feed icon rss

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)  (184)
  • English  (184)
Type of Medium
Publisher
  • American Association for Cancer Research (AACR)  (184)
Language
  • English  (184)
Subjects(RVK)
  • 1
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2012
    In:  Cancer Research Vol. 72, No. 8_Supplement ( 2012-04-15), p. 5335-5335
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 72, No. 8_Supplement ( 2012-04-15), p. 5335-5335
    Abstract: Pancreatic cancer has the worst prognosis of any major malignancy with complex etiology involving both environmental and genetic factors. Despite increasing knowledge in tumor biology, the treatment efficacy in pancreatic cancers has not improved significantly over the past decade. Thus, the identification of novel molecular pathways involved in pancreatic oncogenesis and the development of new and potent therapeutic options are highly desirable. The realization that tumors themselves function as complex organs birthed the concept that cancer cells with tumor-initiating properties of stem cells may be the key drivers of the complex machinery behind oncogenesis. Tumor-initiating cells (TICs) have been studied extensively in the hematopoietic system, however it is not known which subpopulation has the highest tumor-initiating potential in pancreatic cancer. We have identified that the CD133+/CD44+/ALDH1+ cell population derived from 31 pancreatic adenocarcinomas and 6 pancreatic cancer cell lines, has the highest tumorigenic potential, assessed by in vivo dilution tumor propagation assays in highly immunodeficient (NOD-SCID IL2Rγ-/-) mice. In addition, this population has the highest sphere-forming capacity and is highly resistant to gemcitabine treatment in comparison to the other subpopulations. RNA-sequencing analysis in CD133−/CD44−/ALDH1− and CD133+/CD44+/ALDH1+ subpopulations derived from BxPC3 pancreatic cells revealed the transcriptomic alterations between these cell types. Furthermore, gene network analysis revealed enrichment of inflammatory and lipid metabolism networks in pancreatic TICs, suggesting the potential role of inflammation and lipid metabolism in pancreatic TIC function. In addition, a gene network-based FDA-approved chemical library screen revealed that metformin perturbs the activated networks in pancreatic TICs leading to the suppression of their growth. Metformin suppressed pancreatic cancer growth, through suppression of pancreatic TICs, in different pancreatic cancer mouse models. Overall, we propose the identification of a novel pancreatic cancer subpopulation which harbors tumor-initiating properties and is regulated by inflammatory signals, contributing to our understanding of how inflammation is linked to pancreatic oncogenesis in the molecular level. Furthermore, by following a novel strategy, we have identified that metformin suppresses pancreatic tumor growth and relapse through inhibition of pancreatic TIC growth. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 103rd Annual Meeting of the American Association for Cancer Resear ch; 2012 Mar 31-Apr 4; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2012;72(8 Suppl):Abstract nr 5335. doi:1538-7445.AM2012-5335
    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
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 2
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2013
    In:  Cancer Research Vol. 73, No. 9 ( 2013-05-01), p. 2760-2769
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 73, No. 9 ( 2013-05-01), p. 2760-2769
    Abstract: The classic view of metastatic cancer progression is that it is a unidirectional process initiated at the primary tumor site, progressing to variably distant metastatic sites in a fairly predictable, although not perfectly understood, fashion. A Markov chain Monte Carlo mathematical approach can determine a pathway diagram that classifies metastatic tumors as “spreaders” or “sponges” and orders the timescales of progression from site to site. In light of recent experimental evidence highlighting the potential significance of self-seeding of primary tumors, we use a Markov chain Monte Carlo (MCMC) approach, based on large autopsy data sets, to quantify the stochastic, systemic, and often multidirectional aspects of cancer progression. We quantify three types of multidirectional mechanisms of progression: (i) self-seeding of the primary tumor, (ii) reseeding of the primary tumor from a metastatic site (primary reseeding), and (iii) reseeding of metastatic tumors (metastasis reseeding). The model shows that the combined characteristics of the primary and the first metastatic site to which it spreads largely determine the future pathways and timescales of systemic disease. Cancer Res; 73(9); 2760–9. ©2013 AACR.
    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
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 3
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 75, No. 9_Supplement ( 2015-05-01), p. PD5-3-PD5-3
    Abstract: Background: Phosphatidylinositol 3-kinase (PI3K) hyperactivation plays a role in endocrine therapy resistance. Adding an α-selective PI3K inhibitor (BYL719) to hormonal therapy may therefore overcome resistance in HR+ MBC. We report results from a phase I study to evaluate the safety and preliminary efficacy of BYL719 plus an AI in patients (pts) with HR+ MBC. Methods: This 3+3 dose-escalation trial studied daily oral BYL719 added to standard dose letrozole (L, Arm A) or exemestane (E, Arm B), and later examined intermittent dosing (Arm C, L + BYL719 every other week; Arm D, E + BYL719 on 5 of 7 days weekly). Pts with HR+ MBC, any/no PIK3CA mutation, and on L/E were eligible. A cycle (C) was 28 days (d). Endpoints were dose-limiting toxicity (DLT), tolerability (CTCAE 4.0), and efficacy (RECIST v1.1). Paired tumor biopsies were performed for genomic and proteomic correlatives. Serial plasma was collected to quantify cell-free (cf) DNA and mutant allele fraction. Results: 14 pts (median (M) age: 55 (30-69) yrs), 7 each on Arms A and B, received a M of 76d (6-312+) of BYL719 + L or E. All were evaluable for toxicity, 10 for response. PIK3CA status was mutant(MT)/wild-type(WT)/unknown in 8/5/1 pts. M number of prior MBC therapies was 2 (1-12) in Arm A, 6 (2-14) in Arm B. Arms had similar toxicities. On Arm A, BYL719 was given at 300mg daily (DL0) to 3 pts who completed the 28d DLT period. 2 pts had 3 distinct DLTs: maculopapular rash (N=1), hyperglycemia (N=1), abdominal pain (N=1). Dose was de-escalated (DL-1=250mg) with no DLT in 3 enrolled pts. On Arm B, DL0, 1 pt experienced DLT (maculopapular rash) of 3 initially enrolled pts. Arm B expansion at DL0 had 1 additional pt with DLT (rash). Clinically significant, treatment-related toxicities included grade (G)≥4: none; G3: maculopapular rash (N=8, including 1 pt treated at DL-1), hyperglycemia (N=1) and G1/2: fatigue (N=7), nausea (N=7), and hyperglycemia (N=6). Toxicity required 6 dose reductions in 4pts and discontinuation in 2 pts. M duration on study for PIK3CA MT vs. WT was 169.5d vs. 69.5d, respectively. In pts with PIK3CA–MT MBC evaluable for response (n=7), 6 had clinical benefit: 1 PR (pt heavily pre-treated, including prior L, MBC to liver, Arm A, now C10+ after DLT); SD (n=5, included -29.9%, -19%, -12%), and POD (n=1). In pts with PIK3CA-WT MBC evaluable for response (n=3), 2 had SD (no changes ≥+/-5%) and 1 had POD. Serial cfDNA analysis in 4 pts with SD or PR demonstrated a decrease of & gt;90% in the PIK3CA mutant allele fraction on treatment. Due to toxicity seen with continuous BYL719, the study was amended to explore intermittent dosing schedules (Arm C, L; Arm D, E; DL0=250mg), with 5 pts enrolled, 3 of whom have completed the DLT period with no DLTs, and 1 pt with G3 rash. Correlative studies including serial cf DNA collection from these pts is ongoing. Conclusions: Continuously dosed BYL719 with L or E shows promising antitumor activity. Skin toxicity warranted evaluation of alternative schedules. Mutant allele fraction may be an early predictor of response and may serve as a pharmacodynamic marker during intermittent treatment. Safety, efficacy, and correlative data from study Arm C and Arm D will also be presented. Citation Format: Payal D Shah, Mary Ellen Moynahan, Shanu Modi, Betty Ann Caravella, Farrah M Datko, Stephen Zamora, Elizabeth Comen, Theresa Gilewski, Steven M Sugarman, Gabriella D'Andrea, Diana E Lake, Shari B Goldfarb, Sujata Patil, Anne Covey, Michael F Berger, Mario E Lacouture, Larry Norton, Clifford A Hudis, Jose Baselga, Sarat Chandarlapaty, Maura N Dickler. Phase I trial: PI3Kα inhibitor BYL719 plus aromatase inhibitor (AI) for patients with hormone receptor-positive (HR+) metastatic breast cancer (MBC) [abstract]. In: Proceedings of the Thirty-Seventh Annual CTRC-AACR San Antonio Breast Cancer Symposium: 2014 Dec 9-13; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2015;75(9 Suppl):Abstract nr PD5-3.
    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
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 4
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2015
    In:  Cancer Research Vol. 75, No. 15_Supplement ( 2015-08-01), p. 2639-2639
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 75, No. 15_Supplement ( 2015-08-01), p. 2639-2639
    Abstract: BNC101 is a monoclonal antibody (mAb) targeting LGR5 that has successfully completed IND-enabling studies in preparation for Phase I clinical studies in 2015. LGR5 is a validated cancer stem cell (CSC) receptor overexpressed in colorectal cancer (CRC), pancreatic cancer and most other solid tumors. Loss and gain-of-function studies indicate that LGR5 is a functional cancer receptor involved in tumor growth and survival. Sorted LGR5+ primary CRC are highly tumorigenic compared to LGR5- cells in limiting dilution in vivo xenograft studies. BNC101 was selected as the lead LGR5 candidate for clinical development using Bionomics’ CSC Rx Discovery™ platform. Key lead selection criteria for BNC101 were anti-tumor and anti-CSC activity in vitro and in vivo, in patient-derived xenograft (PDX) tumor models from multiple tumor types. CSC activity was evaluated by functional in vivo limiting dilution assays (LDA) using tumor cells treated with BNC101 +/- standard-of-care chemotherapy (SOC) combinations. In LDA studies with the K-Ras mutant CRC PDX tumors CT1 and CT3, 5/8 (63%) and 6/8 (75%) mice implanted with serially diluted cells from BNC101 treated tumors remained tumor free, compared to 1/8 (13%) and 2/8 (25%) in the control groups. Combination with SOC further improved BNC101 activity against CT3 tumors, where 8/8 mice re-implanted for LDA had 6 months progression-free-survival (vs 3/8 in chemo group). In the pancreatic PDX model JH109, BNC101 combined with SOC flat-lined established tumors in 3/7 mice, compared to 0/7 tumors eradicated with control SOC alone. BNC101 is also active in multiple pancreatic, triple-negative breast (TNBC) and small-cell lung (SCLC) cancer xenograft models. Ongoing translational studies have identified a panel of Wnt genes modulated by BNC101 treatment that could potentially be used to identify BNC101 responsive versus resistant patients in the clinic. Furthermore, BNC101 treatment significantly reduced LGR5+ HLA+ circulating-tumor-cells (CTCs) in the peripheral blood of tumor-bearing mice, providing a pharmacodynamic (PD) biomarker of target engagement to further evaluate in clinical studies. Altogether, the preclinical data package supports our therapeutic hypothesis that BNC101 targeting of LGR5+ CSCs will significantly improve PFS and OS in solid tumor indications. Additional translational biomarker data to support the clinical development of BNC101 will also be presented. Citation Format: Peter Chu, Kristen Smith, Farbod Shojaei, Colin Walsh, John Norton, Jose Iglesias, Christopher Reyes. Preclinical evaluation and biomarker identification for the anti-LGR5 mAb BNC101 in K-Ras mutant CRC and other solid tumor indications. [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 2639. doi:10.1158/1538-7445.AM2015-2639
    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
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 5
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 72, No. 24_Supplement ( 2012-12-15), p. P2-10-01-P2-10-01
    Abstract: Background: Intrinsic breast cancer subtypes determined by the PAM50 assay are reported to be prognostic independent of standard clinicopathological variables. The CALGB (Alliance) 9741 adjuvant trial randomized treatment in a 2×2 factorial design to (i) 2-wk (dose dense; DD) vs 3-wk therapy and (ii) sequential vs concurrent doxorubicin, cyclophosphamide, paclitaxel (A & gt;T & gt;C vs AC & gt;T). DD therapy improved disease-free survival and overall survival (OS) (Citron et al. JCO 2003.). A significant interaction between intrinsic breast cancer subtypes and the use of DD therapy was hypothesized. Methods: C9741 was conducted in collaboration with ECOG, NCCTG, and SWOG. Biospecimens were collected from 1652 of 2005 subjects. Multiplexed gene expression profiling (NanoString Technologies, Inc) generated PAM50 subtype calls (luminal A, luminal B, HER2-enriched, basal-like), risk of relapse (ROR) score, and proliferation score for 1321 cases (80%). Excluded samples were due to insufficient tumor (n = 181) or RNA (n = 150). The primary endpoint was relapse-free survival (RFS). The primary analysis to determine if the clinical benefit of DD therapy is dependent on intrinsic subtype was conducted as a test of interaction in a Cox proportional hazards model (3 degrees of freedom, df; alpha=0.05). Similar analyses were performed for ROR score and proliferation score as continuous measures of risk (1 df). PAM50 subtype, ROR score, and proliferation score were evaluated in separate multivariate models adjusting for number of positive nodes, menopausal status, dose density, and sequence of therapy. Results: Improved outcomes for DD therapy in the evaluable subset mirrored results from the complete dataset (HR = 1.20; 95%CI 0.99–1.44) with a median follow-up of 12.3 yrs. Subtype analysis identified 32% luminal A (n = 417), 26% luminal B (n = 341), 20% HER2-enriched (n = 267), and 22% basal-like (n = 296). Intrinsic subtypes were prognostic of RFS (p & lt; 0.0001) irrespective of treatment assignment with HRs relative to the luminal A subgroup of 1.50 (luminal B), 1.70 (HER2-enriched) and 1.66 (basal-like). No subtype specific treatment effect on RFS was identified (p = 0.44). ROR scores (range 0–100, median 60) were also prognostic of RFS (HR = 1.12 for a 10-unit change; 95% CI 1.07–1.18; p & lt; 0.0001), but no association with DD therapy benefit was seen (p = 0.58). Similar results were obtained for the proliferation score, for OS as a secondary endpoint, and from multivariate models with clinical covariates. Conclusion: The prognostic value of PAM50 subtype, ROR score, and proliferation score remains highly significant in patients treated with contemporary adjuvant anthracycline and taxane based chemotherapy. Intrinsic subtype did not predict for improved survival with the 2-wk DD vs 3-wk treatment schedule; we hypothesize that this is because the prognostic differences by subtype outweighed the modest but significant clinical benefit of DD chemotherapy administration for the overall population. Planned clinical validation studies will assess the ability of PAM50 and other gene signatures to stratify patients and individualize systemic therapy regimens based on the expected risk of distant recurrence. Citation Information: Cancer Res 2012;72(24 Suppl):Abstract nr P2-10-01.
    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
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 6
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2010
    In:  Cancer Research Vol. 70, No. 24_Supplement ( 2010-12-15), p. P5-08-01-P5-08-01
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 70, No. 24_Supplement ( 2010-12-15), p. P5-08-01-P5-08-01
    Abstract: Background: Many breast cancer survivors of child-bearing age wish to become parents after therapy and are concerned about the possibility of treatment-induced infertility. Educating patients about the effects of therapy on fertility, early menopause, and fertility preservation options prior to treatment may optimize a survivor's quality of life after treatment. It is unclear whether oncologists feel qualified to discuss fertility issues with their patients, and if not, what barriers prevent such discussions. Methods: An IRB approved cross-sectional survey was developed at Memorial Sloan-Kettering Cancer Center (MSKCC) in order for clinicians to self-evaluate their knowledge, attitudes, and behaviors regarding fertility preservation. Survey items were derived from existing surveys in the literature and input from a multidisciplinary committee. The web-based survey was systematically administered to all MSKCC ambulatory clinicians. Repeated email reminders were sent to optimize responses. Results: 76 breast cancer clinicians at MSKCC and our regional network sites completed the survey between 2/9/09 and 2/25/09. Among respondents, there was widespread agreement (97% (70/72)) that patients should be informed of fertility preservation options, but fewer respondents (51% (37/72)) consistently discussed effects of treatment on fertility with their patients. Only 47% of clinicians (35/74) reported access to information about effects of treatment on fertility. Many physicians cited lack of training in fertility preservation, time constraints, and lack of referral information as barriers to educating patients. No significant difference existed in practice or knowledge between physicians who were practicing for ≤ 5 years vs ≥ 5 years. Conclusions: Physicians report that lack of education, resources and insufficient time hinder fertility preservation discussions with patients. Physicians might benefit from educational efforts regarding the effects of treatment on fertility and new fertility preservation techniques in order to effectively counsel their patients. Time limitations may be overcome by developing educational resources and collaborating with other clinical staff (e.g., nurses) to provide this vital information to patients. These findings are informing an institution-wide educational fertility program. Citation Information: Cancer Res 2010;70(24 Suppl):Abstract nr P5-08-01.
    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
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 7
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 70, No. 24_Supplement ( 2010-12-15), p. P6-11-12-P6-11-12
    Abstract: Background Capecitabine (C) administered for 14 days followed by a 7 day rest (14—7) in combination with lapatinib (L) improved TTP in patients (pts) with HER2(+) metastatic breast cancer (MBC) that progressed following trastuzumab (T). Our mathematical modeling of C dosing predicted 7 days of therapy followed by 7 days of rest (7—7) to maximize efficacy and minimize drug-related toxicity. In xenograph models, C (7—7) was better tolerated than C (14—7) at higher doses and led to improved survival. We established the maximum tolerated dose of C (7—7) to be 2,000 mg twice daily and have studied C (7—7) in combination with targeted therapies. We now report the results of C (7—7) with L in pts with trastuzumab-refractory HER2(+) MBC. Methods Eligible pts had measurable, HER2(+) MBC with progression following T. HER2(+) is defined as 3+ on IHC or FISH-amplified. Pts had normal LVEF by MUGA, ECOG performance status (PS) ≥2, and & lt;3 chemotherapy regimens (CRx) for MBC (no prior fluoropyrimidine). Therapy was C (2,000 mg orally twice daily, 7—7) with L (1,250 mg orally daily); cycle length was 4 weeks. Pts were evaluated for toxicity every 4 weeks and for LVEF by MUGA every 12 weeks. Response was assessed every 12 weeks. Primary endpoint was response rate (RR). Secondary endpoints included toxicity by NCI CTCv3, stable disease (SD) & gt; 6 months, and progression free survival (PFS). Using a Simon optimal 2-stage design, with alpha 10% and power 90% to discriminate between RR 10% and 25%, 21 pts were planned for the 1st stage. If & gt;2 pts responded, then 29 additional pts were to have been enrolled to 2nd stage. If & gt;7/50 pts responded, C (7—7) with L would be considered worthy of further study. Results As of 6/14/10, 22 pts were enrolled on study with the following characteristics: median (med) age 55 years (42-72), med PS 0 (0-2), ER/PR(+) 12, HER2(+) 22, med LVEF 64% (51-70%). Sites of MBC include viscera 16, brain 3. Prior therapy includes 8 pts with adjuvant anthracycline and taxane exposure, 4 pts with adjuvant T, 18 pts with metastatic T, and a med of 1 CRx for MBC. After a med of 5 cycles (1-16), 18 patients are evaluable for response: 0 CR, 3 PR, 6 SD & gt;6 months, 9 SD & lt;6 months. Med PFS is 36 weeks (95% CI 23-not reached). Four pts are too early for response assessment. Nine pts remain on treatment. One pt experienced grade (gr) 4 treatment-related anemia and had refused supportive erythropoietin. Treatment-related gr 2 or 3 toxicities in & gt;10% of patients include: hand-foot syndrome (gr 3: 5%, gr2: 41%), diarrhea (gr 3: 0%, gr 2: 23%), elevated AST/ALT or bilirubin (gr 3: 0%, gr 2: 18%). There have been no declines in LVEF. Conclusion Due to slower than expected accrual and an anticipated randomized, phase III trial comparing C (7—7) with C (14—7), we closed this study to further accrual. However, our data suggests that C (7—7) with lapatinib is active, well-tolerated, feasible and associated with mild gastrointestinal toxicity in pts with trastuzumab-refractory, HER2(+) MBC. Citation Information: Cancer Res 2010;70(24 Suppl):Abstract nr P6-11-12.
    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
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 8
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2011
    In:  Cancer Research Vol. 71, No. 24_Supplement ( 2011-12-15), p. P1-01-03-P1-01-03
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 71, No. 24_Supplement ( 2011-12-15), p. P1-01-03-P1-01-03
    Abstract: Background: Using murine mammary tumor models, recent research conducted by our laboratory at the Sloan-Kettering Institute indicates that select neutrophils are mobilized by a primary breast tumor and uniquely have the capacity to inhibit metastatic seeding in the lung (Granot Z et al. unpublished). We sought to evaluate the cytotoxic role of select neutrophils in the peripheral blood of breast cancer patients as contrasted with those from women without breast cancer and women with DCIS. In addition, we sought to determine whether the addition of select chemokines to neutrophils from a DCIS patient could induce cytotoxicity. Methods: Neutrophils were purified from the blood of 21 newly diagnosed pre-operative breast cancer patients without evidence of metastatic disease, 9 healthy female volunteers with no history of any cancer, and 3 patients with newly diagnosed DCIS. Cytotoxicity was evaluated by incubating isolated neutrophils with luciferase labeled MDA-MB-231 cells. Luciferase activity, as a reflection of% cell kill, was measured using a Bio-Tek microplate luminescence reader. Neutrophils from a DCIS patient with low cytotoxicity were then co-cultured with various CC chemokines (CCl2, CCL3 and CCL5) or CXCL chemokines (CXCL1, CXCL12, and CXCL16) at 100ng/ml. Results: Significant cytotoxicity was notably observed when MDA-MB-231 cells were co-cultured with neutrophils purified from patients with invasive tumors. Pre-operative breast cancer patients (n=21) had a cell kill range of 0–30% (mean = 12.1%), whereas healthy subjects (n=9) had a cell kill range of 0.2-8% (mean = 2.6%), p & lt;0.004. DCIS patients (N=3) had a cell kill range of 3–4% (mean = 2.7). The addition of select chemokines to neutrophils from a DCIS patient with low cytotoxicity (3.2%) resulted in significant increases in cytotoxicity. Table 1 indicates the relative cytotoxicity percentages from the addition of each chemokine. Conclusions: To date, this preliminary work is the first to demonstrate the cytotoxic role of select neutrophils in the peripheral blood of breast cancer patients as contrasted with those from women without breast cancer. We further demonstrate the novel induction of neutrophil cytotoxicity by select chemokines. Further studies are needed to evaluate the prognostic and therapeutic role of cytotoxic neutrophils as well as the role of chemokines in neutrophil cytotoxicity. Citation Information: Cancer Res 2011;71(24 Suppl):Abstract nr P1-01-03.
    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
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 9
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 71, No. 24_Supplement ( 2011-12-15), p. P4-16-06-P4-16-06
    Abstract: Background BM develops in 65–70% of pts with MBC. RANK and its ligand (RANK-L) can be critical in the development and progression of BM. Src overexpression and deregulation occurs in many solid tumors but it has not been fully characterized although an association between Src activity defined by a gene expression signature and BM particularly in ER+ pts has been described. (Zhang XH et al. Cancer Cell. 2009) Our goal was to elucidate the relationship between Src and RANK expression in BT and BM in relation to estrogen-/progesterone-receptor (ER/PR)/HER2 expression and tumor histology (invasive ductal carcinoma (IDC) vs invasive lobular carcinoma (ILC)). Methods: Immunohistochemistry (IHC) for RANK (R & D Systems clone 80707) and Src (Cell Applications Inc. Phospho Tyr-416) protein expression was performed on archived paraffin embedded BT and BM. Scoring: 0=negative, 1+=weak, 2+=intermediate, 3+=strong and the percent of positive tumor cells; RANK+ = 2–3+, & gt; 1% of cells; Src+ = 1–3+; & gt; 1% of cells. Associations between RANK/Src expression and tumor characteristics were assessed using the chi-square test or McNemar's test for pairs, as appropriate. Results: From the MSKCC database, using an IRB-approved waiver of consent, we identified 54 pts with MBC who underwent surgical biopsy of a metastatic bone lesion at our center between 2005–2010, and had tissue available for further testing. 17 corresponding BT samples were identified. At the time of diagnosis, 43 (79.5%) primary tumors were ER or PR (+); 6 (11%) were HER2+; 41 (76%) were invasive ductal carcinoma. 87% of BM expressed RANK and 44% expressed Src. (Table 1) No significant correlation between RANK or Src expression in BM and ER/PR/HER2 status of BT was observed. A significant correlation between RANK expression and BT histology was observed, (p=0.0016): 93% of IDC were RANK (+), in comparison to 50% of invasive lobular carcinomas. RANK expression was not significantly different between primary tumor and metastatic bone samples (p=0.99). There was a borderline significant difference in Src expression between primary and metastatic site (p=.06). Conclusions: In our cohort, no correlation between RANK or Src by IHC and ER/PR/HER2 was identified but RANK expression was more common in IDC than ILC. Fidelity was high for RANK between primary and metastatic lesions while Src expression may possibly vary. Citation Information: Cancer Res 2011;71(24 Suppl):Abstract nr P4-16-06.
    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
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 10
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 71, No. 24_Supplement ( 2011-12-15), p. PD09-08-PD09-08
    Abstract: Background: Hyperactivation of the PI3K-AKT-mTOR pathway is a postulated mechanism of resistance to anti-HER2 therapies and has also been described in triple-negative breast tumors. In HER2−amplified (HER2+) laboratory models, inhibition of this pathway induces activation of upstream receptor tyrosine kinases such as HER3. In triple-negative breast cancer (TN), HER1 overexpression has been identified and models show sensitivity to combined HER1 and mTOR inhibition. We hypothesize that dual inhibition of the PI3K pathway, HER1/2, and induced HER3 may be highly effective in patients with HER2+ or TN breast cancer (BC). This phase I/II trial is designed to determine the tolerability and possible efficacy of the mTOR inhibitor temsirolimus (T) plus the HER1/2 inhibitor neratinib (N) in patients with trastuzumab-refractory, HER2+ or TN BC. We will also explore mutational activation of the PI3K pathway in trastuzumab-refractory tumors as it relates to response to the T-N combination. Methods: The phase I dose-escalation study evaluated T (flat dose IV weekly) plus N (240 mg oral daily) in patients with metastatic HER2+ or TN BC. Cycle length was 4 weeks. Phase I end points included definition of maximum tolerated dose (MTD) and response rate (RR) per RECIST. The phase II study has a Simon two-stage design and evaluates the HER2+ and TN patients separately. Phase II endpoints include progression free survival and duration of response. Response was evaluated radiographically every 8 weeks, toxicity assessed every 2 weeks. All patients underwent biopsy of metastatic disease for biomarker assessment. Activating mutations in PIK3CA were assayed using the Sequenom MassARRAY system. Expression of PTEN was assessed by immunohistochemistry utilizing a published scoring system. Results: The phase I study enrolled 8 HER2+ patients who received a median of 5 (1-13) cycles of therapy. All patients had received trastuzumab and a median of 5.5 (2-12) prior lines of therapy. Frequent treatment-related grade 2 events were: hyperglycemia (4/8), elevated CPK (3/8), diarrhea (2/8), and rash (2/8). Grade 3 diarrhea was the dose-limiting toxicity. Other grade 3 toxicity was hyperglycemia (1/8); hematologic toxicities were not observed. The MTD of temsirolimus with neratinib is 8 mg IV weekly. Six patients treated at MTD were evaluable for response; 4 patients had PR, 1 had SD for a RR of 67%. PI3K pathway activation, through PIK3CA mutational activation or PTEN loss, was identified in 4/6 tumors analyzed and did not preclude response to temsirolimus and neratinib. Updated results reflecting the phase II patients will be reported. Conclusions: Temsirolimus plus neratinib is active in trastuzumab-refractory HER2+ patients. The phase II study is ongoing and additional efficacy and safety data in both HER2−amplified and triple-negative breast cancer patients will be presented. Citation Information: Cancer Res 2011;71(24 Suppl):Abstract nr PD09-08.
    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
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. Further information can be found on the KOBV privacy pages