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
Filter
  • American Association for Cancer Research (AACR)  (12)
  • 1
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 73, No. 8_Supplement ( 2013-04-15), p. LB-79-LB-79
    Abstract: Background: The EGFR monoclonal antibody cetuximab is approved for treating head and neck squamous cell cancer (SCCHN) in combination with radiation therapy for locally advanced disease and chemotherapy for recurrent and metastatic disease. Intrinsic or compensatory HER3 signaling and sustained PI3K/Akt activation may play a role in resistance to EGFR-targeted therapy. However, the role of HER3 in resistance to EGFR-targeted therapy in SCCHN has not been elucidated. Therefore, we first tested whether the combination of MM-121/SAR 256212 (M), a HER3 antibody being co-developed by Merrimack Pharmaceuticals and Sanofi, with cetuximab (C) may be more effective than M or C alone in treatment of SCCHN. Methods: We screened for the expression of EGFR, HER3 and their activated forms in 12 SCCHN cell lines. We then evaluated the activity of C, M, and their combination (CM) both in vitro by colony formation assay and in vivo by using SCCHN xenograft models (Tu212 and SCC47). In the in vivo experiment, mice were treated by intraperitoneal injection (i.p.) of C, M or CM twice per week for 4 weeks. The experimental groups include a PBS control, C (6.25ug/dose), M at low dose (300ug/dose, LD), M at high dose (600ug/dose, HD), the combination with LD (CMLD), and the combination with HD (CMHD). In addition, we examined the effect of M and C on HER3 and its downstream pathways in SCCHN cell lines by western blot assay. Results: pHER3 was detected in 9/12 while pEGFR, EGFR and HER3 were expressed in all of 12 SCCHN cell lines. The colony formation assay showed that Tu212 cells were significantly inhibited by CM compared to the control (p & lt;0.001), M (p & lt;0.001) and C (p=0.009) alone. Similar results were also observed using the SCC47 cell line. Western blot analysis confirmed the down-regulation of activated HER3 and its downstream phospho-Akt and phospho-S6 ribosomal protein by CM. Our in vivo study showed significant tumor growth inhibition in both MM-121 LD (p & lt;0.001) and MM-121HD (p & lt;0.001) compared to the control. Moreover, the treatment with both CMLD and CMHD significantly suppressed Tu212 xenograft tumor growth compared to the PBS control (both p & lt;0.0001), C alone (both p & lt;0.0001), and MLD (p=0.0046 and 0.0008, respectively), but only CMHD showed significant inhibition compared to MHD (p = 0.02). There was no significant difference between CMLD and CMHD (p=0.5994). Studies using another SCCHN xenograft model (SCC47) showed a similar inhibitory effect from the treatments by C, M and CM. Conclusion: This study shows that the combination of cetuximab and MM-121 is significantly more active than MM-121 or cetuximab alone in models of head and neck cancer in which both EGFR and HER3 are activated. Further studies will be expanded to understand the underlying mechanisms of this combination. (This study was supported by Merrimack Pharmaceuticals Inc.) Citation Format: Ning Jiang, Dongsheng Wang, Zhongliang Hu, Aminur M. Rahman, Hongzheng Zhang, Ruhul A. Amin, Xiaojing Wang, Zhengjia Chen, Shin M. Dong, Gabriela Garcia, Gavin MacBeath, Jun Ma, Fadlo R. Khuri, Nabil F. Saba, Georgia Z. Chen. Combined treatment with HER3 antibody MM-121/SAR 256212 and EGFR antibody cetuximab in pre-clinical models of head and neck cancer. [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 LB-79. doi:10.1158/1538-7445.AM2013-LB-79
    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 ...
  • 2
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2022
    In:  Cancer Research Vol. 82, No. 12_Supplement ( 2022-06-15), p. 1837-1837
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 82, No. 12_Supplement ( 2022-06-15), p. 1837-1837
    Abstract: Actinomycin D (ActD) was the first anti-cancer antibiotic approved for the management of human cancers. However, the notorious toxicity profile limits its widespread application in cancers including cancers of the aerodigestive tract. Recent studies show that combining low-dose ActD with existing chemotherapies could potentially reduce the toxicity of chemotherapy drugs through p53 mediated cyclotherapy (protection of normal cells while eliminating cancer cells), a recently coined concept. An understanding of ActD’s effect on p53 signaling is critical for meaningful application of ActD in cyclotherapy-based combinations. This study evaluated the antitumor efficacy and mechanism of action of ActD in aerodigestive tract cancers. We found that ActD strongly inhibited the growth of a panel of aerodigestive tract cancer cell lines and induced efficient apoptosis although the sensitivity varies among cell lines. The IC50 values of ActD spanned between 0.021-2.96nM. Mechanistic studies revealed that ActD time- and dose-dependently increased the expression of total and phosphorylated p53 (ser15). Furthermore, ActD-induced apoptosis is dependent on p53 in cells expressing wild-type p53. Further studies demonstrated that ActD induced context-dependent differential expression of downstream targets p21 and PUMA without significant effects on p27. Interestingly, our study revealed that p53-p21 is the predominant pathway activated by low-dose ActD in cells with wild type p53. Therefore, our findings further rationalize the premise for future studies to explore the application of low-dose ActD as a chemoprotectant for cyclotherapy combinations in aerodigestive tract cancers. (Supported by NIH Grant P20GM103434 to the West Virginia IDeA Network for Biomedical Research Excellence) Citation Format: Adeoluwa A. Adeluola, A. R. M Ruhul Amin. Low dose actinomycin D preferentially activates p53-p21 pathway in aerodigestive tract cancers: Implication for cyclotherapy [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2022; 2022 Apr 8-13. Philadelphia (PA): AACR; Cancer Res 2022;82(12_Suppl):Abstract nr 1837.
    Type of Medium: Online Resource
    ISSN: 1538-7445
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2022
    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
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2011
    In:  Cancer Research Vol. 71, No. 8_Supplement ( 2011-04-15), p. 5425-5425
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 71, No. 8_Supplement ( 2011-04-15), p. 5425-5425
    Abstract: Curcumin, 1, 7-bis (4-hydroxy-3-methoxyphenyl)-1, 6-heptadien-3, 5-dione, is the major bioactive compound isolated from the rhizome of Curcuma longa (turmeric). Despite its high margin of safety (no dose-limiting toxicity at doses up to 10 g/day in human) and efficacy against various types of cancers, the potential utility of curcumin as a chemopreventive/chemotherapeutic drug is compromised by its low bioavailability and poor selectivity. To circumvent these problems, more potent and selective curcumin analogues have been introduced. In the current study, we investigated one such synthetic analogue, (1E, 4E)-1, 5-bis (2, 4, 6-trimethoxyphenyl) penta-1, 4-dien-3-one (FLLL-22), against the head and neck cancer cell line Tu212 and lung cancer cell line H460. We conducted an SRB assay to determine IC50 values which showed that FLLL-12 was 24.6-fold and 31.6-fold more potent than natural curcumin against Tu212 and H460 cells, respectively. FLLL-22 also more strongly inhibited colony formation in both cell lines than that of natural curcumin. FLLL-22 at 1 μM was sufficient to completely inhibit colony formation, whereas up to 5 μM of curcumin had only minimal effects on colony formation. FLLL-22 also induced apoptosis more strongly (∼10-fold) than natural curcumin as evidenced by annexin V-PE staining and cleavage of PARP. Interestingly, FLLL-22, but not natural curcumin, activates DNA double strand breaks as evidenced by phosphorylation of H2AX and activation of p53 pathway. Although FLLL-22 strongly phosphorylated H2AX at doses of 3 and 5 μM, no H2AX phosphorylation was observed after curcumin treatment with doses 20 and 30 μM. At these doses, both compounds induced comparable apoptosis. FLLL-22 increased the protein level of p53, its phosphorylation at Ser15 and Ser392, and downstream target p21. A slight increase in p53 expression was observed with 20 μM curcumin, but not with 30 μM. Ablation of p53 by shRNA inhibited FLLL-22-induced p21, suggesting that FLLL-22 activates p53-dependent transcription. We also found that treatment with both FLLL-22 and curcumin inhibited phosphorylation of AKT, S6 and expression of Bcl-2. Taken together, our data strongly suggest that FLLL-22 is a potent curcumin analogue which activates the DNA damage pathway to induce apoptosis and might be a suitable anti-cancer drug for further preclinical and clinical development (supported by U01CA101244, R01CA112643, and P50CA128613 to DMS). Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 102nd Annual Meeting of the American Association for Cancer Research; 2011 Apr 2-6; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2011;71(8 Suppl):Abstract nr 5425. doi:10.1158/1538-7445.AM2011-5425
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2011
    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. 73, No. 8_Supplement ( 2013-04-15), p. 3189-3189
    Abstract: Curcumin is the major bioactive compound isolated from the rhizome of Curcuma longa (turmeric). Despite its high margin of safety (no dose-limiting toxicity at doses up to 10 g/day in human) and efficacy against various types of cancers, the potential utility of curcumin as a chemopreventive/chemotherapeutic drug is compromised by its low bioavailability and poor selectivity. To circumvent these problems, more potent and selective curcumin analogues have been introduced. In the current study, we investigated one such synthetic analogue, FLLL-12, which was reported to exhibit potent anti-tumor activity against prostate, colon and breast cancer cell lines. However, its mechanism of growth inhibition has yet to be elucidated. Methods: MSK-Leuk1 is an oral premalignant cell line. Other cell lines used in the study are fully transformed head and neck cancer cell lines isolated from cancer patients. Genotyping was conducted to confirm the identity of the cells. SRB assay was used for measuring growth inhibition. Annexin V staining was conducted for apoptosis assay. Expression of mRNAs and proteins were measured by RT-PCR and Western blotting, respectively. Result: IC50 values and apoptosis assay results suggested that FLLL-12 was 10-25-fold more potent than natural curcumin against head and neck cancers. FLLL-12 also strongly inhibited the expression of p-EGFR, p-AKT, and Bcl-2. Overexpression of constitutively active AKT or Bcl-2 significantly inhibited FLLL-12 induced apoptosis. Interestingly, FLLL-12 not only inhibited phosphorylated EGFR and AKT but also total EGFR and AKT proteins. Mechanistic studies revealed that FLLL-12 had an insignificant effect on mRNA expression of EGFR, AKT and Bcl-2 as compared to protein expression. Pretreatment with the proteasome inhibitor MG132 failed to rescue the expression of EGFR, AKT and Bcl-2 proteins inhibited by FLLL-12 treatment. Moreover, FLLL-12 failed to inhibit EGFR and AKT protein expression or induction of apoptosis when global protein synthesis was inhibited by cyclohexamide pretreatment. These results suggest that FLLL-12 inhibited the expression of EGFR, AKT and Bcl-2 at the translational level. Finally, FLLL-12 strongly inhibited phosphorylation of mTOR and its downstream targets pS6 and p-4E-BP1 which are important for cap-dependent protein translation. Conclusion: Our results strongly suggest that FLLL-12 is a potent curcumin analog which induced apoptosis of head and neck cancer cell lines by inhibiting cap-dependent translation of survival proteins including EGFR, AKT and Bcl-2 by inhibiting mTOR-mediated protein translational pathways. Future in vivo studies using appropriate animal models are warranted for further development of this promising compound for cancer prevention and treatment. (Dr. Amin's research is supported by R03CA159369 and Robbins Scholar Award). Citation Format: A.R.M. Ruhul Amin, Mohammad A. Rahman, Dongsheng Wang, Fadlo R. Khuri, James R. Fuchs, Zhuo G. Chen, Dong M. Shin. Potent curcumin analogue FLLL-12 targets protein translational pathways to inhibit EGFR, AKT and Bcl-2: potential role in apoptosis induction. [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 3189. doi:10.1158/1538-7445.AM2013-3189
    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 ...
  • 5
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2015
    In:  Cancer Research Vol. 75, No. 15_Supplement ( 2015-08-01), p. 29-29
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 75, No. 15_Supplement ( 2015-08-01), p. 29-29
    Abstract: Background: Drug-associated toxicity is one of the major challenges in the management of cancer patients. Unlike chemotherapy drugs, the safety of natural compounds such as curcumin has been well established. However, the potential use of curcumin in cancer treatment has been compromised by its low bioavailability, limited tissue distribution, and rapid biotransformation leading to low efficacy. To circumvent these problems, more potent and bioavailable analogs have been synthesized. In the current study, we investigated the mechanism of anti-tumor effect of one such analog, FLLL12, in lung cancer cells. Methods: A panel of premalignant and malignant lung cancer cell lines was used for the study. SRB assay was used to measure cell growth inhibition and IC50. Annexin-V staining was conducted for apoptosis assay. Expression of mRNAs and proteins were measured by RT-PCR and Western blotting, respectively. Small molecule chemical inhibitors and siRNA-mediated knockdown strategies were used to inactivate and shut down the expression of the relevant proteins, respectively. Results: IC50 values (0.63-1.67 μM for FLLL12 as compared to 6.06-12.4 μM for curcumin, depending on the cell lines) and apoptosis results (annexin V staining and cleavage of PARP and caspase 3) suggest that FLLL12 is 5-10-fold more potent than curcumin against lung cancer cells. Moreover, FLLL12 induced the expression of death receptor-5 (DR5). Ablation of the expression of the components of the extrinsic apoptotic pathway (DR5, caspase 8 and BID) significantly protected cells from FLLL12-induced apoptosis as evidenced by reduced annexin V staining (p = 0.0008, p = 0.0001 and p = 0.0007 for DR5, caspase 8 and BID, respectively) and cleavage of PARP and caspase 3. Analysis of mRNA expression by RT-PCR revealed that FLLL-12 had no significant effect on the expression of DR5 mRNA. Interestingly, inhibition of global phosphatase activity by phosphatase inhibitor cocktail (PIC) completely abolished DR5 expression and significantly inhibited apoptosis (p = 0.0007 and p = 0.001, respectively) and the cleavage of casepase-3 and PARP. Similarly, inhibition of protein tyrosine phosphatases (PTPs) by sodium orthovanadate, but not by the alkaline phosphatase inhibitor imidazole, inhibited DR5 expression, apoptosis (p = 0.006) and cleavage of caspase-3 and PARP, suggesting the involvement of PTPs in the regulation of DR5 expression. FLLL12 also induced the expression of p53 and p73. However, inactivation of these proteins with their dominant negative construct or siRNA had no significant effects on apoptosis induction. Conclusions: Our results strongly suggest that FLLL12 induces apoptosis of lung cancer cell lines by posttranscriptional regulation of DR5 through activation of protein tyrosine phosphatase(s). This study was supported by NCI R03 CA171663, NCI P50 CA128613 and Robbins Scholar Award of Winship Cancer Institute of Emory University. Citation Format: Abedul Haque, Mohammad A. Rahman, James R. Fuchs, Zhuo G. Chen, Fadlo R. Khuri, Dong M. Shin, A.R.M. Ruhul Amin. Potent curcumin analog FLLL12 induces apoptosis in lung cancer cells through death receptor-5-dependent pathway. [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 29. doi:10.1158/1538-7445.AM2015-29
    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 ...
  • 6
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2022
    In:  Cancer Prevention Research Vol. 15, No. 12_Supplement_2 ( 2022-12-01), p. A004-A004
    In: Cancer Prevention Research, American Association for Cancer Research (AACR), Vol. 15, No. 12_Supplement_2 ( 2022-12-01), p. A004-A004
    Abstract: Head and neck cancer (HNC) is a devastating disease and the 6th most common cancer worldwide. Most HNC patients are diagnosed with advanced stage disease for which the 5-year survival is below 50%, stressing the need for chemoprevention. Recently, we have reported that the combination of resveratrol and EGCG induces synergistic apoptosis and inhibits xenografted HNC growth by inhibiting AKT-mTOR pathway. In this study, we have investigated the chemopreventive efficacy of resveratrol, EGCG and their combination using 4NQO-induced oral carcinogenesis model. C57BL/6 mice were exposed to 4-NQO (50 μg/ml) via drinking water for 10 weeks, followed by treatment with vehicle (50% sweetened condense milk), resveratrol (30 mg/kg), EGCG (30 mg/kg) and their combination for 8 weeks, 5 days/week. The mice were sacrificed on week 24 and the number of visible and microscopic lesions were counted. Resveratrol alone and in combination with EGCG significantly inhibited the number of visible lesions whereas the number of microscopic lesions and lesion area were significantly inhibited only in the combination group. Furthermore, RNASeq and qPCR analysis using a HNC cell line identified GDF15, ATF3, p21, p27 and Bim as significantly upregulated genes with GDF15 being the most upregulated one. Expression of GDF15 and ATF3 proteins were confirmed by western blotting. Taken together, our data strongly demonstrate the chemopreventive potential of the combination of EGCG and resveratrol and paves the way for further clinical developments. (Supported by NIH Grant P20GM103434 to the West Virginia IDeA Network for Biomedical Research Excellence) Citation Format: Adeoluwa Adeluola, Lukmon Raji, Saroj Sigdel, A.S.M. Anisuzzaman, A. R. M. Ruhul Amin. Chemoprevention of head and neck cancers by the combination of epigallocatechin gallate (EGCG) and resveratrol [abstract]. In: Proceedings of the Second Biennial NCI Meeting: Translational Advances in Cancer Prevention Agent Development (TACPAD); 2022 Sep 7-9. Philadelphia (PA): AACR; Can Prev Res 2022;15(12 Suppl_2): Abstract nr A004.
    Type of Medium: Online Resource
    ISSN: 1940-6215
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2022
    detail.hit.zdb_id: 2422346-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 7
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2010
    In:  Cancer Research Vol. 70, No. 8_Supplement ( 2010-04-15), p. 3677-3677
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 70, No. 8_Supplement ( 2010-04-15), p. 3677-3677
    Abstract: Inhibition of specific genes by small interfering RNAs (siRNA) offers a promising therapeutic approach in oncology. However, siRNA therapy is hindered by poor stability under physiological conditions and limited intracellular uptake. Ribonucleotide reductase subunit M2 (RRM2) is an essential protein for DNA synthesis and is responsible for the reduction of ribonucleotides to deoxyribonucleotides, providing a balanced supply of precursors for DNA synthesis and repair. In this study we investigated the efficacy and mechanism of action of a targeted nanoparticle formulation of siRNA against RRM2. The clinical version of this nanoparticle is denoted as CALAA-01 and is currently used in a Phase I clinical trial. The nanoparticle consists of a cyclodextrin-containing polymer, a polyethylene glycol steric stabilization agent, and human transferrin as a targeting ligand for binding to transferrin receptors (TfR) that are typically up regulated in cancer cells. Initially, we screened several head and neck squamous cell carcinoma (HNSCC) and lung cancer cell lines and found various degrees of expression of TfR and RRM2. Suppression of RRM2 by transfection of 5 nM siRNA for 72 hours in vitro strongly inhibited the cell growth in HNSCC (Tu212∼75% and M4e∼60%) and in lung cancer cell lines (A549 and H460 ∼80%). Using xenograft (Tu212) as an in-vivo model, we found nanoparticles (10mg/Kg) injected via the tail vein in four dosing schedules (Day 1, 3, 8 and 10) significantly reduced RRM2 expression and its activity in xenograft tumors. RRM2 activity was reduced by an average 1.88-fold in the RRM2 siRNA treated group compare to the formulated control siRNA group (p=0.04). We monitored tumor growth in treated and control groups (8 mice in each group) for 28 days and found that tumor growth reduced by an average 2.9- fold in the treated group compared to formulated control siRNA (p=0.004). The nanoparticle delivered RRM2 siRNA significantly suppressed cell proliferation and induced apoptosis as evidenced by xenograft tumor tissue staining with Ki67 and terminal deoxynucleotidyl transferase dUTP nick and labeling, respectively. Mechanistic studies (in-vitro) have revealed siRNA-mediated suppression of RRM2 increases apoptosis in a p53-independent manner. Interestingly, suppression of RRM2 by 5nM siRNA for 72 hours increased activation of caspase 9 and caspase 3 at least 2-fold with concomitant increase in the levels of cleaved poly-ADP-ribose polymerase (PARP), thus suggesting a vital role for p73 in the observed apoptosis through an intrinsic pathway. Thus this study demonstrates that targeted nanoparticles can deliver RRM2 siRNA to head and neck tumors in mice and highlights RRM2 as an excellent target to induce potent apoptosis and tumor growth inhibition. (Supported by NIH/NCI grant U54CA119338-04) Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 101st Annual Meeting of the American Association for Cancer Research; 2010 Apr 17-21; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2010;70(8 Suppl):Abstract nr 3677.
    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 ...
  • 8
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2014
    In:  Cancer Research Vol. 74, No. 19_Supplement ( 2014-10-01), p. 2283-2283
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 74, No. 19_Supplement ( 2014-10-01), p. 2283-2283
    Abstract: Background: Curcumin is the major bioactive compound isolated from the rhizome of Curcuma longa (turmeric). Despite its high margin of safety and efficacy against various types of cancers, the potential utility of curcumin as a chemopreventive/chemotherapeutic drug is compromised by its low bioavailability and poor selectivity. To circumvent these problems, more potent and selective curcumin analogues have been developed. In the current study, we investigated the mechanism of apoptosis of one such synthetic analogue, FLLL-12, against lung cancers. This compound was reported to exhibit potent anti-tumor activity against prostate, colon and breast cancer cell lines. However, its mechanism of growth inhibition has yet to be elucidated. Methods: Multiple premalignant and fully transformed lung cancer cell lines were used throughout the study. SRB assay was used to measure cell growth inhibition. Annexin V staining was conducted for apoptosis assay. Expression of mRNAs and proteins were measured by RT-PCR and Western blotting, respectively. Gene overexpression and knockdown strategies were used to activate or shut down of specific proteins. Results: IC50 values and apoptosis assay results showed that FLLL-12 was ∼10-fold more potent than the natural parent compound, curcumin, against lung cancer cells. Further mechanistic studies revealed that FLLL-12 induced the expression of DR5, inhibited the protein expression of EGFR, p-AKT, AKT and Bcl-2 and increased the expression of BIM. Analysis of mRNA expression suggested that FLLL-12 strongly inhibited the mRNA expression of EGFR and AKT, whereas the expression of Bcl-2 and Bim mRNAs remained unchanged. FLLL-12 also induced the expression of p53 and p73. However, inactivation of these proteins with their dominant negative construct or siRNA had no significant effects on apoptosis induction. Conclusions: Our results strongly suggest that FLLL-12 is a potent curcumin analog that induces apoptosis of lung cancer cell lines by targeting: (1) intrinsic pathways via transcriptional inhibition of EGFR and AKT and induction of BIM, and (2) extrinsic pathway via induction of DR5. Future in vivo studies using appropriate animal models are warranted for further development of this promising compound for cancer prevention and/or treatment for lung cancer. (Supported by R03CA171663, P50CA128613 and Robbins Scholar Award of Winship Cancer Institute of Emory University). Citation Format: A.R.M. Ruhul Amin, Abedul Haque, Mohammad A. Rahman, James R. Fuchs, Zhuo G. Chen, Dong M. Shin. Potent curcumin analog FLLL-12 targets both intrinsic and extrinsic signaling pathways to induce apoptosis in lung cancers. [abstract]. In: Proceedings of the 105th Annual Meeting of the American Association for Cancer Research; 2014 Apr 5-9; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2014;74(19 Suppl):Abstract nr 2283. doi:10.1158/1538-7445.AM2014-2283
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2014
    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 ...
  • 9
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 74, No. 19_Supplement ( 2014-10-01), p. 2269-2269
    Abstract: Purpose: Effective chemopreventive strategies could save millions of lives. We have previously reported that the combination of the EGFR-TKI erlotinib and the green tea constituent epigallocatechin-3-gallate (EGCG) exhibited synergistic chemopreventive effects in head and neck cancers by inducing the expression of Bim, p21, p27, and inhibiting the phosphorylation of ERK and AKT and expression of Bcl-2. In the current study, we further investigated the mechanism of regulation of Bim, Bcl-2, p21 and p27, and their role in apoptosis. Methods: Well characterized and genetically validated squamous cell carcinoma of the head and neck cell lines were used throughout the study. Annexin V staining was conducted for apoptosis assay. Expression of mRNAs and proteins were measured by RT-PCR and Western blotting, respectively. Gene overexpression and knockdown strategies were used to activate or shut down the expression of specific proteins. Results: siRNA-mediated silencing of Bim significantly inhibited apoptosis induced by the combination of erlotinib and EGCG (p=). On the other hand, overexpression of Bcl-2 significantly protected cells from apoptosis (p=), whereas overexpression of constitutively AKT had minimal effect on apoptosis (actual values). Analysis of mRNA expression by RT-PCR revealed that neither erlotinib, EGCG nor their combination had any significant effects on the mRNA expression of Bim, p21, p27 or Bcl-2, suggesting post-transcriptional regulation of Bim, p21, p27 and Bcl-2 by the combination of erlotinib and EGCG. Furthermore, we found that erlotinib or the combination of EGCG and erlotinib inhibited the phosphorylation of Bim, and that inhibition of protein translation by cycloheximide pretreatment stabilized Bim, suggesting posttranslational regulation of Bim. Conclusion: Our results strongly suggest that the combination of erlotinib and EGCG induces apoptosis of SCCHN cells by regulating Bim and Bcl-2 at the post-translational level. Currently, a clinical trial is underway at Winship Cancer Institute of Emory University to inhibit or reverse the progression of oral premalignant lesions using this combination. (This study is supported by R03CA159369, P50CA128613 and Robbins Scholar Award of Winship Cancer Institute). Citation Format: Abedul Haque, Mohammad A. Rahman, Zhuo G. Chen, Dong M. Shin, A.R.M. Ruhul Amin. Combination of erlotinib and epigallocatechin-3-gallate induces apoptosis of squamous cell carcinoma of the head and neck through posttranslational regulation of Bim and Bcl-2. [abstract]. In: Proceedings of the 105th Annual Meeting of the American Association for Cancer Research; 2014 Apr 5-9; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2014;74(19 Suppl):Abstract nr 2269. doi:10.1158/1538-7445.AM2014-2269
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2014
    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 ...
  • 10
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2022
    In:  Cancer Research Vol. 82, No. 12_Supplement ( 2022-06-15), p. 3245-3245
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 82, No. 12_Supplement ( 2022-06-15), p. 3245-3245
    Abstract: Background: In recent years, oncology research has led to the discovery of targeted therapies aimed at specific genetic abnormalities unique to cancer cells. In particular, the overexpression of epidermal growth factor receptor (EGFR) and constitutive activation of the phosphoinositide-3-kinase/protein kinase B (PI3K/AKT) pathway are common aberrations in head and neck cancer. However, intrinsic and acquired resistance are continuously posing challenges to success. Previously, we reported that the combination of EGFR inhibitor erlotinib with PI3K inhibitor BKM120 had synergistic effects that induced efficient apoptosis in a panel of head and neck cancer cell lines, but some cells were still resistant. The purpose of the current study is to understand the mechanism of resistance to EGFR and PI3-K co-targeting. Hypothesis: Src-Met signaling confers apoptosis resistance to EGFR and PI3K cotargeting by regulating downstream genes expresiion. Methods: Apoptosis was measured by annexin V-PE staining. Western blotting and qPCR were used for measuring the expression of proteins and mRNA, respectively. RNASeq was conducted from RNA samples treated with (Erlotinib + BKM120) ± dasatinib (Src inhibitor)/crizotinib(Met inhibitor). Results: Biochemical studies revealed that JHU022 cells, which were resistant to the combination treatment, had an overactivation of c-Met receptor tyrosine kinase that was regulated by Src kinases. Pharmacological or genetic inactivation of both Met or Src kinase sensitized this cell line to apoptosis induced by EGFR and PI3K co-targeting. RNA-Seq analysis identified that a total of 291 genes were modulated (2-folds) by both crizotinib and dasatinib. Pathway enrichment suggested that many of these genes were associated with cell cycle, DNA replication, and cellular response to DNA damage. CPA4 was identified as the most downregulated gene and GDF15 as one of the top upregulated genes. Conclusions: SRC-Met signaling confers resistant to EGFR and PI3K cotargeting by modulating genes associated with cell cycle, DNA replication, and cellular response to DNA damage. Citation Format: Adeoluwa A. Adeluola, A.R.M. Ruhul Amin. Genes associated with Src-Met driven resistance of head & neck cancers to EGFR-PI3K cotargeting [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2022; 2022 Apr 8-13. Philadelphia (PA): AACR; Cancer Res 2022;82(12_Suppl):Abstract nr 3245.
    Type of Medium: Online Resource
    ISSN: 1538-7445
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2022
    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...