FINDINGS Patients reported good usability, high satisfaction, and modest impact on discussions with their providers. Providers reported modest usability, modest impact on discussions with patients, and had varied reactions as to how the system affected practice. Documentation of symptoms was largely absent before and after implementation. CONCLUSIONS This system demonstrated good usability and satisfaction but had only a modest impact on symptom-related discussions and no impact on documentation. IMPLICATIONS FOR NURSING A computerized system can help address barriers to symptom assessment but may not improve documentation unless it can be integrated into existing medical records systems.
Journal of Thoracic Oncology | 2011
Anjaiah Srirangam; Monica Milani; Ranjana Mitra; Zhijun Guo; Mariangellys Rodriguez; Hitesh Kathuria; Seiji Fukuda; Anthony E. Rizzardi; Stephen C. Schmechel; David G. Skalnik; Louis M. Pelus; David A. Potter
Introduction: Ritonavir is a potential therapeutic agent in lung cancer, but its targets in lung adenocarcinoma are unknown, as are candidate biomarkers for its activity. Methods: RNAi was used to identify genes whose expression affects ritonavir sensitivity. Synergy between ritonavir, gemcitabine, and cisplatin was tested by isobologram analysis. Results: Ritonavir inhibits growth of K-ras mutant lung adenocarcinoma lines A549, H522, H23, and K-ras wild-type line H838. Ritonavir causes G0/G1 arrest and apoptosis. Associated with G0/G1 arrest, ritonavir down-regulates cyclin-dependent kinases, cyclin D1, and retinoblastoma protein phosphorylation. Associated with induction of apoptosis, ritonavir reduces survivin messenger RNA and protein levels more than twofold. Ritonavir inhibits phosphorylation of c-Src and signal transducer and activator of transcription protein 3, which are important events for survivin gene expression and cell growth, and induces cleavage of PARP1. Although knock down of survivin, c-Src, or signal transducer and activator of transcription protein 3 inhibits cell growth, only survivin knock down enhances ritonavir inhibition of growth and survivin overexpression promotes ritonavir resistance. Ritonavir was tested in combination with gemcitabine or cisplatin, exhibiting synergistic and additive effects, respectively. The combination of ritonavir/gemcitabine/cisplatin is synergistic in the A549 line and additive in the H522 line, at clinically feasible ritonavir concentrations (<10 &mgr;M). Conclusions: Ritonavir is of interest for lung adenocarcinoma therapeutics, and survivin is an important target and potential biomarker for its sensitivity. Ritonavir cooperation with gemcitabine/cisplatin might be explained by involvement of PARP1 in repair of cisplatin-mediated DNA damage and survivin in repair of gemcitabine-mediated double-stranded DNA breaks.
Histopathology | 2005
John D. Henley; Summerlin Dj; David A. Potter; Tomich C
our group and others have shown that MUC1 is overexpressed in high nuclear grade and advanced stage RCCs. Moreover, we have also demonstrated that primary RCCs which express MUC1 diffusely are associated with a poorer prognosis and frequent metastases. In the present study, we have shown that MUC1 expression is increased and diffuse in metastases from RCCs. Interestingly, we observed, in several cases, preferential localization of MUC1+ tumour cells around large intratumoral vessels, though this did not reach statistical significance. Moreover, MUC1 is known to interact with many ligands on the cell surface such as intercellular adhesion molecule-1, which is a cell adhesion molecule expressed by endothelial cells and initiates a calcium signal. The value of E-cadherin in renal cancer is more controversial. In the present study, we observed that E-cadherin was expressed in only 27% (6 ⁄ 22) of metastases and in 3 ⁄ 10 renal primary tumours (30%). No direct relationship between MUC1 and E-cadherin expression was seen, which confirms the results of a recent study. MUC1 is thought to be implicated in tumour progression and metastases by several mechanisms. MUC1 interacts with many ligands on the cell surface and could limit the immune host recognition by steric hindrance. MUC1 also plays a role in different intracellular signalling pathways. The cytoplasmic tail of MUC1 binds b-catenin, which is a member of the WNT signalling cascade. The interaction between MUC1 and b-catenin ⁄ E-cadherin leads to the disruption of cell adhesion, which could favour cell detachment and migration. Our study highlights that MUC1 is diffusely expressed in metastases of RCCs and confirms that MUC1 could be a potential therapeutic target for disseminated renal cancer.