56 research outputs found

    SKFlow: Learning Optical Flow with Super Kernels

    Full text link
    Optical flow estimation is a classical yet challenging task in computer vision. One of the essential factors in accurately predicting optical flow is to alleviate occlusions between frames. However, it is still a thorny problem for current top-performing optical flow estimation methods due to insufficient local evidence to model occluded areas. In this paper, we propose the Super Kernel Flow Network (SKFlow), a CNN architecture to ameliorate the impacts of occlusions on optical flow estimation. SKFlow benefits from the super kernels which bring enlarged receptive fields to complement the absent matching information and recover the occluded motions. We present efficient super kernel designs by utilizing conical connections and hybrid depth-wise convolutions. Extensive experiments demonstrate the effectiveness of SKFlow on multiple benchmarks, especially in the occluded areas. Without pre-trained backbones on ImageNet and with a modest increase in computation, SKFlow achieves compelling performance and ranks 1st\textbf{1st} among currently published methods on the Sintel benchmark. On the challenging Sintel clean and final passes (test), SKFlow surpasses the best-published result in the unmatched areas (7.967.96 and 12.5012.50) by 9.09%9.09\% and 7.92%7.92\%. The code is available at \href{https://github.com/littlespray/SKFlow}{https://github.com/littlespray/SKFlow}.Comment: Accepted to NeurIPS 202

    Actively implementing an evidence-based feeding guideline for critically ill patients (NEED): a multicenter, cluster-randomized, controlled trial

    Get PDF
    Background: Previous cluster-randomized controlled trials evaluating the impact of implementing evidence-based guidelines for nutrition therapy in critical illness do not consistently demonstrate patient benefits. A large-scale, sufficiently powered study is therefore warranted to ascertain the effects of guideline implementation on patient-centered outcomes. Methods: We conducted a multicenter, cluster-randomized, parallel-controlled trial in intensive care units (ICUs) across China. We developed an evidence-based feeding guideline. ICUs randomly allocated to the guideline group formed a local "intervention team", which actively implemented the guideline using standardized educational materials, a graphical feeding protocol, and live online education outreach meetings conducted by members of the study management committee. ICUs assigned to the control group remained unaware of the guideline content. All ICUs enrolled patients who were expected to stay in the ICU longer than seven days. The primary outcome was all-cause mortality within 28 days of enrollment. Results: Forty-eight ICUs were randomized to the guideline group and 49 to the control group. From March 2018 to July 2019, the guideline ICUs enrolled 1399 patients, and the control ICUs enrolled 1373 patients. Implementation of the guideline resulted in significantly earlier EN initiation (1.20 vs. 1.55 mean days to initiation of EN; difference − 0.40 [95% CI − 0.71 to − 0.09]; P = 0.01) and delayed PN initiation (1.29 vs. 0.80 mean days to start of PN; difference 1.06 [95% CI 0.44 to 1.67]; P = 0.001). There was no significant difference in 28-day mortality (14.2% vs. 15.2%; difference − 1.6% [95% CI − 4.3% to 1.2%]; P = 0.42) between groups. Conclusions: In this large-scale, multicenter trial, active implementation of an evidence-based feeding guideline reduced the time to commencement of EN and overall PN use but did not translate to a reduction in mortality from critical illness. Trial registration: ISRCTN, ISRCTN12233792. Registered November 20th, 2017

    Actively implementing an evidence-based feeding guideline for critically ill patients (NEED): a multicenter, cluster-randomized, controlled trial.

    Get PDF
    BackgroundPrevious cluster-randomized controlled trials evaluating the impact of implementing evidence-based guidelines for nutrition therapy in critical illness do not consistently demonstrate patient benefits. A large-scale, sufficiently powered study is therefore warranted to ascertain the effects of guideline implementation on patient-centered outcomes.MethodsWe conducted a multicenter, cluster-randomized, parallel-controlled trial in intensive care units (ICUs) across China. We developed an evidence-based feeding guideline. ICUs randomly allocated to the guideline group formed a local "intervention team", which actively implemented the guideline using standardized educational materials, a graphical feeding protocol, and live online education outreach meetings conducted by members of the study management committee. ICUs assigned to the control group remained unaware of the guideline content. All ICUs enrolled patients who were expected to stay in the ICU longer than seven days. The primary outcome was all-cause mortality within 28 days of enrollment.ResultsForty-eight ICUs were randomized to the guideline group and 49 to the control group. From March 2018 to July 2019, the guideline ICUs enrolled 1399 patients, and the control ICUs enrolled 1373 patients. Implementation of the guideline resulted in significantly earlier EN initiation (1.20 vs. 1.55 mean days to initiation of EN; difference - 0.40 [95% CI - 0.71 to - 0.09]; P = 0.01) and delayed PN initiation (1.29 vs. 0.80 mean days to start of PN; difference 1.06 [95% CI 0.44 to 1.67]; P = 0.001). There was no significant difference in 28-day mortality (14.2% vs. 15.2%; difference - 1.6% [95% CI - 4.3% to 1.2%]; P = 0.42) between groups.ConclusionsIn this large-scale, multicenter trial, active implementation of an evidence-based feeding guideline reduced the time to commencement of EN and overall PN use but did not translate to a reduction in mortality from critical illness.Trial registrationISRCTN, ISRCTN12233792 . Registered November 20th, 2017

    Actively implementing an evidence-based feeding guideline for critically ill patients (NEED): a multicenter, cluster-randomized, controlled trial (vol 26, 46, 2022)

    Get PDF
    BackgroundPrevious cluster-randomized controlled trials evaluating the impact of implementing evidence-based guidelines for nutrition therapy in critical illness do not consistently demonstrate patient benefits. A large-scale, sufficiently powered study is therefore warranted to ascertain the effects of guideline implementation on patient-centered outcomes.MethodsWe conducted a multicenter, cluster-randomized, parallel-controlled trial in intensive care units (ICUs) across China. We developed an evidence-based feeding guideline. ICUs randomly allocated to the guideline group formed a local "intervention team", which actively implemented the guideline using standardized educational materials, a graphical feeding protocol, and live online education outreach meetings conducted by members of the study management committee. ICUs assigned to the control group remained unaware of the guideline content. All ICUs enrolled patients who were expected to stay in the ICU longer than seven days. The primary outcome was all-cause mortality within 28 days of enrollment.ResultsForty-eight ICUs were randomized to the guideline group and 49 to the control group. From March 2018 to July 2019, the guideline ICUs enrolled 1399 patients, and the control ICUs enrolled 1373 patients. Implementation of the guideline resulted in significantly earlier EN initiation (1.20 vs. 1.55 mean days to initiation of EN; difference - 0.40 [95% CI - 0.71 to - 0.09]; P = 0.01) and delayed PN initiation (1.29 vs. 0.80 mean days to start of PN; difference 1.06 [95% CI 0.44 to 1.67]; P = 0.001). There was no significant difference in 28-day mortality (14.2% vs. 15.2%; difference - 1.6% [95% CI - 4.3% to 1.2%]; P = 0.42) between groups.ConclusionsIn this large-scale, multicenter trial, active implementation of an evidence-based feeding guideline reduced the time to commencement of EN and overall PN use but did not translate to a reduction in mortality from critical illness.Trial registrationISRCTN, ISRCTN12233792 . Registered November 20th, 2017

    SYNTHESIS AND STRUCTURES OF TRIS

    No full text

    Medical supervised masked autoencoders: Crafting a better masking strategy and efficient fine-tuning schedule for medical image classification

    Full text link
    Masked autoencoders (MAEs) have displayed significant potential in the classification and semantic segmentation of medical images in the last year. Due to the high similarity of human tissues, even slight changes in medical images may represent diseased tissues, necessitating fine-grained inspection to pinpoint diseased tissues. The random masking strategy of MAEs is likely to result in areas of lesions being overlooked by the model. At the same time, inconsistencies between the pre-training and fine-tuning phases impede the performance and efficiency of MAE in medical image classification. To address these issues, we propose a medical supervised masked autoencoder (MSMAE) in this paper. In the pre-training phase, MSMAE precisely masks medical images via the attention maps obtained from supervised training, contributing to the representation learning of human tissue in the lesion area. During the fine-tuning phase, MSMAE is also driven by attention to the accurate masking of medical images. This improves the computational efficiency of the MSMAE while increasing the difficulty of fine-tuning, which indirectly improves the quality of MSMAE medical diagnosis. Extensive experiments demonstrate that MSMAE achieves state-of-the-art performance in case with three official medical datasets for various diseases. Meanwhile, transfer learning for MSMAE also demonstrates the great potential of our approach for medical semantic segmentation tasks. Moreover, the MSMAE accelerates the inference time in the fine-tuning phase by 11.2% and reduces the number of floating-point operations (FLOPs) by 74.08% compared to a traditional MAE

    A generalized solid strengthening rule for biocompatible Zn-based alloys, a comparison with Mg-based alloys

    No full text
    Solid solution strengthening has been widely used in designing various high-performance biocompatible Mg-based alloys, but its transferability to other biocompatible metals such as Zn-based alloys is questionable or nearly absent. In the present study, an ab initio informed Peierls-Nabarro model and Leyson et al.'s strengthening model are used for a systematic investigation on solute strengthening in Zn-based alloys, which is compared with the widely studied Mg-based alloys. Although an inverse relationship was revealed between volume misfit epsilon (b) and chemical misfit epsilon (SFE) for both Zn-based and Mg-based alloys, most solutes would however result in positive epsilon (b) and negative epsilon (SFE) for Zn-based alloys, differing from Mg-based alloys. With epsilon (b) and epsilon (SFE) as two key descriptors, a generalized scaling diagram is finally drawn for a fast evaluation of solid solution strengthening in Zn-based alloys, indicating that the alkaline-earth and rare earth elements are better strengtheners for Zn-based alloys, which provides a general rule in designing novel biocompatible materials.Web of Science2140226382262

    All-Optical Non-Inverted Parity Generator and Checker Based on Semiconductor Optical Amplifiers

    No full text
    An all-optical non-inverted parity generator and checker based on semiconductor optical amplifiers (SOAs) are proposed with four-wave mixing (FWM) and cross-gain modulation (XGM) non-linear effects. A 2-bit parity generator and checker using by exclusive NOR (XNOR) and exclusive OR (XOR) gates are implemented by first SOA and second SOA with 10 Gb/s return-to-zero (RZ) code, respectively. The parity and check bits are provided by adjusting the center wavelength of the tunable optical bandpass filter (TOBPF). A saturable absorber (SA) is used to reduce the negative effect of small signal clock (Clk) probe light to improve extinction ratio (ER) and optical signal-to-noise ratio (OSNR). For Pe and Ce (even parity bit and even check bit) without Clk probe light, ER and OSNR still maintain good performance because of the amplified effect of SOA. For Po (odd parity bit), ER and OSNR are improved to 1 dB difference for the original value. For Co (odd check bit), ER is deteriorated by 4 dB without SA, while OSNR is deteriorated by 12 dB. ER and OSNR are improved by about 2 dB for the original value with the SA. This design has the advantages of simple structure and great integration capability and low cost
    corecore