GLORIA

GEOMAR Library Ocean Research Information Access

Your email was sent successfully. Check your inbox.

An error occurred while sending the email. Please try again.

Proceed reservation?

Export
  • 1
    In: Clinical Cancer Research, American Association for Cancer Research (AACR), Vol. 26, No. 21 ( 2020-11-01), p. 5720-5734
    Abstract: Docetaxel plays an indispensable role in the management of advanced prostate cancer. However, more than half of patients do not respond to docetaxel, and those good responders frequently experience significant cumulative toxicity, which limits its dose duration and intensity. Hence, a second agent that could increase the initial efficacy of docetaxel and maintain tolerability at biologically effective doses may improve outcomes for patients. Experimental Design: We determined phosphodiesterase 5 (PDE5) expression levels in human and genetically engineered mouse (GEM) prostate tissues and tumor-derived cell lines. Furthermore, we investigated the therapeutic benefits and underlying mechanism of PDE5 inhibitor sildenafil in combination with docetaxel using in vitro, Pten conditional knockout (cKO), derived tumoroid and xenograft prostate cancer models. Results: PDE5 expression was higher in both human and mouse prostate tumors and cancer cell lines compared with normal tissues/cells. In GEM prostate-derived cell lines, PDE5 expression increased from normal prostate (wild-type) epithelial cells to androgen-dependent and castrated prostate-derived cell lines. The addition of physiologically achievable concentrations of sildenafil enhanced docetaxel-induced prostate cancer cell growth inhibition and apoptosis in vitro, reduced murine 3D tumoroid growth, and in vivo tumorigenicity as compared with docetaxel alone. Furthermore, sildenafil enhanced docetaxel-induced NO and cGMP levels thereby augmenting antitumor activity. Conclusions: Our results demonstrate that sildenafil's addition could sensitize docetaxel chemotherapy in prostate cancer cells at much lesser concentration than needed for inducing cell death. Thus, the combinatorial treatment of sildenafil and docetaxel may improve anticancer efficacy and reduce chemotherapy-induced side-effects among patients with advanced prostate cancer.
    Type of Medium: Online Resource
    ISSN: 1078-0432 , 1557-3265
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2020
    detail.hit.zdb_id: 1225457-5
    detail.hit.zdb_id: 2036787-9
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 73, No. 8_Supplement ( 2013-04-15), p. 4044-4044
    Abstract: The limited effectiveness of therapy for patients with advanced stage Head and Neck Squamous Cell Carcinoma (HNSCC) or recurrent disease is a reflection of an incomplete understanding of the molecular basis of HNSCC pathogenesis. Cellular senescence is an extremely stable form of cell cycle arrest that limits the proliferation of damaged cells and may act as a natural barrier to cancer progression. MUC4, a high molecular weight glycoprotein, is overexpressed in many cancers and is implicated in cell proliferation, adhesion, anti-apoptosis, cell cycle regulation, migration and invasion of various carcinomas, but nothing is known about its clinical relevance and the mechanism through which it regulates cancer progression in HNSCC. Therefore, the present study was aimed to investigate clinical relevance and a potential role of MUC4 in cellular senescence in HNSCCs. Using immunohistochemical analysis, we observed a significant up regulation of MUC4 in 79% (68/86) of HNSCC tissue samples compared to only 10% (1/10) of benign tissues. Further, we observed high expression of MUC4 in a majority of HNSCC cell lines tested. Knockdown (KD) of MUC4 with specific shRNA in HNSCC cells markedly decreased cell proliferation. The doubling time increased from 26 h to 36 h and 34 h to 42 h in MUC4 silenced UMSCC-1 and SCC-10B cells, respectively as compared to their control counterparts. MUC4 KD in UMSCC-1 and UMSCC-10B cells resulted in accumulation of cells at the G0/G1 phase with concomitant decrease in the expression of cell cycle regulating proteins like Cyclin-E and Cyclin-D1, and decrease in BrDU incorporation. More importantly, MUC4 KD resulted in the induction of cellular senescence in both cell lines as indicated by an increase in the number of flat, enlarged and senescence-associated β-galactosidase (SA-β-Gal) positive cells. Further, MUC4 KD resulted in the inhibition of FAK signaling, and decreased motility and invasive behavior in both UMSCC-1 and UMSCC-10B cells. Mechanistic dissection of senescent response to MUC4 silencing indicated decreased acetylated histone enrichment at Cyclin-E/Cyclin-D1 promoters, leading to their downregulation. Further, both cell lines UMSCC-1 and UMSCC-10B underwent a P16 and P21 dependent cellular senescence in response to MUC4 KD that requires inactivation of Akt and ERK signaling. In conclusions, these findings suggest a novel role of MUC4 in regulating cellular senescence and provide evidence of the functional role of MUC4 in the proliferation, motility and invasion of HNSCC cells. Therefore, downregulation of MUC4 may be a promising therapeutic approach for Head and Neck cancer treatment. Citation Format: Muzafar A. Macha, Satyanarayana Rachagani, Priya Pai, Maneesh Jain, Williams M. Lydiatt, Russell B. Smith, Sonny L. Johansson, Subodh M. Lele, Sham S. Kakar, Farghaly H. Ibrahim, John H. Lee. MUC4 knockdown induces cellular senescence in head and neck cancer cells. [abstract]. In: Proceedings of the 104th Annual Meeting of the American Association for Cancer Research; 2013 Apr 6-10; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2013;73(8 Suppl):Abstract nr 4044. doi:10.1158/1538-7445.AM2013-4044
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2013
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 75, No. 15_Supplement ( 2015-08-01), p. CT323-CT323
    Abstract: Background: There is an urgent need for better trial designs to assess targeted drug combinations. We proposed a novel intrapatient (intrapt) dose escalation trial design to optimize drug exposures, minimize pharmacokinetic (PK) variability and reduce patient (pt) numbers needed (Yap et al, JCO 2013). In vivo synergy between PARP and PI3K pathway inhibition was seen in BRCA1-related and sporadic cancers (Juvekar et al; Ibrahim et al, Cancer Discov 2012), providing rationale for this study. Methods: Two-stage investigator initiated phase I trial: a) Intrapt dose escalation; b) Recommended phase II combination dose (RP2CD) expansion. Advanced cancer pts received escalating doses of AZD5363 (AZD) BID in 2 parallel arms (4 days on 3 days off [4/7 arm] at 320, 400, 480mg; 2 days on 5 days off [2/7 arm] at 480, 560, 640mg) with Olaparib (Ola) at 300mg BID in 3 weekly cycles. AZD was escalated after each cycle in each pt if drug related toxicities were ≤CTCAE G2. Dose limiting toxicities (DLT) were assessed during the 1st cycle of each dose level (DL). ≥6 evaluable pts were required at each DL. RECIST assessment was done every 3 cycles. Prior PARP or PI3K/AKT inhibitor use was allowed. PK and pharmacodynamics (PD) were assessed in tumor and normal tissue. Targeted +/- whole exome next generation sequencing was assessed in tumor and serial plasma DNA samples in all pts for predictive biomarkers of response. Results: Dose escalation was completed in 7.5 months (m) in 20 pts in 1 center; ≥6 evaluable pts were treated at each of the 3 DLs in both arms. Common ( & gt;15%) G1-2 toxicities were nausea, vomiting, fatigue, diarrhea and anemia. A DLT of G3 rash was seen at 480mg BID 4/7 AZD + 300mg BID Ola. Non DLT G3 anemia (n = 2), diarrhea (n = 2), fatigue (n = 1) and vomiting (n = 1) were seen in 4/7 arm; G3 hyperglycemia (n = 1), transaminitis (n = 1) and fatigue (n = 2) in 2/7 arm. No significant PK interactions were seen between Ola and AZD. Intrapt dose escalation of AZD showed dose dependent increases in PK exposures. Platelet-rich plasma PD showed significant decreases in pSer9 GSK3β post-therapy at all DLs (mean ≥55% [p & lt;0.002] in 4/7 arm and ≥70% [p & lt;0.0001] in 2/7 arm). Confirmed partial responses (PR) were seen in a BRCA wild type PTEN LOH platinum resistant ovarian cancer (PROC) pt for 6m and a BRCA1 mutant (mut) PROC pt for 5.5m+. Unconfirmed PR was seen in a BRCA1 mut breast cancer pt (2.5m+). A BRCA1 mut castration resistant prostate cancer pt has PCWG2 response (PSA 14 to 0.7 μg/L) and tumor response on DW-MRI (8m+). A PI3K/mTOR inhibitor resistant peritoneal mesothelioma pt has stable disease for 9m+ with 66% CA125 decline (202 to 69 U/mL). RP2CD was established at 640mg BID 2/7 AZD + 300mg BID Ola based on tolerability. Conclusion: This novel trial design led to rapid completion of dose escalation. RP2CD expansion (n = 40) is ongoing in: a) germline BRCA mut cancers; b) sporadic cancers with relevant somatic mutations. Citation Format: Vasiliki Michalarea, David Lorente, Juanita Lopez, Suzanne Carreira, Hasina Hassam, Mona Parmar, Nitharsan Sathiyayogan, Alison Turner, Emma Hall, Sonia Serrano Fandos, Satyanarayana Seeramreddi, Shaun Decordova, Karen Swales, Ruth Ruddle, Florence Raynaud, Nina Tunariu, Gerhardt Attard, L. Rhoda Molife, Udai Banerji, Ruth Plummer, Johann S. de Bono, Timothy A. Yap. Accelerated phase I trial of two schedules of the combination of the PARP inhibitor olaparib and AKT inhibitor AZD5363 using a novel intrapatient dose escalation design in advanced cancer patients. [abstract]. In: Proceedings of the 106th Annual Meeting of the American Association for Cancer Research; 2015 Apr 18-22; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2015;75(15 Suppl):Abstract nr CT323. doi:10.1158/1538-7445.AM2015-CT323
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2015
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 78, No. 13_Supplement ( 2018-07-01), p. 5897-5897
    Abstract: Background: Docetaxel remains the first-line therapeutic intervention for metastatic and castration-resistant (CR) prostate cancer (PCa). However, the therapeutic efficacy is limited due to unresponsiveness, toxicity, and drug-resistance. The availability of additional therapies increases the PCa patient survival modestly, but the development of cross-resistance limit the therapeutic efficacy. Hence, there is a need to understand the mechanisms of resistance and identify a novel target for the better management of CR PCa patients. Methods: Docetaxel resistant 22Rv1, LNCaP C-81, and PC3 PCa cells were established, and its resistance phenotype was determined by cell growth inhibition (MTT assay), apoptosis markers (western blot), tumor sphere assay, drug efflux property (SP analysis by FACS), auto fluorescence (FACS). qRT-PCR, western blot, and confocal microscopic analysis were also performed to confirm the drug-resistant marker phenotype. Additional experiments were performed to determine the underlying molecular mechanisms of docetaxel resistance. Results: The docetaxel resistant PCa cells grew well at the higher concentration of docetaxel (120, 15 and 50 nM of docetaxel, respectively) and did not result in apoptosis as measured by cPARP and caspase-3 cleavage. Docetaxel resistant PCa cells confer cross-resistance to second-generation chemotherapeutic agent cabazitaxel and show altered cell proliferation and invasion. Blocking by the ABCB1 specific inhibitor enhances docetaxel-induced cell death on par with parental cells. Side population analysis by flow cytometry confirms the acquired drug efflux property. Side population, autofluorescence and tumor sphere analyses confirmed the drug-resistance and stem-like cell phenotype. qRT-PCR, western blot and confocal microscopy show the abundant expression levels of the drug transporters, ABCB1 and ABCG2. Further, the docetaxel resistant PCa cells show higher stem cell network proteins such as PAF1, POU5F1, NANOG and SOX9 expression levels compared to age-matched control cells. Conversely, Tet-inducible PAF1 knockdown reduces embryonic stem cell network proteins and reverses docetaxel-resistance phenotype. Conclusions: Collectively, our study suggests that the stem cell factors such as PAF1 play a major role in docetaxel resistance and aggressiveness to PCa cells. Understanding the associated mechanisms and targeting these factors could lead a better management approach for CRPCa patients. Citation Format: Sakthivel Muniyan, Rama Krishna Nimmakayala, Saswati Karmakar, Satyanarayana Rachagani, Jawed A. Siddiqui, Parthasarathy Seshacharyulu, Ming-Fong Lin, Kaustubh Datta, Moorthy P. Ponnusamy, Surinder K. Batra. Role of polymerase II associated factor 1, PAF1, in docetaxel resistant prostate cancer cells [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2018; 2018 Apr 14-18; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2018;78(13 Suppl):Abstract nr 5897.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2018
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 79, No. 13_Supplement ( 2019-07-01), p. 4684-4684
    Abstract: Background Glioblastoma (GBM) is the aggressive primary brain tumor with a median survival rate of 14.6 months. Currently, first-line treatment includes surgical resection, chemoradiation, and adjuvant chemotherapy with temozolomide (TMZ). However, GBM recurs most often within 6.9 months. Most of the targeted therapies failed in GBM, possibly due to the co-activation of the receptor tyrosine kinases (RTKs). Genetic analysis on GBM tumor revealed that RTKs are dysregulated, with epidermal growth factor receptor (EGFR) representing 57.4% of the deleted/mutated GBM, about 30 - 40% of GBM patients with EGFR amplification carry an oncogenic gene rearrangement EGFR variant III (EGFRvIII) which is constitutively active. In addition, EGFRvIII co-activates cMET RTKs thereby enriches GBM cancer stem like cells (CSCs). CSCs are relatively radio- and chemo- resistant and play a pivotal role in tumor recurrence/progression. We hypothesize that afatinib and TMZ combination would inhibit EGFRvIII co-activation and tumor progression in GBM model. Methods GBM cell lines U87MG, U87MG transfected with EGFRvIII (U87 EGFRvIII) and SP/CSC isolated from U87 EGFRvIII cells were treated with afatinib, TMZ alone or in combination and analyzed. The in vivo efficacy of these drugs were also analyzed on U87 EGFRvIII orthograft mouse model. Results We observed significantly higher proportion of CSCs in U87 EGFRvIII cells compared to U87MG cells (p = 0.03). While afatinib decreased the percentage of CSCs in both U87MG and U87 EGFRvIII cells (p = 0.02), TMZ only decreased CSC population in U87MG cells. However, combination of afatinib with TMZ significantly decreased the CSCs in both U87MG and U87 EGFRvIII cells (p = 0.02). Our clonogenic (tumorsphere) assay revealed significantly more tumorspheres (p=0.01) formed by U87 EGFRvIII CSCs cells than U87MG CSCs. In addition, we also observed that TMZ significantly decreased the self-renewal properties of U87 CSCs compared to U87 EGFRvIII CSCs. Furthermore, afatinib alone or in combination with TMZ significantly abolished the tumorsphere formation by U87 EGFRvIII CSCs. The underlying mechanism revealed inhibition of cMET RTK co-activation by EGFRvIII using afatinib. Our in vivo studies using U87 EGFRvIII orthograft model revealed significant tumor growth inhibition by afatinib and TMZ combination compared to control and either drug alone. Conclusion Our results strongly support the efficacy of afatinib and TMZ combination in inhibiting EGFRvIII-cMET signaling mediated GBM stemness and prevention of tumor progression. This treatment should be tested in EGFR amplified/mutated GBM patients. Note: This abstract was not presented at the meeting. Citation Format: Raghupathy Vengoji, Muzafar A. Macha, Ramakrishna Nimmakayala, Satyanarayana Rachagani, Kavita Mallya, Maneesh Jain, Moorthy P. Ponnusamy, Surinder K. Batra, Nicole Shonka. Afatinib targets glioblastoma stem cells by inhibiting EGFRVIII-cMet co-activation [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2019; 2019 Mar 29-Apr 3; Atlanta, GA. Philadelphia (PA): AACR; Cancer Res 2019;79(13 Suppl):Abstract nr 4684.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2019
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 6
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 70, No. 16 ( 2010-08-15), p. 6437-6447
    Abstract: Glioblastoma (GBM; grade IV astrocytoma) is a very aggressive form of brain cancer with a poor survival and few qualified predictive markers. This study integrates experimentally validated genes that showed specific upregulation in GBM along with their protein-protein interaction information. A system level analysis was used to construct GBM-specific network. Computation of topological parameters of networks showed scale-free pattern and hierarchical organization. From the large network involving 1,447 proteins, we synthesized subnetworks and annotated them with highly enriched biological processes. A careful dissection of the functional modules, important nodes, and their connections identified two novel intermediary molecules CSK21 and protein phosphatase 1 α (PP1A) connecting the two subnetworks CDC2-PTEN-TOP2A-CAV1-P53 and CDC2-CAV1-RB-P53-PTEN, respectively. Real-time quantitative reverse transcription-PCR analysis revealed CSK21 to be moderately upregulated and PP1A to be overexpressed by 20-fold in GBM tumor samples. Immunohistochemical staining revealed nuclear expression of PP1A only in GBM samples. Thus, CSK21 and PP1A, whose functions are intimately associated with cell cycle regulation, might play key role in gliomagenesis. Cancer Res; 70(16); 6437–47. ©2010 AACR.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2010
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 7
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2016
    In:  Cancer Research Vol. 76, No. 14_Supplement ( 2016-07-15), p. 1726-1726
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 76, No. 14_Supplement ( 2016-07-15), p. 1726-1726
    Abstract: Introduction: Pancreatic cancer (PC) is the third leading cause of cancer related deaths in United States and has a 5 year survival rate of 7.2%. Current front line therapy for PC includes Gemcitabine, 5FU and Abraxane. Cancer stem cells (CSCs), a small subpopulation in cancer comprising of less than 5% of total cancer cells, are responsible for therapy resistance and aggressive metastasis resulting in poor prognosis of cancer including PC. It has already been established that EGFR family members (EGFR and HER2) are involved in CSC maintenance. Therefore, CSCs are important target to reduce the drug resistance and aggressiveness. Afatinib is an FDA approved pan-EGFR inhibitor. Our major goal is to use afatinib to target pancreatic CSC and understand the mechanism of its action. Methods: After identifying the IC50 by MTT assay, PC cell lines (Capan1 and SW1990) were treated with afatinib and gemcitabine and cell lysates were analyzed by western blotting (WB) for EGFR family members and CSC markers like Shh, SOX2, Aldh1 and CD133.CSCs or Side Population (SP) were analyzed in afatinib and gemcitabine treated Capan1 cells by Hoechst based FACS analysis. Hoechst based FACS was used to isolate SP/CSC and NSP (non-side population) from parental PC cell lines and their IC50 for gemcitabine and afatinib were determined. SP and NSP cells were subjected to drug treatments to determine alterations in EGFR family proteins and stem cell markers by WB. Tumor sphere assay (TSA) was performed to determine tumorigenic potential of SP/CSC and NSP fractions with and without drug treatments. We developed Tumor-Organoids from Kras;PdxCre (KC) mice and used them to test effect of afatinib in 3D culture system. Results: Our results showed significant decrease in SP/CSC fraction upon afatinib treatment and an enrichment of SP/CSC fraction on gemcitabine treatment. The SP/CSC cells showed higher sensitivity to afatinib and resistance towards gemcitabine when compared to parental cells suggesting specific targeting on SP/CSCs. Upon afatinib administration, we observed a reduction in phospho forms of EGFR family members and CSC markers like Sox2, Aldh1 and CD133 confirming functionality of the drug and its effect on pancreatic CSCs. A significant decrease was also observed in number and size of tumor spheres formed in vitro with both SP/CSC and NSP. We also confirmed a drastic reduction in the size and number of tumor-organoids following nine days of afatinib treatment compared to untreated controls. Conclusion: Our results demonstrate a novel role of afatinib in targeting pancreatic CSCs and provide a scope to generate targeted therapy towards PC. Citation Format: Garima Kaushik, Parthasarathy Seshacharyulu, Satyanarayana Rachagani, Muzafar A. Macha, Moorthy P. Ponnusamy, Surinder K. Batra. Targeting pancreatic cancer stem cells by afatinib in organoid culture. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 1726.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2016
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 8
    In: Clinical Cancer Research, American Association for Cancer Research (AACR), Vol. 18, No. 22 ( 2012-11-15), p. 6188-6198
    Abstract: Purpose: To study the expression and function of a novel cell-cycle regulatory protein, human ecdysoneless (Ecd), during pancreatic cancer pathogenesis. Experimental Design: Immunohistochemical expression profiling of Ecd was done in nonneoplastic normal pancreatic tissues and pancreatic ductal adenocarcinoma lesions (from tissue microarray and Rapid Autopsy program) as well as precancerous PanIN lesions and metastatic organs. To analyze the biological significance of Ecd in pancreatic cancer progression, Ecd was stably knocked down in pancreatic cancer cell line followed by in vitro and in vivo functional assays. Results: Normal pancreatic ducts showed very weak to no Ecd expression compared to significant positive expression in pancreatic cancer tissues (mean ± SE composite score: 0.3 ± 0.2 and 3.8 ± 0.2 respectively, P & lt; 0.0001) as well as in PanIN precursor lesions with a progressive increase in Ecd expression with increasing dysplasia (PanIN-1–PanIN-3). Analysis of matched primary tumors and metastases from patients with pancreatic cancer revealed that Ecd is highly expressed in both primary pancreatic tumor and in distant metastatic sites. Furthermore, knockdown of Ecd suppressed cell proliferation in vitro and tumorigenicity of pancreatic cancer cells in mice orthotopic tumors. Microarray study revealed that Ecd regulates expression of glucose transporter GLUT4 in pancreatic cancer cells and was subsequently shown to modulate glucose uptake, lactate production, and ATP generation by pancreatic cancer cells. Finally, knockdown of Ecd also reduced level of pAkt, key signaling molecule known to regulate aerobic glycolysis in cancer cells. Conclusion: Ecd is a novel tumor-promoting factor that is differentially expressed in pancreatic cancer and potentially regulates glucose metabolism within cancer cells. Clin Cancer Res; 18(22); 6188–98. ©2012 AACR.
    Type of Medium: Online Resource
    ISSN: 1078-0432 , 1557-3265
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2012
    detail.hit.zdb_id: 1225457-5
    detail.hit.zdb_id: 2036787-9
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 9
    In: Clinical Cancer Research, American Association for Cancer Research (AACR), Vol. 20, No. 3 ( 2014-02-01), p. 688-700
    Abstract: Purpose: MUC4 shows aberrant expression in early pancreatic lesions and a high specificity for pancreatic cancer. It thus has a high potential to be a sensitive and specific biomarker. Unfortunately, its low serum level limits its diagnostic/prognostic potential. We here report that a multifaceted acute phase protein lipocalin 2, regulated by MUC4, could be a potential diagnostic/prognostic marker for pancreatic cancer. Experimental Designs and Results: Overexpression/knockdown, luciferase reporter and molecular inhibition studies revealed that MUC4 regulates lipocalin 2 by stabilizing HER2 and stimulating AKT, which results in the activation of NF-κB. Immunohistochemical analyses of lipocalin 2 and MUC4 showed a significant positive correlation between MUC4 and lipocalin 2 in primary, metastatic tissues (Spearman correlation coefficient 0.71, P = 0.002) from rapid autopsy tissue sample from patients with pancreatic cancer as well as in serum and tissue samples from spontaneous KRASG12D mouse pancreatic cancer model (Spearman correlation coefficient 0.98, P & lt; 0.05). Lipocalin 2 levels increased progressively with disease advancement (344.2 ± 22.8 ng/mL for 10 weeks to 3067.2 ± 572.6 for 50 weeks; P & lt; 0.0001). In human pancreatic cancer cases, significantly elevated levels of lipocalin 2 were observed in patients with pancreatic cancer (148 ± 13.18 ng/mL) in comparison with controls (73.27 ± 4.9 ng/mL, P = 0.014). Analyses of pre- and postchemotherapy patients showed higher lipocalin 2 levels in prechemotherapy patients [121.7 ng/mL; 95% confidence interval (CI), 98.1–150.9] in comparison with the postchemotherapy (92.6 ng/mL; 95% CI, 76.7–111.6; P = 0.06) group. Conclusions: This study delineates the association and the downstream mechanisms of MUC4-regulated elevation of lipocalin-2 (via HER2/AKT/NF-κB) and its clinical significance for prognosis of pancreatic cancer. Clin Cancer Res; 20(3); 688–700. ©2013 AACR.
    Type of Medium: Online Resource
    ISSN: 1078-0432 , 1557-3265
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2014
    detail.hit.zdb_id: 1225457-5
    detail.hit.zdb_id: 2036787-9
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 10
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2018
    In:  Cancer Research Vol. 78, No. 13_Supplement ( 2018-07-01), p. 1909-1909
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 78, No. 13_Supplement ( 2018-07-01), p. 1909-1909
    Abstract: Background: Nuclear receptor related 1 protein (NURR1/NR4A2) is a transcription factor that mediates numerous functions in differentiation/development, maintenance and metabolism. In cancer cells, NR4A2 promotes proliferation, migration, transformation and chemo-resistance in many cancers. Here, we explore the role of NR4A2 in head and neck squamous cell cancers (HNC) tumorigenesis and define its target genes. Methods: NR4A2 expression was determined by immunohistochemistry (IHC) and in publically available databases (Oncomine, TCGA). NR4A2 was knockedout/knockdown by CRISPR or shRNA. Coimmunoprepitation and reverse chromatin immunoprecipitation were done to study the interaction between EGFR/STAT3. ChIPSeq analysis was done by using Illumina NextSeq 500 Genome Analyzer and integrated with microarray (HPV+/- patients) to identify the true target genes of NR4A2 using BETA Software and validated by PCR/qPCR. Results: IHC and database analysis demonstrated overexpression of NR4A2 in HNC patients. NR4A2 was significantly overexpressed in HPV+ HNC patients, SCC47 and SCC104 cell lines. Functionally, CRISPR/shRNA knockout/knockdown of NR4A2 in cell lines significantly decreased colonogenicity, proliferation and migration. Expression of NR4A2 was positively correlated with EGFR expression (R2=0.92), and EGF treatment (100ng/ml; 24-48h) induced NR4A2 (~2 fold) expression coupled with epithelial to mesenchymal transition in SCC1 and SCC10B cells (HPV-). We also observed EGF mediated NR4A2 induction was abrogated by panEGFR inhibitor (afatinib) with concomitant decrease in STAT3 expression. Physical interaction between EGFR and STAT3 with EGF treatment regulated NR4A2 expression. Whole genome integration of ChIPseq and microarray demonstrates 9% (2015) and 23% (3002) of the enriched up and down-regulated target genes, respectively were canonically regulated in HPV+ HNC. ChIP PCR validation indicates enrichment of NR4A2 in some of the target genes, wnt10, p53, cxcl13, cln10 and hoxb8 in HPV+ cells. Conclusions: Our data indicate overexpression of NR4A2 in HNC, and its positive correlation with EGFR expression. We also noted the physical interaction between EGFR and STAT3 which functionally attribute the NR4A2 expression. Integration and validation data showed the differential enrichment of NR4A2 target genes in HPV+/- cell lines which suggest its possible discriminatory role in HNC pathogenesis which needs to be confirmed. Citation Format: Sanjib Chaudhary, Ramesh Pothuraju, Pranita Atri, Satyanarayana Rachagani, Surinder K. Batra, Muzafar A. Macha. NR4A2 role in head and neck cancer: Mechanistic and functional analysis [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2018; 2018 Apr 14-18; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2018;78(13 Suppl):Abstract nr 1909.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2018
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. More information can be found here...