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)
Material
Publisher
  • American Association for Cancer Research (AACR)  (12)
Language
Years
Subjects(RVK)
  • 1
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2019
    In:  Cancer Epidemiology, Biomarkers & Prevention Vol. 28, No. 6 ( 2019-06-01), p. 1059-1066
    In: Cancer Epidemiology, Biomarkers & Prevention, American Association for Cancer Research (AACR), Vol. 28, No. 6 ( 2019-06-01), p. 1059-1066
    Abstract: Management of advanced-stage non–small cell lung cancer (NSCLC) has changed significantly over the past two decades with the development of numerous systemic treatments, including targeted therapies. However, a high proportion of advanced-stage patients are untreated. The role that health insurance plays in receipt of systemic treatments is unclear. Methods: Using California Cancer Registry data (2012–2014), we developed multivariable Poisson regression models to assess the independent effect of health insurance type on systemic treatment utilization among patients with stage IV NSCLC. Systemic treatment information was manually abstracted from treatment text fields. Results: A total of 17,310 patients were evaluated. Patients with Medicaid/other public insurance were significantly less likely to receive any systemic treatments [risk ratio (RR), 0.78; 95% confidence interval (CI), 0.75–0.82], bevacizumab combinations (RR, 0.57; 95% CI, 0.45–0.71), or tyrosine kinase inhibitors (RR, 0.70; 95% CI, 0.60–0.82) compared with the privately insured. Patients with Medicare or dual Medicare–Medicaid insurance were not significantly different from the privately insured in their likelihood of receiving systemic treatments. Conclusions: Substantial disparities in the use of systemic treatments for stage IV NSCLC exist by source of health insurance in California. Patients with Medicaid/other public insurance were significantly less likely to receive systemic treatments compared with their privately insured counterparts. Impact: Source of health insurance influences care received. Further research is warranted to better understand barriers to treatment that patients with Medicaid face.
    Type of Medium: Online Resource
    ISSN: 1055-9965 , 1538-7755
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2019
    detail.hit.zdb_id: 2036781-8
    detail.hit.zdb_id: 1153420-5
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2014
    In:  Cancer Research Vol. 74, No. 19_Supplement ( 2014-10-01), p. 3739-3739
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 74, No. 19_Supplement ( 2014-10-01), p. 3739-3739
    Abstract: Pancreatic ductal adenocarcinoma (PDAC) has a poor prognosis, with poor responses to cytotoxic or targeted agents. PDAC is characterised by abundant desmoplasia and this tumor microenvironment is a major determinant of intrinsic resistance to anticancer therapies. In particular, pancreatic stellate cells/cancer-associated fibroblasts (PSCs/CAFs) are periacinar stromal cells which promote epithelial-mesenchymal transition (EMT) and radioresistance of PDAC. However, this unique tumour microenvironment is not recapitulated in conventional cell monoculture or xenografts, which might attribute to the poor predictivity of these preclinical drug screening models. To address this, we have developed a PDA tumor cell-specific in vitro Fluorescence-Based Co-culture (PDA-FBC) platform to investigate potential mechanisms of stroma-mediated chemoresistance and to evaluate novel agents and combinations. Using K8484 (PDA cell line derived from the LSL-KrasG12D/+; LSL-Trp53R172H/+; Pdx-Cre (KPC) mouse) stably expressing GFP, we selectively measured tumor cell viability in response to gemcitabine, with and without the presence of stromal cells. Monolayers of CAFs (primary fibroblast-like cells isolated from PDA tumour in KPC mice) or NFs (primary fibroblast-like cells from the normal pancreas of paired PC mice) were grown in 96 well plates and K8484 cells were seeded on top, prior to 72 hour drug exposure. When co-cultured with CAFs, K8484-GFP cells were significantly less sensitive to gemcitabine than cells grown in absence of CAFs: gemcitabine GI50 (50% growth inhibition concentration) is 19+/-3.2nM (n=4) in K8484 cell monolayer, 690+/-250nM (n=4) in the presence of CAFs (p = 0.03). Transwell assays revealed that this CAF-mediated chemoresistance was only induced in a cell-cell direct contact-dependent manner. Moreover, similar stroma-mediated gemcitabine resistance was not detected in co-culture of K8484 with NFs from normal pancreas (GI50=21+/-3.3nM). We are now investigating whether the chemoresistance phenotype extends to other cytotoxic agents. To evaluate potential mechanisms of resistance, RNA sequencing is being conducted to compare molecular profiles of K8484-GFP cells co-cultured with CAFs or NFs (with/without gemcitabine), to identify the genes modulated by the tumor-stroma interaction and potentially responsible for the decreased gemcitabine sensitivity in co-culture. In conclusion, we have identified that cancer-associated fibroblasts induce resistance to gemcitabine in KPC PDAC tumour cells, in contrast to fibroblasts from normal pancreas. This platform will have utility in the assessment of novel drug combinations and a mechanistic understanding of the tumor-stroma interactions may identify new candidate drug combinations. Citation Format: Ruiling Xu, Frances M. Richards, Yao Lin, Duncan I. Jodrell. An in vitro fluorescence-based co-culture model identifies tumour microenvironment-mediated chemoresistance in pancreatic cancer. [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 3739. doi:10.1158/1538-7445.AM2014-3739
    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 ...
  • 3
    In: Molecular Cancer Therapeutics, American Association for Cancer Research (AACR), Vol. 22, No. 12_Supplement ( 2023-12-01), p. B132-B132
    Abstract: Background: We used the TH-MYCN mouse model of neuroblastoma (NB) to determine the importance of simultaneously inhibiting both polyamine uptake and synthesis when added to standard-of-care chemotherapy/immunotherapy. Polyamines are essential cations frequently upregulated in tumors. We have shown the benefit of adding DFMO, an irreversible polyamine synthesis inhibitor, to standard-of-care chemotherapy in several NB mouse models (Evageliou, Clin Cancer Res 2016). Based on our work, a randomized Phase 2 COG trial of DFMO, added to chemotherapy/anti-GD2 immunotherapy, is ongoing for relapsed NB (NCT03794349). We recently showed in NB mouse models that DFMO efficacy is enhanced by adding the polyamine uptake inhibitor AMXT1501 (Gamble, Sci Transl Med, 2019). DFMO/AMXT1501 is now in adult clinical trial (NCT05500508). Preclinical modelling of polyamine-targeted therapeutics in TH-MYCN mice has been invaluable for supporting clinical trial design but has yet to include anti-GD2 immunotherapy. Methods: TH-MYCN mice were used to optimize DFMO/AMXT1501 therapy combined with temozolomide/irinotecan (TEM/IRI) or cyclophosphamide/topotecan (CYCLO/TOPO), and anti-GD2 (14G2a) antibody. We studied escalating doses of DFMO and AMXT1501 (n=10/group), with or without TEM/IRI or CYCLO/TOPO. 14G2a regimen was titrated toward human-relevant pharmacokinetic exposures to establish suitable conditions for combination therapies. Results: In the absence of chemo-immunotherapy, highest-dose DFMO (1.5%) and AMXT1501 (2.5 mg/kg/d) had greatest efficacy without increased toxicity. Median mouse survival was 2-fold greater compared with lower dose DFMO/AMXT1501 combinations, suggesting that maximising DFMO dose is critical. Similar improved efficacy was observed in combination with TEM/IRI or CYCLO/TOPO. Combining 14G2a (100ug 2x/wk, 18 wks; McNerney, Oncoimmunology, 2022) with either CYCLO/TOPO (single 5-day chemo cycle) or DFMO/AMXT1501 (1.5%/2.5 mg/kg/d) increased survival compared with either treatment alone. Combining all 5 drugs resulted in 100% survival at 1 year, with minimal toxicity. However substantial dose reductions of 14G2a ( & lt;25µg, 2 doses) and TEM/IRI or CYCLO/TOPO (single 2-day chemo cycle) were needed to identify clinically relevant backbones which achieved human-relevant 14G2a pharmacokinetics. Addition of DFMO alone to the clinically relevant CYCLO/TOPO/14G2a backbone did not result in benefit compared to backbone alone. In contrast, addition of AMXT1501 plus DFMO to the backbone significantly extended survival (P & lt; 0.001). Conclusions: We have increased the utility of the TH-MYCN mouse model for preclinical modelling of polyamine inhibition, and other investigational treatments, by pharmacokinetics-driven optimization of anti-GD2 therapy with standard-of-care chemotherapy backbones. This approach will improve optimisation for future NB clinical trials. Our data highlight the importance of adding AMXT1501 to DFMO for polyamine inhibition and support a planned international DFMO/AMXT1501/chemotherapy/anti-GD2 trial for refractory NB.   Citation Format: Jayne Murray, Ruby Pandher, Lin Xiao, Klaartje Somers, Jennifer Brand, Erin Mosmann, Stephanie Alfred, Frances Kusuma, Adam Kearns, Julie R Park, Lynley V Marshall, Chiara Gorrini, Louis Chesler, Michael D Hogarty, Andrew D J Pearson, Mark Burns, Jamie I Fletcher, David Ziegler, Murray D Norris, Michelle Haber. Addition of AMXT1501 (polyamine uptake inhibitor) plus DFMO (polyamine synthesis inhibitor) to standard-of-care chemotherapy/anti-GD2 antibody in the TH-MYCN mouse neuroblastoma model, enhances efficacy compared to addition of DFMO alone [abstract]. In: Proceedings of the AACR-NCI-EORTC Virtual International Conference on Molecular Targets and Cancer Therapeutics; 2023 Oct 11-15; Boston, MA. Philadelphia (PA): AACR; Mol Cancer Ther 2023;22(12 Suppl):Abstract nr B132.
    Type of Medium: Online Resource
    ISSN: 1538-8514
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2023
    detail.hit.zdb_id: 2062135-8
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    In: Clinical Cancer Research, American Association for Cancer Research (AACR), Vol. 28, No. 7 ( 2022-04-01), p. 1258-1267
    Abstract: HER2 mutations (HER2mut) induce endocrine resistance in estrogen receptor–positive (ER+) breast cancer. Patients and Methods: In this single-arm multi-cohort phase II trial, we evaluated the efficacy of neratinib plus fulvestrant in patients with ER+/HER2mut, HER2 non-amplified metastatic breast cancer (MBC) in the fulvestrant-treated (n = 24) or fulvestrant-naïve cohort (n = 11). Patients with ER-negative (ER−)/HER2mut MBC received neratinib monotherapy in an exploratory ER− cohort (n = 5). Results: The clinical benefit rate [CBR (95% confidence interval)] was 38% (18%–62%), 30% (7%–65%), and 25% (1%–81%) in the fulvestrant-treated, fulvestrant-naïve, and ER− cohorts, respectively. Adding trastuzumab at progression in 5 patients resulted in three partial responses and one stable disease ≥24 weeks. CBR appeared positively associated with lobular histology and negatively associated with HER2 L755 alterations. Acquired HER2mut were detected in 5 of 23 patients at progression. Conclusions: Neratinib and fulvestrant are active for ER+/HER2mut MBC. Our data support further evaluation of dual HER2 blockade for the treatment of HER2mut MBC.
    Type of Medium: Online Resource
    ISSN: 1078-0432 , 1557-3265
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2022
    detail.hit.zdb_id: 1225457-5
    detail.hit.zdb_id: 2036787-9
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 78, No. 13_Supplement ( 2018-07-01), p. 874-874
    Abstract: Although effective in BRAF V600 mutant melanoma, Type 1.5 RAF inhibitors such as vemurafinib and dabrafenib have not proven to be successful in KRAS mutant cancers, neither as single agent nor in combination with MEK inhibitors. Through large-scale cellular compound combination screening we found that Type II RAF inhibitors such as AZ-628 do show synergistic activity with MEK inhibitors in multiple KRAS mutant indications, including NSCLC, colorectal cancer and ovarian cancer. The combination of Type II RAF inhibitors and MEK inhibitors demonstrates robust and durable abrogation of MAPK signaling both on canonical markers of MAPK activity such as pERK and pRSC as well as transcriptional output. We also observe synergistic in vivo tumor growth inhibition in two independent models of KRAS mutant cancer by this combination treatment. We found that treatment with MEK inhibitors alone drives the increase of active RAS-GTP levels and induces CRAF:BRAF hetero-dimerization. These induced dimers are active and able to phosphorylate MEK in vitro. This increased dimerization renders cells sensitive to Type 2 RAF inhibitors. We find that this effect is not limited to KRAS mutant cells, as a subset of KRAS wild-type cells show increased RAS-GTP levels upon MEK inhibitor treatment. These cells also show synergistic sensitivity to Type 2 RAF inhibition. Additionally, we observed significantly higher synergy and higher RAS-GTP levels in KRAS G13D mutant cells, which have intrinsically high GDP exchange and low intrinsic GTP hydrolysis. Finally, we show that GDC-0941 and GDC-0032, two broad PI3K inhibitors, also induce RAS-GTP levels in cells independent of PIK3CA or KRAS mutation status. We subsequently observed a synergistic sensitivity to Type 2 RAF inhibitors in these PI3K inhibitor-treated cells. Overall, we demonstrate that pharmacologic inhibition of MEK or PI3K increases RAS-GTP levels and drives increased CRAF:BRAF hetero-dimerization. This in turn sensitizes cells to Type 2 RAF inhibition, leading to a synergistic drug effect. Combination of these inhibitors may be a viable therapeutic approach in KRAS mutant cancer, and may be especially effective in KRAS G13D-carrying tumors. Citation Format: Christiaan N. Klijn, Ivana Yen, Frances Shanahan, Mark Merchant, Christine Orr, Thomas Hunsaker, Matthew Durk, Hank La, Xiaoling Zhang, Scott Martin, Eva Lin, John Chan, Yihong Yu, Dhara Amin, Amy Gustafson, Scott Foster, Joachim Rudolph, Shiva Malek. Pharmacologically induced RAS-GTP levels and CRAF-BRAF hetero-dimerization drive sensitization to Type II pan-RAF inhibitors in KRAS mutant cancer [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 874.
    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 ...
  • 6
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2012
    In:  Cancer Research Vol. 72, No. 8_Supplement ( 2012-04-15), p. 1924-1924
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 72, No. 8_Supplement ( 2012-04-15), p. 1924-1924
    Abstract: CYC3, an Aurora Kinase A specific inhibitor, suppresses the pancreatic cancer cell growth (72h GI50 2.4μM in MiaPaCa-2 cells and 1.95μM in Panc-1 cells), arrest the cells at M phase and induces apoptosis. In order to evaluate CYC3's clinical potential, we employed mathematical models and simulations to look for possible synergistic combinations of CYC3 with paclitaxel in MiaPaCa-2 and Panc-1 cells. We have identified low concentrations of paclitaxel (3nM) and CYC3 is synergistic in inhibiting pancreatic cell growth, achieving similar effects as high concentrations of paclitaxel (30nM). Liquid Chromatography-Mass Spectrometry (LC-MS) analysis shows that CYC3 does not alter the cellular uptake of paclitaxel, supporting a synergistic mechanism at molecular level. In addition, the combination of CYC3 with low concentration of paclitaxel (3nM) displays less myelotoxicity compared to high concentrations of paclitaxel (30nM) in colony formation assay using Colony Forming Unit of Granulocyte and Macrophage (CFU-GM) (60-70% vs 100% inhibition), suggesting a potentially safer but equally efficient application of paclitaxel in patients. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 103rd Annual Meeting of the American Association for Cancer Research; 2012 Mar 31-Apr 4; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2012;72(8 Suppl):Abstract nr 1924. doi:1538-7445.AM2012-1924
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2012
    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: Clinical Cancer Research, American Association for Cancer Research (AACR), Vol. 29, No. 17 ( 2023-09-01), p. 3340-3351
    Abstract: Plasma circulating tumor DNA (ctDNA) analysis is used for genotyping advanced non–small cell lung cancer (NSCLC); monitoring dynamic ctDNA changes may be used to predict outcomes. Patients and Methods: This was a retrospective, exploratory analysis of two phase III trials [AURA3 (NCT02151981), FLAURA (NCT02296125)]. All patients had EGFR mutation-positive (EGFRm; ex19del or L858R) advanced NSCLC; AURA3 also included T790M-positive NSCLC. Osimertinib (FLAURA, AURA3), or comparator EGFR–tyrosine kinase inhibitor (EGFR-TKI; gefitinib/erlotinib; FLAURA), or platinum-based doublet chemotherapy (AURA3) was given. Plasma EGFRm was analyzed at baseline and Weeks 3/6 by droplet digital PCR. Outcomes were assessed by detectable/non-detectable baseline plasma EGFRm and plasma EGFRm clearance (non-detection) at Weeks 3/6. Results: In AURA3 (n = 291), non-detectable versus detectable baseline plasma EGFRm had longer median progression-free survival [mPFS; HR, 0.48; 95% confidence interval (CI), 0.33–0.68; P & lt; 0.0001]. In patients with Week 3 clearance versus non-clearance (n = 184), respectively, mPFS (months; 95% CI) was 10.9 (8.3–12.6) versus 5.7 (4.1–9.7) with osimertinib and 6.2 (4.0–9.7) versus 4.2 (4.0–5.1) with platinum-pemetrexed. In FLAURA (n = 499), mPFS was longer with non-detectable versus detectable baseline plasma EGFRm (HR, 0.54; 95% CI, 0.41–0.70; P & lt; 0.0001). For Week 3 clearance versus non-clearance (n = 334), respectively, mPFS was 19.8 (15.1 to not calculable) versus 11.3 (9.5–16.5) with osimertinib and 10.8 (9.7–11.1) versus 7.0 (5.6–8.3) with comparator EGFR-TKI. Similar outcomes were observed by Week 6 clearance/non-clearance. Conclusions: Plasma EGFRm analysis as early as 3 weeks on-treatment has the potential to predict outcomes in EGFRm advanced NSCLC.
    Type of Medium: Online Resource
    ISSN: 1078-0432 , 1557-3265
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2023
    detail.hit.zdb_id: 1225457-5
    detail.hit.zdb_id: 2036787-9
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 8
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 77, No. 13_Supplement ( 2017-07-01), p. 4967-4967
    Abstract: While activating mutations in the KRAS oncogene frequently drive tumorigenesis in human cancers (40% CRC, 20% NSCLC) through constitutive activation of the MAPK pathway, there are currently no targeted treatments available for KRAS mutant cancers. Inhibitors of the individual nodes of the MAPK pathway have been developed, but these molecules have been largely ineffective against KRAS mutant tumors in the clinic. Multiple studies have shown rational combinations of MAPK inhibitors may have anti-tumor activity in KRAS mutant models. In order to understand the versatility of combining RAF inhibitors in this context, we conducted a library screen consisting of 430 small molecule tool compounds in combination with RAF inhibitor AZ-628. Here we show: RAF inhibitors combine especially well with other MAPK pathway inhibitors in KRAS mutant tumor cells. In particular, Type II RAF inhibitors are synergistic with the MEK inhibitor Cobimetinib in vitro and exhibit tumor regressing efficacy in xenograft mouse model studies in vivo. Mechanistically, we have found the MEK inhibitor disables ERK induced negative feedback on the MAPK pathway resulting in activation of CRAF in a KRAS dependent manner. The combination of RAF with MEK inhibition blunts KRAS-dependent activation of CRAF kinase activity and robustly inhibits MAPK signaling, thereby driving efficacy in KRAS mutant tumors. Broad cell line profiling with the combination of RAF and MEK inhibitors demonstrates that a majority of KRAS mutant lung and colorectal tumor lines exhibit synergy with the combination. Therefore, combining a Type II pan-RAF inhibitor with a MEK inhibitor has the potential to improve the therapeutic outcome in KRAS mutant cancers. Citation Format: Ivana Yen, Christiaan Klijn, Frances Shanahan, Mark Merchant, Christine Orr, Thomas Hunsaker, Matthew Durk, Hank La, Xiaolin Zhang, Scott Martin, Eva Lin, John Chan, Yihong Yu, Amy Gustafson, Joachim Rudolph, Shiva Malek. RAF kinase inhibition synergizes with MEK inhibitors in KRAS mutant tumors [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 4967. doi:10.1158/1538-7445.AM2017-4967
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2017
    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. 83, No. 11_Supplement ( 2023-06-02), p. B048-B048
    Abstract: Introduction Many biomarker assays improve sensitivity in diagnosing high grade prostate cancer (PCa), but their performance during active surveillance, after a diagnosis of PCa, is not well understood. Procedures In the multicenter Canary PASS cohort, we explored the contribution of four distinct biomarker panels (blood-based: 4Kpanel, phi; urine-based: PCA3, MDxSelect RNA) and clinical variables in predicting 4-year extreme biopsy reclassification to Gleason Grade Group 3 (GG3) or above. Biomarker assays were performed between 2014-2020 on subcohorts including 727-1,176 participants; outcomes were collected through March 2020. Models were built using a) biomarker, b) biomarker + prostate volume, c) biomarker + biopsy variables d) biomarker + prostate volume + biopsy variables, and, for urine markers e) biomarker + prostate volume + biopsy variables + serum PSA. Clinical PSA was used as a reference. Partly conditional Cox proportional hazards regression models for residual time to event were constructed based on information available at each prediction time, accounting for competing risk. The probability of extreme reclassification within 4 years was calculated after diagnosis and after first follow-up biopsy (Bx1) and the accuracy was assessed with the cross-validated receiver operating characteristic (ROC) curve analysis. Results Cross validated Areas Under the Curve (AUCs) for predictions made with biomarkers ranged from 0.648 (95% CI: 0.581, 0.716) to 0.755 (95% CI: 0.687, 0.824) after diagnosis and from 0.588 (95% CI: 0.480, 0.696) to 0.669 (95% CI: 0.558, 0.780) after Bx1, and were consistently higher than AUCs for clinical PSA at both timepoints; the difference in AUC between biomarker and PSA was statistically significant only for 4Kpanel and phi. Adding clinical variables to predictive models improved AUCs to varying degrees; AUCs for full models with all variables were 0.702 (95% CI: 0.641, 0.764) to 0.776 (95% CI: 0.715, 0.837) after diagnosis and 0.740 (95% CI: 0.656, 0.824) to 0.763 (95% CI: 0.691, 0.836) after Bx1. Adding clinical variables to blood biomarkers resulted in less incremental improvement than for urine markers. A limitation of this study is that prostate MRI was not part of the PASS protocol, although MRI data collected suggests minimal predictive ability during active surveillance. Conclusions Our results suggest that early during prostate cancer active surveillance the blood-based biomarkers of the 4Kpanel and phi may predict future reclassification to higher grade cancer as well as or better than common clinical variables. Clinical variables improve the performance of the urine-based markers of PCA3 or MDxSelect. External validation studies are needed. Citation Format: Lisa F. Newcomb, Yingye Zheng, Menghan Liu, James D. Brooks, Peter R. Carroll, Atreya Dash, William J. Ellis, Christopher J. Filson, Martin E. Gleave, Michael A. Liss, Frances M. Martin, Todd M. Morgan, Peter S. Nelson, Andrew A. Wagner, Daniel W. Lin. Performance of diagnostic biomarkers in the Canary Prostate cancer Active Surveillance Study (PASS) [abstract]. In: Proceedings of the AACR Special Conference: Advances in Prostate Cancer Research; 2023 Mar 15-18; Denver, Colorado. Philadelphia (PA): AACR; Cancer Res 2023;83(11 Suppl):Abstract nr B048.
    Type of Medium: Online Resource
    ISSN: 1538-7445
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2023
    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: Clinical Cancer Research, American Association for Cancer Research (AACR), Vol. 27, No. 15 ( 2021-08-01), p. 4338-4352
    Abstract: We investigated whether targeting chromatin stability through a combination of the curaxin CBL0137 with the histone deacetylase (HDAC) inhibitor, panobinostat, constitutes an effective multimodal treatment for high-risk neuroblastoma. Experimental Design: The effects of the drug combination on cancer growth were examined in vitro and in animal models of MYCN-amplified neuroblastoma. The molecular mechanisms of action were analyzed by multiple techniques including whole transcriptome profiling, immune deconvolution analysis, immunofluorescence, flow cytometry, pulsed-field gel electrophoresis, assays to assess cell growth and apoptosis, and a range of cell-based reporter systems to examine histone eviction, heterochromatin transcription, and chromatin compaction. Results: The combination of CBL0137 and panobinostat enhanced nucleosome destabilization, induced an IFN response, inhibited DNA damage repair, and synergistically suppressed cancer cell growth. Similar synergistic effects were observed when combining CBL0137 with other HDAC inhibitors. The CBL0137/panobinostat combination significantly delayed cancer progression in xenograft models of poor outcome high-risk neuroblastoma. Complete tumor regression was achieved in the transgenic Th-MYCN neuroblastoma model which was accompanied by induction of a type I IFN and immune response. Tumor transplantation experiments further confirmed that the presence of a competent adaptive immune system component allowed the exploitation of the full potential of the drug combination. Conclusions: The combination of CBL0137 and panobinostat is effective and well-tolerated in preclinical models of aggressive high-risk neuroblastoma, warranting further preclinical and clinical investigation in other pediatric cancers. On the basis of its potential to boost IFN and immune responses in cancer models, the drug combination holds promising potential for addition to immunotherapies.
    Type of Medium: Online Resource
    ISSN: 1078-0432 , 1557-3265
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2021
    detail.hit.zdb_id: 1225457-5
    detail.hit.zdb_id: 2036787-9
    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...