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: Molecular Cancer Therapeutics, American Association for Cancer Research (AACR), Vol. 18, No. 12_Supplement ( 2019-12-01), p. B107-B107
    Abstract: Background: PI3K/mTOR pathway is mutated in 10-20% of colorectal cancer (CRC) specimens and has been associated with poor survival. Phosphatidic acid (PA) is a central lipid membrane metabolite and lipid second messenger which has been shown to target mTOR. It is thought that PA lipid signaling to mTOR in part promotes mTOR mediated cancer cell growth, proliferation and survival. Diacylglycerol kinases (DGKs) are one of several mechanisms of PA generation. In this study, we found diacylglycerol kinases to be synthetically lethal in mTOR inhibitor resistant CRC. We evaluated the anti-proliferative, pharmacodynamic and metabolic effects of dual inhibition with mTOR (TAK-228) and DGK (ritanserin and R59022) inhibitors. Methods: A synthetic lethal screen was performed with two TAK-228 resistant colorectal cancer cell lines (HCT116 and SW620). Subsequent experiments were performed with one TAK-228 sensitive (DLD1) and one resistant (HCT116) CRC cell lines. Efficacy of TAK-228/ritanserin and TAK-228/R59022 combination therapy was evaluated with CellTiter-Glo cell viability and clonogenic colony formation assays. Global metabolomics profiling of DLD1 and HCT116 cells upon treatment with TAK-228, R59022, and in combination was performed using ultra high pressure liquid chromatography coupled to mass spectrometry. Pharmacologic DGK inhibition was phenocopied using lentiviral shRNA knockdown of DGKα. Immunoblotting was performed to evaluate mechanism of action of TAK-228 combination therapy. Results: TAK-228 combined with ritanserin and R59022 demonstrated decreased cell viability and colony formation as compared to either single agent. Immunoblotting confirmed TAK-228 abrogates PI3K/mTOR pathway activity. DGK inhibition alone resulted in a compensatory activation of mTOR signaling. DGK inhibition disrupted the phosphatidic acid pathway in DLD1 and HCT116 as evidenced by a decrease in PA synthesis and elevation of glycerol 3-phosphate levels, respectively: altering energy metabolism. Specifically, in HCT116, glucose utilization, glutaminolysis, and Krebs cycle anaplerosis were elevated; while one carbon metabolism was decreased. Lentiviral shRNA transduction resulted in DGKα knockdown as evaluated by RT-PCR and immunoblot. Phenocopy combination therapy with TAK-228 and DGKα knockdown resulted in an increased sensitivity to mTOR inhibition compared to mock transduced control. Conclusions: Pharmacologic and shRNA knockdown inhibition of DGK in combination with mTOR inhibition resulted in decreased cancer cell viability and decreased colony formation. Pharmacologic inhibition of mTOR and DGK, alone or in combination, alter metabolic wiring in crucial pathways such as energy metabolism, nucleotide biosynthesis, and the generation of lipid precursors. Impaired phosphatidic acid production may sensitize cells to mTOR inhibition. These results suggest a therapeutic anti-cancer advantage of simultaneously targeting lipid signaling/metabolism via diacylglycerol kinases and mTOR. Citation Format: Peter J Klauck, Hayley J Hawkins, Madison Weber, Stacey M Bagby, Christopher H Lieu, Sarah J Hartman, Betelehem W Yacob, Kelly D Sullivan, Monica Brown, Julie A Reisz, Angelo D’Alessandro, Wells A Messersmith, S Gail Eckhardt, Todd M Pitts. Metabolic reprogramming enhances the efficacy of mTOR inhibition in colorectal cancer [abstract]. In: Proceedings of the AACR-NCI-EORTC International Conference on Molecular Targets and Cancer Therapeutics; 2019 Oct 26-30; Boston, MA. Philadelphia (PA): AACR; Mol Cancer Ther 2019;18(12 Suppl):Abstract nr B107. doi:10.1158/1535-7163.TARG-19-B107
    Type of Medium: Online Resource
    ISSN: 1535-7163 , 1538-8514
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2019
    detail.hit.zdb_id: 2062135-8
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    In: BMC Cancer, Springer Science and Business Media LLC, Vol. 20, No. 1 ( 2020-12)
    Abstract: Triple-negative breast cancer (TNBC) is an aggressive breast cancer subtype with limited systemic treatment options. RX-5902 is a novel anti-cancer agent that inhibits phosphorylated-p68 and thus attenuates nuclear β-catenin signaling. The purpose of this study was to evaluate the ability of β-catenin signaling blockade to enhance the efficacy of anti-CTLA-4 and anti-PD-1 immune checkpoint blockade in immunocompetent, preclinical models of TNBC. Methods Treatment with RX-5902, anti-PD-1, anti-CTLA-4 or the combination was investigated in BALB/c mice injected with the 4 T1 TNBC cell line. Humanized BALB/c-Rag2 null Il2rγ null SIRPα NOD (hu-CB-BRGS) mice transplanted with a human immune system were implanted with MDA-MB-231 cells. Mice were randomized into treatment groups according to human hematopoietic chimerism and treated with RX-5902, anti-PD-1 or the combination. At sacrifice, bone marrow, lymph nodes, spleen and tumors were harvested for flow cytometry analysis of human immune cells. Results The addition of RX-5902 to CTLA-4 or PD-1 inhibitors resulted in decreased tumor growth in the 4 T1 and human immune system and MDA-MB-231 xenograft models. Immunologic analyses demonstrated a significant increase in the number of activated T cells in tumor infiltrating lymphocytes (TILs) with RX-5902 treatment compared to vehicle ( p   〈  0.05). In the RX-5902/nivolumab combination group, there was a significant increase in the percentage of CD4+ T cells in TILs and increased systemic granzyme B production ( p   〈  0.01). Conclusions Conclusions: RX-5902 enhanced the efficacy of nivolumab in a humanized, preclinical model of TNBC. Several changes in immunologic profiles were noted in mice treated with RX-5902 and the combination, including an increase in activated TILs and a decrease in human myeloid populations, that are often associated with immunosuppression in a tumor microenvironment. RX-5902 also was shown to potentiate the effects of checkpoint inhibitors of CTLA4 and the PD-1 inhibitor in the 4 T-1 murine TNBC model. These findings indicate that RX-5902 may have important immunomodulatory, as well as anti-tumor activity, in TNBC when combined with a checkpoint inhibitor.
    Type of Medium: Online Resource
    ISSN: 1471-2407
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2020
    detail.hit.zdb_id: 2041352-X
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2022
    In:  Cancer Research Vol. 82, No. 12_Supplement ( 2022-06-15), p. 1063-1063
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 82, No. 12_Supplement ( 2022-06-15), p. 1063-1063
    Abstract: Background: Triple-negative breast cancer (TNBC) is a subtype of breast cancer that lacks the expression of the estrogen receptor, progesterone receptor, and human epidermal growth factor-2 over-expression. Compared to other subtypes of breast cancer, TNBC is associated with a higher risk for metastatic recurrence. While immunotherapy, PARP inhibitors, and sacituzumab govitecan have shown benefit in a subset of TNBC patients, chemotherapy with doxorubicin remains a mainstay of treatment. Senescence is a cellular phenomenon where cells are committed to an arrested state, however, senescent cells are able to secrete pro-tumorigenic factors which can help to promote tumor progression and invasion. It has been suggested that senescence is a potential mechanism of resistance in doxorubicin (dox) treated cells. Transitioning senescence mediated resistance to apoptosis is crucial to the treatment of TNBC. The objective of this study was to evaluate the combination BCL-2 and HDAC inhibitors with dox to overcome resistance and induce apoptosis. Experimental Procedures: TNBC cell lines resistant and sensitive to doxorubicin were identified using a CellTiter-Glo Viability Assay and resistant cell lines were selected for further analysis. To assess proliferation, dox resistant TNBC cell lines were plated in 96-well plates. Cells were exposed to vehicle control, dox, BCL-2 inhibitor, HDAC inhibitor or the combination of dox and BCL-2 inhibitor or HDAC inhibitor for 72 hours. Cellular proliferation was assessed using the BioSpa live cell analysis system. Apoptosis at 24 hours was analyzed by flow cytometry using Annexin V on cells treated in combination and as single agents. Immunoblotting was performed to evaluate the downstream effects of apoptosis and senescence of drugs as single agents and in combination. Results: The addition of a BCL-2 inhibitor and HDAC inhibitor to doxorubicin resulted in decreased proliferation in resistant TNBC cell lines compared to single agents and vehicle control by live cell microscopy. Furthermore, the combination of dox and a BCL-2 inhibitor resulted in increased apoptosis when compared to single agents and control using Annexin V staining. The cyclin dependent kinase inhibitors p21 and p16 were over-expressed in cells exposed to combination treatment. Apoptotic proteins BAD, PUMA, cleaved PARP and the anti-apoptotic protein BCL-2 were upregulated in cells treated with a BCL-2 inhibitor with or without dox. The pro-mitotic protein cyclin-B1 was downregulated in resistant cells treated with single agent BCL-2 inhibitor and in combination with dox. Additional mechanistic studies are ongoing. Conclusion: The combination of doxorubicin with BCL-2 and HDAC inhibitors resulted in decreased cellular proliferation and increased apoptosis. Potential senolytic drugs used with dox represent an exciting potential to overcome dox resistance and warrant further investigation. Citation Format: Anna R. Schreiber, Stephen Smoots, Betelehem W. Yacob, Adrian TA Dominguez, Cecilia Levandowski, Jennifer R. Diamond, Todd M. Pitts. Combination strategies to overcome doxorubicin induced senescence in triple-negative breast cancer [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 1063.
    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 ...
  • 4
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 79, No. 13_Supplement ( 2019-07-01), p. 4720-4720
    Abstract: Background: AZD0156 is an oral inhibitor of ATM, a serine threonine kinase that plays a key role in DNA damage response associated with DNA double strand breaks. Topoisomerase-I inhibitors like irinotecan induce single-strand DNA breaks, which are converted to double-strand breaks during DNA replication. Thus the combination of AZD0156 and irinotecan is a rational combination for clinical use. Irinotecan is used clinically to treat a variety of malignancies, including colorectal cancer (CRC), usually in combination with 5-fluorouracil (5FU) as FOLFIRI. An ongoing phase 1 clinical trial is evaluating AZD0156 in combination with single-agent irinotecan and FOLFIRI in patients with refractory cancers (NCT02588105). The purpose of this study is to evaluate AZD0156 in combination with irinotecan and 5FU in preclinical models of CRC to help inform clinical use. Methods: Anti-proliferative effects of single-agent AZD0156 and combination therapy with SN38 (active metabolite of irinotecan) and 5FU were evaluated in CRC cell lines using the Cell-Titer Glo assay. Immunoblotting and cell cycle analysis were performed to determine the mechanism of enhanced combination effects. Four CRC patient derived xenograft (PDX) models were treated with AZD0156, irinotecan, and 5FU alone and in combination for assessment of tumor growth inhibition (TGI). Results: An enhanced antiproliferative effect was observed with the combination treatment over either single agent. A more significant synergistic effect was demonstrated with the combination of AZD0156 and SN38 as compared with the combination of AZD0156 and 5FU. Cell cycle data demonstrated enhanced cell cycle arrest with combination therapy as compared to single agents. Immunoblotting results suggest a decrease in phosphorylated gamma-H2AX in cell lines treated with combination therapies. Increased TGI was observed in CRC PDX models treated with the combination of AZD0156 and irinotecan as compared to single-agent therapy in 3 of 4 models. There was not a significant change in TGI with the addition of 5FU for triplet therapy in the majority of models. Conclusions: The combination of AZD0156 with irinotecan is synergistic in in vitro models and is associated with increased TGI in CRC PDX in vivo models. The addition of 5FU to AZD0156 and irinotecan did not result in increased TGI as compared to doublet therapy in CRC PDX models, though did not decrease the AZD0156/irinotecan combination effect. An ongoing clinical trial is evaluating this combination in patients with cancers refractory to standard treatments (NCT02588105). Citation Format: S. Lindsey Davis, Marina I. Schlaepfer, Stacey M. Bagby, Sarah J. Hartman, Betelehem W. Yacob, Tonia Tse, Dennis M. Simmons, Jennifer R. Diamond, Christopher H. Lieu, Alexis D. Leal, Elaine B. Cadogan, Gareth D. Hughes, Stephen T. Durant, Wells A. Messersmith, Todd M. Pitts. Ataxia telangiectasia mutated (ATM) kinase inhibitor AZD0156 in combination with 5-fluorouracil and irinotecan in preclinical models of colorectal cancer [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 4720.
    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 ...
  • 5
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 80, No. 16_Supplement ( 2020-08-15), p. 6387-6387
    Abstract: Pancreatic ductal adenocarcinoma (PDAC) is currently the fourth leading cause of cancer deaths with more than 56,000 new cases estimated to be diagnosed in 2019. Current treatment options for PDAC include radiation and chemotherapeutic regimens, however these targeted therapies are ineffective for patients with advanced disease progression. Additionally, the dense stromal nature of PDAC tumors create challenges to target the cancer cells resulting in incomplete cell killing and eventual drug resistance. Recent evidence has shown that CPT1A, an enzyme that regulates the entry of lipids into mitochondria for β-oxidation, is strongly expressed in several cancers. CPT1A is located on the mitochondrial membrane and potentially interacts with BCL-2, an anti-apoptotic protein that promotes tumor maintenance and metastasis. Metabolic stress can activate the anti-apoptotic effects of BCL-2, reprograming metabolism to use fat oxidation for cancer survival. Therefore, a co-inhibition using the selective BCL-2 inhibitor, venetoclax, with agents that inhibit CPT1A and β-oxidation, could be a novel strategy for PDAC. There are few studies considering CPT1A as a therapeutic target for PDAC. Current available drugs to target these pathways include the anti-anginal ranolazine, and CPT1A inhibitors etomoxir and perhexiline. Previous studies have shown that expression of BCL-2 by tumor cells is necessary for BCL-2 inhibitors to be effective. We initially wanted to determine the expression of BCL-2 and CPT1A in PDAC cells utilizing western blot and rtPCR, and to confirm their proximity using a proximity ligation assay (PLA). PDAC cells were then plated in 96 well plates and Cell Titer-Glo assays were performed to determine effective concentrations of single agent venetoclax, etomoxir, and perhexiline. The effects of these drugs in combination were then evaluated using a clonogenic assay, which was analyzed using the ImageJ colony area plugin. PDAC cells were then exposed to the combinations and western blots were performed to evaluate changes downstream effectors. We have confirmed the expression of BCL-2 and CPT1A on the mitochondrial membrane using Westerns, rtPCR, and a PLA on several PDAC lines. Though single agent drugs had little effect on cell viability, the combination of venetoclax with CPT1A and β-oxidation inhibitors decreased colony formation in some PDAC cell lines. Western blot analysis revealed the drug combinations affected the phosphorylation of AKT and 4E-BP1 and expression of the pro-apoptotic protein BID. These data suggest that co-targeting BCL-2 and CPT1A have potential for anti-tumor effects in PDAC. Additional research into the role of CPT1A in PDAC biology will elucidate the optimal dosing concentrations and mechanisms for further studies. Citation Format: Sarah J. Hartman, Nathalie Nadales, Stacey M. Bagby, Betelehem W. Yacob, Brian L. Gittleman, Adriana Estrada-Bernal, Anh T. Le, Christopher H. Lieu, S. Lindsey Davis, Alexis D. Leal, Jennifer R. Diamond, Wells A. Messersmith, Isabel R. Schlaepfer, Todd M. Pitts. Therapeutic targeting of lipid oxidation and apoptosis in pancreatic ductal adenocarcinoma [abstract]. In: Proceedings of the Annual Meeting of the American Association for Cancer Research 2020; 2020 Apr 27-28 and Jun 22-24. Philadelphia (PA): AACR; Cancer Res 2020;80(16 Suppl):Abstract nr 6387.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2020
    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. 80, No. 16_Supplement ( 2020-08-15), p. 6647-6647
    Abstract: Immunotherapies that target immune regulatory checkpoints such as CTLA-4 and PD-1 are widely used among many cancer types and have shown positive results in CRC with high microsatellite instability. However, in microsatellite stable (MSS) CRC there is a dismal response rate of 0%. The limited efficacy has shown to be partially due to the lack of T-cells in the tumor microenvironment and/or no activation/regulation of paramount cells in the immune system. The Wnt pathway is the most commonly altered pathway in CRC and is highly involved in driving tumor initiation and progression. Recent evidence also demonstrates the Wnt pathway is involved in T-lymphocyte development, maturation/activation of CD8+ effector T cells and recruitment of dendritic cells. Therefore, targeting the Wnt pathway utilizing a Porcupine (PORCN) inhibitor (ETC-159) in MSS CRC may be a promising strategy to sensitize tumors to immune checkpoint inhibition. Human Immune System BRGS (BALB/c, Rag2−/−, IL2RγC−/−, NODSIRPα) mice were engrafted with MSS CRC PDX (hPDX). The hPDX were randomized according to human chimerism into the following drug treatments groups: Vehicle, ETC-159, nivolumab, and the combination. Treatments began when tumors reached 100-300mm3 and tumors were measured twice weekly. At the end of study, sera, lymph nodes, spleen, and tumor tissue were collected for immunohistochemistry, single cell suspensions, and flow cytometry analysis. Combination therapy resulted in a significant decrease in tumor volume compared to both single agents and vehicle. Flow cytometric analysis demonstrated an increase in human immune cells, in particular human CD4 and CD8 cells in the combination compared to the vehicle and nivolumab treated groups. Additionally, these T-cells showed increased signs of activation and effector function, as indicated by increased CD69+ expression, effector memory subsets, and granzyme B+ cells in the TILs, with a further reduction in Treg populations, suggesting an overall increase in inflammation. An increase in MHC II expression on tumor cells was observed in the ETC-159 single agent with a statistically significant increase in the combination treated tumors demonstrating enhanced antigen presentation. Furthermore, PD-1 expression was upregulated on CD4+ T-cells in the ETC-159 single agent. Lastly, VECTRA analysis corroborates the flow cytometry data showing a changing tumor immune landscape through an increase in CD4+ and CD8+ T cells in the tumor and surrounding stroma. Our data demonstrates the combination treatment of ETC-159 + nivolumab in MSS CRC hPDX show increased tumor infiltration of human immune cells. Further preclinical data is compulsory but these results support further development of this combination in clinical trials. Citation Format: Stacey M. Bagby, Sarah J. Hartman, Natalie M. Navarro, Betelehem W. Yacob, Jeremy Shulman, Jessica Barkow, Christopher H. Lieu, S. Lindsey Davis, Alexis D. Leal, Wells A. Messersmith, Angela Minic, Kimberly R. Jordan, Julie Lang, Todd M. Pitts. Sensitizing microsatellite stable colorectal cancer to immune checkpoint therapy utilizing Wnt pathway inhibition [abstract]. In: Proceedings of the Annual Meeting of the American Association for Cancer Research 2020; 2020 Apr 27-28 and Jun 22-24. Philadelphia (PA): AACR; Cancer Res 2020;80(16 Suppl):Abstract nr 6647.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2020
    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
    In: Endocrine-Related Cancer, Bioscientifica, Vol. 26, No. 10 ( 2019-10), p. 765-778
    Abstract: Adrenocortical carcinoma (ACC) is an aggressive orphan malignancy with less than 35% 5-year survival and 75% recurrence. Surgery remains the primary therapy and mitotane, an adrenolytic, is the only FDA-approved drug with wide-range toxicities and poor tolerability. There are no targeted agents available to date. For the last three decades, H295R cell line and its xenograft were the only available preclinical models. We recently developed two new ACC patient-derived xenograft mouse models and corresponding cell lines (CU-ACC1 and CU-ACC2) to advance research in the field. Here, we have utilized these novel models along with H295R cells to establish the mitotic PDZ-binding kinase (PBK) as a promising therapeutic target. PBK is overexpressed in ACC samples and correlates with poor survival. We show that PBK is regulated by FOXM1 and targeting PBK via shRNA decreased cell proliferation, clonogenicity and anchorage-independent growth in ACC cell lines. PBK silencing inhibited pAkt, pp38MAPK and pHistone H3 altering the cell cycle. Therapeutically, targeting PBK with the small-molecule inhibitor HITOPK032 phenocopied PBK-specific modulation of pAkt and pHistone H3, but also induced apoptosis via activation of JNK. Consistent with in vitro findings, treatment of CU-ACC1 PDXs with HITOPK032 significantly reduced tumor growth by 5-fold ( P   〈  0.01). Treated tumor tissues demonstrated increased rates of apoptosis and JNK activation, with decreased pAkt and Histone H3 phosphorylation, consistent with effects observed in ACC cell lines. Together these studies elucidate the mechanism of PBK in ACC tumorigenesis and establish the potential therapeutic potential of HITOPK032 in ACC patients.
    Type of Medium: Online Resource
    ISSN: 1351-0088 , 1479-6821
    Language: Unknown
    Publisher: Bioscientifica
    Publication Date: 2019
    detail.hit.zdb_id: 2010895-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 8
    In: BMC Cancer, Springer Science and Business Media LLC, Vol. 22, No. 1 ( 2022-10-29)
    Abstract: AZD0156 is an oral inhibitor of ATM, a serine threonine kinase that plays a key role in DNA damage response (DDR) associated with double-strand breaks. Topoisomerase-I inhibitor irinotecan is used clinically to treat colorectal cancer (CRC), often in combination with 5-fluorouracil (5FU). AZD0156 in combination with irinotecan and 5FU was evaluated in preclinical models of CRC to determine whether low doses of AZD0156 enhance the cytotoxicity of irinotecan in chemotherapy regimens used in the clinic. Methods Anti-proliferative effects of single-agent AZD0156, the active metabolite of irinotecan (SN38), and combination therapy were evaluated in 12 CRC cell lines. Additional assessment with clonogenic assay, cell cycle analysis, and immunoblotting were performed in 4 selected cell lines. Four colorectal cancer patient derived xenograft (PDX) models were treated with AZD0156, irinotecan, or 5FU alone and in combination for assessment of tumor growth inhibition (TGI). Immunofluorescence was performed on tumor tissues. The DDR mutation profile was compared across in vitro and in vivo models. Results Enhanced effects on cellular proliferation and regrowth were observed with the combination of AZD0156 and SN38 in select models. In cell cycle analysis of these models, increased G2/M arrest was observed with combination treatment over either single agent. Immunoblotting results suggest an increase in DDR associated with irinotecan therapy, with a reduced effect noted when combined with AZD0156, which is more pronounced in some models. Increased TGI was observed with the combination of AZD0156 and irinotecan as compared to single-agent therapy in some PDX models. The DDR mutation profile was variable across models. Conclusions AZD0156 and irinotecan provide a rational and active combination in preclinical colorectal cancer models. Variability across in vivo and in vitro results may be related to the variable DDR mutation profiles of the models evaluated. Further understanding of the implications of individual DDR mutation profiles may help better identify patients more likely to benefit from treatment with the combination of AZD0156 and irinotecan in the clinical setting.
    Type of Medium: Online Resource
    ISSN: 1471-2407
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2022
    detail.hit.zdb_id: 2041352-X
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 9
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 80, No. 4_Supplement ( 2020-02-15), p. P1-19-25-P1-19-25
    Abstract: Background: Triple-negative breast cancer (TNBC) is an aggressive subtype defined by lack of hormone receptor expression and non-amplified HER2. TNBC accounts for approximately 15% of breast cancer cases, however, is associated with an increased risk of cancer recurrence, brain metastasis, and death due to metastatic breast cancer. Mutations in p53 are common in TNBC, occurring in approximately 85% of tumors. While a number of promising targeted therapies are on the horizon in TNBC including immunotherapy, there remains an unmet need for active targeted therapies where chemotherapy remains the standard treatment for metastatic disease and results in a median survival of 12-18 months. Adavosertib (AZD1775) is a potent, small molecule, ATP-competitive inhibitor of the Wee1 kinase that potentiates the activity of many DNA-damaging chemotherapeutics and is currently in clinical development for multiple indications. AZD1775 potentiates the activity of DNA-damaging and antimetabolite chemotherapeutics in preclinical models without TP53-deficiency, possibly due to baseline replicative stress or compromised DNA repair proficiency. A previous unbiased screen of CTEP compounds in TNBC PDX models demonstrated that the combination of adavosertib and capecitabine/5FU had greater anti-proliferative effects than either of the single agents. The purpose of this study was to further investigate the combination of adavosertib and capecitabine/5FU in preclinical TNBC models. Methods: HCC1937, CAL51, MDA-MB-231 and MDA-MB-468 cells were plated in 96-well plates and exposed to increasing concentrations of adavosertib, 5FU, or the combination. Cellular proliferation was assessed in real-time using IncuCyte® Live Cell Analysis. Apoptosis was assessed via the Caspase-Glo 3/7 assay system. Western blotting was used to assess changes in expression of CDC2, phospho-CDC2, H2AX, and Bcl-xL. TNBC PDX models CU_TNBC_012 and CU_TNBC_013 were treated with vehicle, adavosertib, capecitabine, or the combination and assessed for tumor growth inhibition. Results: From the initial PDX screen, two of the four TNBC PDX models demonstrated a better response in the combination treatment than either of the single agents. As confirmation, two PDX models were expanded for statistical comparison. Both PDX models demonstrated a significant growth inhibition in the combination versus either of the single agents. (TNBC012, p & lt;0.05 combo vs adavosertib or capecitabine, TNBC013, p & lt;0.01 combo vs adavosertib or capecitabine ). An enhanced antiproliferative effect was observed in the adavosertib/5FU combination treatment as measured by live cell analysis. An increase in apoptosis was observed in two of the four cell lines in the combination when compared to single agent treatment. Treatment with single agent adavosertib resulted in an increase in p-cdc2 in a dose dependent manner that was also observed in the combination treatment. Similar results were observed with γH2AX in two of the four cell lines tested. No significant changes were observed in Bcl-xL following treatment in any of the cell lines. Conclusions: The combination of adavosertib and capecitabine/5-FU demonstrated enhanced combination effects both in vitro and in vivo in preclinical models of TNBC. These results support the clinical investigation of this combination in patients with TNBC, including those with brain metastasis given the CNS penetration of both agents. Citation Format: Todd M Pitts, Dennis M Simmons, Kyrie Dailey, Stacey M Bagby, Sarah J Hartman, Betelehem W Yacob, Brian Gittleman, John J Tentler, Diana Cittely, D. Ryan Ormond, Wells A Messersmith, S Gail Eckhardt, Jennifer R Diamond. Wee1 inhibition enhances the anti-tumor effects of capecitabine in preclinical models of triple negative breast cancer [abstract]. In: Proceedings of the 2019 San Antonio Breast Cancer Symposium; 2019 Dec 10-14; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2020;80(4 Suppl):Abstract nr P1-19-25.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2020
    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
    In: Frontiers in Oncology, Frontiers Media SA, Vol. 11 ( 2021-3-25)
    Abstract: Pancreatic ductal adenocarcinoma (PDAC) is a highly lethal cancer with high incidences of p53 mutations. AZD1775 (adavosertib, previously MK-1775) is a small molecule WEE1 inhibitor that abrogates the G2M checkpoint and can potentially synergize with DNA damaging therapies commonly used in PDAC treatment. The purpose of this study was to identify combination partners for AZD1775, including standard chemotherapy or targeted agents, in PDAC preclinical models. Low powered preliminary screens demonstrated that two of the four PDX models responded better to the combinations of AZD1775 with irinotecan or capecitabine than to either single agent. Following the screens, two full powered PDAC PDX models of differing p53 status were tested with the combinations of AZD1775 and irinotecan or capecitabine. The combinations of AZD1775 and SN38 or 5-FU were also tested on PDAC cell lines. Cellular proliferation was measured using an IncuCyte Live Cell Imager and apoptosis was measured using a Caspase-Glo 3/7 assay. Flow cytometry was conducted to measure alterations in cell cycle distribution. Western blot analysis was used to determine the effects of the drug combinations on downstream effectors. In PDX models with mutated p53 status, there was significant tumor growth inhibition from the combination of AZD1775 with irinotecan or capecitabine ( P ≤ 0.03), while PDX models with wild type p53 did not show anti-tumor synergy from the same combinations ( P ≥ 0.08). The combination of AZD1775 with SN38 or 5-FU significantly decreased proliferation in all PDAC cell lines, and enhanced apoptosis in multiple cell lines. Cell cycle distribution was disrupted from the combination of AZD1775 with SN38 or 5-FU which was recorded as G2M arrest and decreased G1 phase. AZD1775 inhibited phospho-CDC2 and increased the expression of γ H2AX that was either maintained or enhanced after combination with SN38 or 5-FU. The combination of AZD1775 with irinotecan/SN38 or capecitabine/5-FU showed anti-tumor effects in vivo and in vitro in PDAC models. These results support further investigation for these combination strategies to enhance outcomes for PDAC patients.
    Type of Medium: Online Resource
    ISSN: 2234-943X
    Language: Unknown
    Publisher: Frontiers Media SA
    Publication Date: 2021
    detail.hit.zdb_id: 2649216-7
    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...