146 research outputs found

    Observing the earliest moments of supernovae using strong gravitational lenses

    Get PDF
    We determine the viability of exploiting lensing time delays to observe strongly gravitationally lensed supernovae (gLSNe) from first light. Assuming a plausible discovery strategy, the Legacy Survey of Space and Time (LSST) and the Zwicky Transient Facility (ZTF) will discover \sim 110 and \sim 1 systems per year before the supernova (SN) explosion in the final image respectively. Systems will be identified 11.79.3+29.811.7^{+29.8}_{-9.3} days before the final explosion. We then explore the possibility of performing early-time observations for Type IIP and Type Ia SNe in LSST-discovered systems. Using a simulated Type IIP explosion, we predict that the shock breakout in one trailing image per year will peak at \lesssim 24.1 mag (\lesssim 23.3) in the BB-band (F218WF218W), however evolving over a timescale of \sim 30 minutes. Using an analytic model of Type Ia companion interaction, we find that in the BB-band we should observe at least one shock cooling emission event per year that peaks at \lesssim 26.3 mag (\lesssim 29.6) assuming all Type Ia gLSNe have a 1 M_\odot red giant (main sequence) companion. We perform Bayesian analysis to investigate how well deep observations with 1 hour exposures on the European Extremely Large Telescope would discriminate between Type Ia progenitor populations. We find that if all Type Ia SNe evolved from the double-degenerate channel, then observations of the lack of early blue flux in 10 (50) trailing images would rule out more than 27% (19%) of the population having 1 M_\odot main sequence companions at 95% confidence.Comment: 17 pages, 15 figures (including appendices). Accepted by MNRAS 3rd May 202

    The Impact of Microlensing on the Standardisation of Strongly Lensed Type Ia Supernovae

    Get PDF
    We investigate the effect of microlensing on the standardisation of strongly lensed Type Ia supernovae (GLSNe Ia). We present predictions for the amount of scatter induced by microlensing across a range of plausible strong lens macromodels. We find that lensed images in regions of low convergence, shear and stellar density are standardisable, where the microlensing scatter is < 0.15 magnitudes, comparable to the intrinsic dispersion of for a typical SN Ia. These standardisable configurations correspond to asymmetric lenses with an image located far outside the Einstein radius of the lens. Symmetric and small Einstein radius lenses (< 0.5 arcsec) are not standardisable. We apply our model to the recently discovered GLSN Ia iPTF16geu and find that the large discrepancy between the observed flux and the macromodel predictions from More et al. (2017) cannot be explained by microlensing alone. Using the mock GLSNe Ia catalogue of Goldstein et al. (2017), we predict that ~ 22% of GLSNe Ia discovered by LSST will be standardisable, with a median Einstein radius of 0.9 arcseconds and a median time-delay of 41 days. By breaking the mass-sheet degeneracy the full LSST GLSNe Ia sample will be able to detect systematics in H0 at the 0.5% level.Comment: 11 pages, 8 Figures. Accepted by MNRAS May 17 201

    Observing the earliest moments of supernovae using strong gravitational lenses

    Get PDF
    We determine the viability of exploiting lensing time delays to observe strongly gravitationally lensed supernovae (gLSNe) from first light. Assuming a plausible discovery strategy, the Legacy Survey of Space and Time (LSST) and the Zwicky Transient Facility (ZTF) will discover ∼110 and ∼1 systems per year before the supernova (SN) explosion in the final image, respectively. Systems will be identified 11.7^(+29.8)_(−9.3) d before the final explosion. We then explore the possibility of performing early-time observations for Type IIP and Type Ia SNe in LSST-discovered systems. Using a simulated Type IIP explosion, we predict that the shock breakout in one trailing image per year will peak at ≲24.1 mag (≲23.3) in the B-band (F218W), however evolving over a time-scale of ∼30 min. Using an analytic model of Type Ia companion interaction, we find that in the B-band we should observe at least one shock cooling emission event per year that peaks at ≲26.3 mag (≲29.6) assuming all Type Ia gLSNe have a 1 M_⊙ red giant (main sequence) companion. We perform Bayesian analysis to investigate how well deep observations with 1 h exposures on the European Extremely Large Telescope would discriminate between Type Ia progenitor populations. We find that if all Type Ia SNe evolved from the double-degenerate channel, then observations of the lack of early blue flux in 10 (50) trailing images would rule out more than 27 per cent (19 per cent) of the population having 1 M_⊙ main sequence companions at 95 per cent confidence

    White matter structure and myelin-related gene expression alterations with experience in adult rats

    Get PDF
    White matter (WM) plasticity during adulthood is a recently described phenomenon by which experience can shape brain structure. It has been observed in humans using diffusion tensor imaging (DTI) and myelination has been suggested as a possible mechanism. Here, we set out to identify molecular and cellular changes associated with WM plasticity measured by DTI. We combined DTI, immunohistochemistry and mRNA expression analysis and examined the effects of somatosensory experience in adult rats. First, we observed experience-induced DTI differences in WM and in grey matter structure. C-Fos mRNA expression, a marker of cortical activity, in the barrel cortex correlated with the MRI WM metrics, indicating that molecular correlates of cortical activity relate to macroscale measures of WM structure. Analysis of myelin-related genes revealed higher myelin basic protein (MBP) mRNA expression. Higher MBP protein expression was also found via immunohistochemistry in WM. Finally, unbiased RNA sequencing analysis identified 134 differentially expressed genes encoding proteins in- volved in functions related to cell proliferation and differentiation, regulation of myelination and neuronal activity modulation. In conclusion, macroscale measures of WM plasticity are supported by both molecular and cellular evidence and confirm that myelination is one of the underlying mechanisms

    Capivasertib, an AKT Kinase Inhibitor, as Monotherapy or in Combination With Fulvestrant in Patients With AKT1 E17K-Mutant, ER-Positive Metastatic Breast Cancer.

    Get PDF
    PURPOSE:The activating mutation AKT1 E17K occurs in ~7% of ER+ metastatic breast cancer (MBC). We report, from a multipart, first-in-human, Phase I study (NCT01226316), tolerability and activity of capivasertib, an oral AKT inhibitor, as monotherapy or combined with fulvestrant in expansion cohorts of AKT1 E17K-mutant ER+ MBC patients. PATIENTS AND METHODS:Patients with an AKT1 E17K mutation, detected by local (NGS) or central (plasma-based BEAMing) testing, received capivasertib 480 mg bid, 4 days on, 3 days off, weekly or 400 mg bid combined with fulvestrant at the labeled dose. Study endpoints included safety, objective response rate (ORR; RECIST v1.1), progression-free survival (PFS) and clinical benefit rate at 24 weeks (CBR24). Biomarker analyses were conducted in the combination cohort. RESULTS:From October 2013 to August 2018, 63 heavily pretreated patients received capivasertib (20 monotherapy, 43 combination). ORR was 20% with monotherapy, and within the combination cohort was 36% in fulvestrant-pretreated and 20% in fulvestrant-naïve patients, although this latter group may have had more aggressive disease at baseline. AKT1 E17K mutations were detectable in plasma by BEAMing (95%, 41/43), ddPCR (80%, 33/41) and NGS (76%, 31/41). A 50% decrease in AKT1 E17K at cycle 2 day 1 was associated with improved PFS. Combination therapy appeared more tolerable than monotherapy (most frequent grade ≥3 adverse events: rash [9% vs 20%], hyperglycemia [5% vs 30%], diarrhea [5% vs 10%]). CONCLUSIONS:Capivasertib demonstrated clinically meaningful activity in heavily pretreated AKT1 E17K-mutant ER+ MBC patients, including those with prior disease progression on fulvestrant. Tolerability and activity appeared improved by the combination

    Phase I Trial of the PARP Inhibitor Olaparib and AKT Inhibitor Capivasertib in Patients with <i>BRCA1/2</i>- and Non-<i>BRCA1/2</i>-Mutant Cancers.

    Get PDF
    Preclinical studies have demonstrated synergy between PARP and PI3K/AKT pathway inhibitors in BRCA1 and BRCA2 (BRCA1/2)-deficient and BRCA1/2-proficient tumors. We conducted an investigator-initiated phase I trial utilizing a prospective intrapatient dose- escalation design to assess two schedules of capivasertib (AKT inhibitor) with olaparib (PARP inhibitor) in 64 patients with advanced solid tumors. Dose expansions enrolled germline BRCA1/2-mutant tumors, or BRCA1/2 wild-type cancers harboring somatic DNA damage response (DDR) or PI3K-AKT pathway alterations. The combination was well tolerated. Recommended phase II doses for the two schedules were: olaparib 300 mg twice a day with either capivasertib 400 mg twice a day 4 days on, 3 days off, or capivasertib 640 mg twice a day 2 days on, 5 days off. Pharmacokinetics were dose proportional. Pharmacodynamic studies confirmed phosphorylated (p) GSK3β suppression, increased pERK, and decreased BRCA1 expression. Twenty-five (44.6%) of 56 evaluable patients achieved clinical benefit (RECIST complete response/partial response or stable disease ≥ 4 months), including patients with tumors harboring germline BRCA1/2 mutations and BRCA1/2 wild-type cancers with or without DDR and PI3K-AKT pathway alterations. SIGNIFICANCE: In the first trial to combine PARP and AKT inhibitors, a prospective intrapatient dose- escalation design demonstrated safety, tolerability, and pharmacokinetic-pharmacodynamic activity and assessed predictive biomarkers of response/resistance. Antitumor activity was observed in patients harboring tumors with germline BRCA1/2 mutations and BRCA1/2 wild-type cancers with or without somatic DDR and/or PI3K-AKT pathway alterations.This article is highlighted in the In This Issue feature, p. 1426

    Fulvestrant plus capivasertib versus placebo after relapse or progression on an aromatase inhibitor in metastatic, oestrogen receptor-positive, HER2-negative breast cancer (FAKTION): overall survival, updated progression-free survival, and expanded biomarker analysis from a randomised, phase 2 trial

    Get PDF
    Background Capivasertib, an AKT inhibitor, added to fulvestrant, was previously reported to improve progression-free survival in women with aromatase inhibitor-resistant oestrogen receptor (ER)-positive, HER2-negative advanced breast cancer. The benefit appeared to be independent of the phosphoinositide 3-kinase (PI3K)/AKT/phosphatase and tensin homologue (PTEN) pathway alteration status of tumours, as ascertained using assays available at the time. Here, we report updated progression-free survival and overall survival results, and a prespecified examination of the effect of PI3K/AKT/PTEN pathway alterations identified by an expanded genetic testing panel on treatment outcomes. Methods This randomised, multicentre, double-blind, placebo-controlled, phase 2 trial recruited postmenopausal adult women aged at least 18 years with ER-positive, HER2-negative, metastatic or locally advanced inoperable breast cancer and an Eastern Cooperative Oncology Group performance status of 0–2, who had relapsed or progressed on an aromatase inhibitor, from across 19 hospitals in the UK. Participants were randomly assigned (1:1) to receive intramuscular fulvestrant 500 mg (day 1) every 28 days (plus a 500 mg loading dose on day 15 of cycle 1) with either capivasertib 400 mg or matching placebo, orally twice daily on an intermittent weekly schedule of 4 days on and 3 days off, starting on cycle 1 day 15. Treatment continued until disease progression, unacceptable toxicity, loss to follow-up, or withdrawal of consent. Treatment was allocated by an interactive web-response system using a minimisation method (with a 20% random element) and the following minimisation factors: measurable or non-measurable disease, primary or secondary aromatase inhibitor resistance, PIK3CA status, and PTEN status. The primary endpoint was progression-free survival in the intention-to-treat population. Secondary endpoints shown in this Article were overall survival and safety in the intention-to-treat population, and the effect of tumour PI3K/AKT/PTEN pathway status identified by an expanded testing panel that included next-generation sequencing assays. Recruitment is complete. The trial is registered with ClinicalTrials.gov, number NCT01992952. Findings Between March 16, 2015, and March 6, 2018, 183 participants were screened for eligibility and 140 (77%) were randomly assigned to receive fulvestrant plus capivasertib (n=69) or fulvestrant plus placebo (n=71). Median follow-up at the data cut-off of Nov 25, 2021, was 58·5 months (IQR 45·9–64·1) for participants treated with fulvestrant plus capivasertib and 62·3 months (IQR 62·1–70·3) for fulvestrant plus placebo. Updated median progression-free survival was 10·3 months (95% CI 5·0–13·4) in the group receiving fulvestrant plus capivasertib compared with 4·8 months (3·1–7·9) for fulvestrant plus placebo (adjusted hazard ratio [HR] 0·56 [95% CI 0·38–0·81]; two-sided p=0·0023). Median overall survival in the capivasertib versus placebo groups was 29·3 months (95% CI 23·7–39·0) versus 23·4 months (18·7–32·7; adjusted HR 0·66 [95% CI 0·45–0·97]; two-sided p=0·035). The expanded biomarker panel identified an expanded pathway-altered subgroup that contained 76 participants (54% of the intention-to-treat population). Median progression-free survival in the expanded pathway-altered subgroup for participants receiving capivasertib (n=39) was 12·8 months (95% CI 6·6–18·8) compared with 4·6 months (2·8–7·9) in the placebo group (n=37; adjusted HR 0·44 [95% CI 0·26–0·72]; two-sided p=0·0014). Median overall survival for the expanded pathway-altered subgroup receiving capivasertib was 38·9 months (95% CI 23·3–50·7) compared with 20·0 months (14·8–31·4) for those receiving placebo (adjusted HR 0·46 [95% CI 0·27–0·79]; two-sided p=0·0047). By contrast, there were no statistically significant differences in progression-free or overall survival in the expanded pathway non-altered subgroup treated with capivasertib (n=30) versus placebo (n=34). One additional serious adverse event (pneumonia) in the capivasertib group had occurred subsequent to the primary analysis. One death, due to atypical pulmonary infection, was assessed as possibly related to capivasertib treatment. Interpretation Updated FAKTION data showed that capivasertib addition to fulvestrant extends the survival of participants with aromatase inhibitor-resistant ER-positive, HER2-negative advanced breast cancer. The expanded biomarker testing suggested that capivasertib predominantly benefits patients with PI3K/AKT/PTEN pathway-altered tumours. Phase 3 data are needed to substantiate the results, including in patients with previous CDK4/6 inhibitor exposure who were not included in the FAKTION trial. Funding AstraZeneca and Cancer Research UK

    Proliferation and AKT activity biomarker analyses after capivasertib (AZD5363) treatment of patients with ER+ invasive breast cancer (STAKT)

    Get PDF
    Purpose: The STAKT study examined short-term exposure (4.5 days) to the oral selective pan-AKT inhibitor capivasertib (AZD5363) to determine if this drug can reach its therapeutic target in sufficient concentration to significantly modulate key biomarkers of the AKT pathway and tumor proliferation. Patients and Methods: STAKT was a two-stage, double-blind, randomized, placebo-controlled, “window-of-opportunity” study in patients with newly diagnosed ER+ invasive breast cancer. Stage 1 assessed capivasertib 480 mg b.i.d. (recommended monotherapy dose) and placebo, and stage 2 assessed capivasertib 360 and 240 mg b.i.d. Primary endpoints were changes from baseline in AKT pathway markers pPRAS40, pGSK3β, and proliferation protein Ki67. Pharmacologic and pharmacodynamic properties were analyzed from blood sampling, and tolerability by adverse-event monitoring. Results: After 4.5 days' exposure, capivasertib 480 mg b.i.d. (n = 17) produced significant decreases from baseline versus placebo (n = 11) in pGSK3β (H-score absolute change: −55.3, P = 0.006) and pPRAS40 (−83.8, P < 0.0001), and a decrease in Ki67 (absolute change in percentage positive nuclei: −9.6%, P = 0.031). Significant changes also occurred in secondary signaling biomarker pS6 (−42.3, P = 0.004), while pAKT (and nuclear FOXO3a) also increased in accordance with capivasertib's mechanism (pAKT: 81.3, P = 0.005). At doses of 360 mg b.i.d. (n = 5) and 240 mg b.i.d. (n = 6), changes in primary and secondary biomarkers were also observed, albeit of smaller magnitude. Biomarker modulation was dose and concentration dependent, and no new safety signals were evident. Conclusions: Capivasertib 480 mg b.i.d. rapidly modulates key biomarkers of the AKT pathway and decreases proliferation marker Ki67, suggesting future potential as an effective therapy in AKT-dependent breast cancers
    corecore