new

Get trending papers in your email inbox!

Subscribe

Daily Papers

byAK and the research community

Sep 2

SADM: Sequence-Aware Diffusion Model for Longitudinal Medical Image Generation

Human organs constantly undergo anatomical changes due to a complex mix of short-term (e.g., heartbeat) and long-term (e.g., aging) factors. Evidently, prior knowledge of these factors will be beneficial when modeling their future state, i.e., via image generation. However, most of the medical image generation tasks only rely on the input from a single image, thus ignoring the sequential dependency even when longitudinal data is available. Sequence-aware deep generative models, where model input is a sequence of ordered and timestamped images, are still underexplored in the medical imaging domain that is featured by several unique challenges: 1) Sequences with various lengths; 2) Missing data or frame, and 3) High dimensionality. To this end, we propose a sequence-aware diffusion model (SADM) for the generation of longitudinal medical images. Recently, diffusion models have shown promising results in high-fidelity image generation. Our method extends this new technique by introducing a sequence-aware transformer as the conditional module in a diffusion model. The novel design enables learning longitudinal dependency even with missing data during training and allows autoregressive generation of a sequence of images during inference. Our extensive experiments on 3D longitudinal medical images demonstrate the effectiveness of SADM compared with baselines and alternative methods. The code is available at https://github.com/ubc-tea/SADM-Longitudinal-Medical-Image-Generation.

METAGENE-1: Metagenomic Foundation Model for Pandemic Monitoring

We pretrain METAGENE-1, a 7-billion-parameter autoregressive transformer model, which we refer to as a metagenomic foundation model, on a novel corpus of diverse metagenomic DNA and RNA sequences comprising over 1.5 trillion base pairs. This dataset is sourced from a large collection of human wastewater samples, processed and sequenced using deep metagenomic (next-generation) sequencing methods. Unlike genomic models that focus on individual genomes or curated sets of specific species, the aim of METAGENE-1 is to capture the full distribution of genomic information present within this wastewater, to aid in tasks relevant to pandemic monitoring and pathogen detection. We carry out byte-pair encoding (BPE) tokenization on our dataset, tailored for metagenomic sequences, and then pretrain our model. In this paper, we first detail the pretraining dataset, tokenization strategy, and model architecture, highlighting the considerations and design choices that enable the effective modeling of metagenomic data. We then show results of pretraining this model on our metagenomic dataset, providing details about our losses, system metrics, and training stability over the course of pretraining. Finally, we demonstrate the performance of METAGENE-1, which achieves state-of-the-art results on a set of genomic benchmarks and new evaluations focused on human-pathogen detection and genomic sequence embedding, showcasing its potential for public health applications in pandemic monitoring, biosurveillance, and early detection of emerging health threats.

SuSana Distancia is all you need: Enforcing class separability in metric learning via two novel distance-based loss functions for few-shot image classification

Few-shot learning is a challenging area of research that aims to learn new concepts with only a few labeled samples of data. Recent works based on metric-learning approaches leverage the meta-learning approach, which is encompassed by episodic tasks that make use a support (training) and query set (test) with the objective of learning a similarity comparison metric between those sets. Due to the lack of data, the learning process of the embedding network becomes an important part of the few-shot task. Previous works have addressed this problem using metric learning approaches, but the properties of the underlying latent space and the separability of the difference classes on it was not entirely enforced. In this work, we propose two different loss functions which consider the importance of the embedding vectors by looking at the intra-class and inter-class distance between the few data. The first loss function is the Proto-Triplet Loss, which is based on the original triplet loss with the modifications needed to better work on few-shot scenarios. The second loss function, which we dub ICNN loss is based on an inter and intra class nearest neighbors score, which help us to assess the quality of embeddings obtained from the trained network. Our results, obtained from a extensive experimental setup show a significant improvement in accuracy in the miniImagenNet benchmark compared to other metric-based few-shot learning methods by a margin of 2%, demonstrating the capability of these loss functions to allow the network to generalize better to previously unseen classes. In our experiments, we demonstrate competitive generalization capabilities to other domains, such as the Caltech CUB, Dogs and Cars datasets compared with the state of the art.

TinyViT: Fast Pretraining Distillation for Small Vision Transformers

Vision transformer (ViT) recently has drawn great attention in computer vision due to its remarkable model capability. However, most prevailing ViT models suffer from huge number of parameters, restricting their applicability on devices with limited resources. To alleviate this issue, we propose TinyViT, a new family of tiny and efficient small vision transformers pretrained on large-scale datasets with our proposed fast distillation framework. The central idea is to transfer knowledge from large pretrained models to small ones, while enabling small models to get the dividends of massive pretraining data. More specifically, we apply distillation during pretraining for knowledge transfer. The logits of large teacher models are sparsified and stored in disk in advance to save the memory cost and computation overheads. The tiny student transformers are automatically scaled down from a large pretrained model with computation and parameter constraints. Comprehensive experiments demonstrate the efficacy of TinyViT. It achieves a top-1 accuracy of 84.8% on ImageNet-1k with only 21M parameters, being comparable to Swin-B pretrained on ImageNet-21k while using 4.2 times fewer parameters. Moreover, increasing image resolutions, TinyViT can reach 86.5% accuracy, being slightly better than Swin-L while using only 11% parameters. Last but not the least, we demonstrate a good transfer ability of TinyViT on various downstream tasks. Code and models are available at https://github.com/microsoft/Cream/tree/main/TinyViT.

TokenUnify: Scalable Autoregressive Visual Pre-training with Mixture Token Prediction

Autoregressive next-token prediction is a standard pretraining method for large-scale language models, but its application to vision tasks is hindered by the non-sequential nature of image data, leading to cumulative errors. Most vision models employ masked autoencoder (MAE) based pretraining, which faces scalability issues. To address these challenges, we introduce TokenUnify, a novel pretraining method that integrates random token prediction, next-token prediction, and next-all token prediction. We provide theoretical evidence demonstrating that TokenUnify mitigates cumulative errors in visual autoregression. Cooperated with TokenUnify, we have assembled a large-scale electron microscopy (EM) image dataset with ultra-high resolution, ideal for creating spatially correlated long sequences. This dataset includes over 120 million annotated voxels, making it the largest neuron segmentation dataset to date and providing a unified benchmark for experimental validation. Leveraging the Mamba network inherently suited for long-sequence modeling on this dataset, TokenUnify not only reduces the computational complexity but also leads to a significant 45\% improvement in segmentation performance on downstream EM neuron segmentation tasks compared to existing methods. Furthermore, TokenUnify demonstrates superior scalability over MAE and traditional autoregressive methods, effectively bridging the gap between pretraining strategies for language and vision models. Code is available at https://github.com/ydchen0806/TokenUnify.

Memory-Augmented Incomplete Multimodal Survival Prediction via Cross-Slide and Gene-Attentive Hypergraph Learning

Multimodal pathology-genomic analysis is critical for cancer survival prediction. However, existing approaches predominantly integrate formalin-fixed paraffin-embedded (FFPE) slides with genomic data, while neglecting the availability of other preservation slides, such as Fresh Froze (FF) slides. Moreover, as the high-resolution spatial nature of pathology data tends to dominate the cross-modality fusion process, it hinders effective multimodal fusion and leads to modality imbalance challenges between pathology and genomics. These methods also typically require complete data modalities, limiting their clinical applicability with incomplete modalities, such as missing either pathology or genomic data. In this paper, we propose a multimodal survival prediction framework that leverages hypergraph learning to effectively integrate multi-WSI information and cross-modality interactions between pathology slides and genomics data while addressing modality imbalance. In addition, we introduce a memory mechanism that stores previously learned paired pathology-genomic features and dynamically compensates for incomplete modalities. Experiments on five TCGA datasets demonstrate that our model outperforms advanced methods by over 2.3% in C-Index. Under incomplete modality scenarios, our approach surpasses pathology-only (3.3%) and gene-only models (7.9%). Code: https://github.com/MCPathology/M2Surv

Scaling Autoregressive Models for Content-Rich Text-to-Image Generation

We present the Pathways Autoregressive Text-to-Image (Parti) model, which generates high-fidelity photorealistic images and supports content-rich synthesis involving complex compositions and world knowledge. Parti treats text-to-image generation as a sequence-to-sequence modeling problem, akin to machine translation, with sequences of image tokens as the target outputs rather than text tokens in another language. This strategy can naturally tap into the rich body of prior work on large language models, which have seen continued advances in capabilities and performance through scaling data and model sizes. Our approach is simple: First, Parti uses a Transformer-based image tokenizer, ViT-VQGAN, to encode images as sequences of discrete tokens. Second, we achieve consistent quality improvements by scaling the encoder-decoder Transformer model up to 20B parameters, with a new state-of-the-art zero-shot FID score of 7.23 and finetuned FID score of 3.22 on MS-COCO. Our detailed analysis on Localized Narratives as well as PartiPrompts (P2), a new holistic benchmark of over 1600 English prompts, demonstrate the effectiveness of Parti across a wide variety of categories and difficulty aspects. We also explore and highlight limitations of our models in order to define and exemplify key areas of focus for further improvements. See https://parti.research.google/ for high-resolution images.

OTSeq2Set: An Optimal Transport Enhanced Sequence-to-Set Model for Extreme Multi-label Text Classification

Extreme multi-label text classification (XMTC) is the task of finding the most relevant subset labels from an extremely large-scale label collection. Recently, some deep learning models have achieved state-of-the-art results in XMTC tasks. These models commonly predict scores for all labels by a fully connected layer as the last layer of the model. However, such models can't predict a relatively complete and variable-length label subset for each document, because they select positive labels relevant to the document by a fixed threshold or take top k labels in descending order of scores. A less popular type of deep learning models called sequence-to-sequence (Seq2Seq) focus on predicting variable-length positive labels in sequence style. However, the labels in XMTC tasks are essentially an unordered set rather than an ordered sequence, the default order of labels restrains Seq2Seq models in training. To address this limitation in Seq2Seq, we propose an autoregressive sequence-to-set model for XMTC tasks named OTSeq2Set. Our model generates predictions in student-forcing scheme and is trained by a loss function based on bipartite matching which enables permutation-invariance. Meanwhile, we use the optimal transport distance as a measurement to force the model to focus on the closest labels in semantic label space. Experiments show that OTSeq2Set outperforms other competitive baselines on 4 benchmark datasets. Especially, on the Wikipedia dataset with 31k labels, it outperforms the state-of-the-art Seq2Seq method by 16.34% in micro-F1 score. The code is available at https://github.com/caojie54/OTSeq2Set.

Lossless Acceleration for Seq2seq Generation with Aggressive Decoding

We study lossless acceleration for seq2seq generation with a novel decoding algorithm -- Aggressive Decoding. Unlike the previous efforts (e.g., non-autoregressive decoding) speeding up seq2seq generation at the cost of quality loss, our approach aims to yield the identical (or better) generation compared with autoregressive decoding but in a significant speedup, achieved by innovative cooperation of aggressive decoding and verification that are both efficient due to parallel computing. We propose two Aggressive Decoding paradigms for 2 kinds of seq2seq tasks: 1) For the seq2seq tasks whose inputs and outputs are highly similar (e.g., Grammatical Error Correction), we propose Input-guided Aggressive Decoding (IAD) that aggressively copies from the input sentence as drafted decoded tokens to verify in parallel; 2) For other general seq2seq tasks (e.g., Machine Translation), we propose Generalized Aggressive Decoding (GAD) that first employs an additional non-autoregressive decoding model for aggressive decoding and then verifies in parallel in the autoregressive manner. We test Aggressive Decoding on the most popular 6-layer Transformer model on GPU in multiple seq2seq tasks: 1) For IAD, we show that it can introduce a 7x-9x speedup for the Transformer in Grammatical Error Correction and Text Simplification tasks with the identical results as greedy decoding; 2) For GAD, we observe a 3x-5x speedup with the identical or even better quality in two important seq2seq tasks: Machine Translation and Abstractive Summarization. Moreover, Aggressive Decoding can benefit even more from stronger computing devices that are better at parallel computing. Given the lossless quality as well as significant and promising speedup, we believe Aggressive Decoding may potentially evolve into a de facto standard for efficient and lossless seq2seq generation in the near future.

White-Box Diffusion Transformer for single-cell RNA-seq generation

As a powerful tool for characterizing cellular subpopulations and cellular heterogeneity, single cell RNA sequencing (scRNA-seq) technology offers advantages of high throughput and multidimensional analysis. However, the process of data acquisition is often constrained by high cost and limited sample availability. To overcome these limitations, we propose a hybrid model based on Diffusion model and White-Box transformer that aims to generate synthetic and biologically plausible scRNA-seq data. Diffusion model progressively introduce noise into the data and then recover the original data through a denoising process, a forward and reverse process that is particularly suitable for generating complex data distributions. White-Box transformer is a deep learning architecture that emphasizes mathematical interpretability. By minimizing the encoding rate of the data and maximizing the sparsity of the representation, it not only reduces the computational burden, but also provides clear insight into underlying structure. Our White-Box Diffusion Transformer combines the generative capabilities of Diffusion model with the mathematical interpretability of White-Box transformer. Through experiments using six different single-cell RNA-Seq datasets, we visualize both generated and real data using t-SNE dimensionality reduction technique, as well as quantify similarity between generated and real data using various metrics to demonstrate comparable performance of White-Box Diffusion Transformer and Diffusion Transformer in generating scRNA-seq data alongside significant improvements in training efficiency and resource utilization. Our code is available at https://github.com/lingximamo/White-Box-Diffusion-Transformer

Multimodal Optimal Transport-based Co-Attention Transformer with Global Structure Consistency for Survival Prediction

Survival prediction is a complicated ordinal regression task that aims to predict the ranking risk of death, which generally benefits from the integration of histology and genomic data. Despite the progress in joint learning from pathology and genomics, existing methods still suffer from challenging issues: 1) Due to the large size of pathological images, it is difficult to effectively represent the gigapixel whole slide images (WSIs). 2) Interactions within tumor microenvironment (TME) in histology are essential for survival analysis. Although current approaches attempt to model these interactions via co-attention between histology and genomic data, they focus on only dense local similarity across modalities, which fails to capture global consistency between potential structures, i.e. TME-related interactions of histology and co-expression of genomic data. To address these challenges, we propose a Multimodal Optimal Transport-based Co-Attention Transformer framework with global structure consistency, in which optimal transport (OT) is applied to match patches of a WSI and genes embeddings for selecting informative patches to represent the gigapixel WSI. More importantly, OT-based co-attention provides a global awareness to effectively capture structural interactions within TME for survival prediction. To overcome high computational complexity of OT, we propose a robust and efficient implementation over micro-batch of WSI patches by approximating the original OT with unbalanced mini-batch OT. Extensive experiments show the superiority of our method on five benchmark datasets compared to the state-of-the-art methods. The code is released.

MambaMIL: Enhancing Long Sequence Modeling with Sequence Reordering in Computational Pathology

Multiple Instance Learning (MIL) has emerged as a dominant paradigm to extract discriminative feature representations within Whole Slide Images (WSIs) in computational pathology. Despite driving notable progress, existing MIL approaches suffer from limitations in facilitating comprehensive and efficient interactions among instances, as well as challenges related to time-consuming computations and overfitting. In this paper, we incorporate the Selective Scan Space State Sequential Model (Mamba) in Multiple Instance Learning (MIL) for long sequence modeling with linear complexity, termed as MambaMIL. By inheriting the capability of vanilla Mamba, MambaMIL demonstrates the ability to comprehensively understand and perceive long sequences of instances. Furthermore, we propose the Sequence Reordering Mamba (SR-Mamba) aware of the order and distribution of instances, which exploits the inherent valuable information embedded within the long sequences. With the SR-Mamba as the core component, MambaMIL can effectively capture more discriminative features and mitigate the challenges associated with overfitting and high computational overhead. Extensive experiments on two public challenging tasks across nine diverse datasets demonstrate that our proposed framework performs favorably against state-of-the-art MIL methods. The code is released at https://github.com/isyangshu/MambaMIL.

Single-Cell Omics Arena: A Benchmark Study for Large Language Models on Cell Type Annotation Using Single-Cell Data

Over the past decade, the revolution in single-cell sequencing has enabled the simultaneous molecular profiling of various modalities across thousands of individual cells, allowing scientists to investigate the diverse functions of complex tissues and uncover underlying disease mechanisms. Among all the analytical steps, assigning individual cells to specific types is fundamental for understanding cellular heterogeneity. However, this process is usually labor-intensive and requires extensive expert knowledge. Recent advances in large language models (LLMs) have demonstrated their ability to efficiently process and synthesize vast corpora of text to automatically extract essential biological knowledge, such as marker genes, potentially promoting more efficient and automated cell type annotations. To thoroughly evaluate the capability of modern instruction-tuned LLMs in automating the cell type identification process, we introduce SOAR, a comprehensive benchmarking study of LLMs for cell type annotation tasks in single-cell genomics. Specifically, we assess the performance of 8 instruction-tuned LLMs across 11 datasets, spanning multiple cell types and species. Our study explores the potential of LLMs to accurately classify and annotate cell types in single-cell RNA sequencing (scRNA-seq) data, while extending their application to multiomics data through cross-modality translation. Additionally, we evaluate the effectiveness of chain-of-thought (CoT) prompting techniques in generating detailed biological insights during the annotation process. The results demonstrate that LLMs can provide robust interpretations of single-cell data without requiring additional fine-tuning, advancing the automation of cell type annotation in genomics research.

Foundation Models for Zero-Shot Segmentation of Scientific Images without AI-Ready Data

Zero-shot and prompt-based technologies capitalized on using frequently occurring images to transform visual reasoning tasks, which explains why such technologies struggle with valuable yet scarce scientific image sets. In this work, we propose Zenesis, a comprehensive no-code interactive platform designed to minimize barriers posed by data readiness for scientific images. We develop lightweight multi-modal adaptation techniques that enable zero-shot operation on raw scientific data, along with human-in-the-loop refinement and heuristic-based temporal enhancement options. We demonstrate the performance of our approach through comprehensive comparison and validation on challenging Focused Ion Beam Scanning Electron Microscopy (FIB-SEM) data of catalyst-loaded membranes. Zenesis significantly outperforms baseline methods, achieving an average accuracy of 0.947, an Intersection over Union (IOU) of 0.858, and a Dice score of 0.923 for amorphous catalyst samples and accuracy of 0.987, an IOU of 0.857, and a Dice score of 0.923 for crystalline samples. These results mark a substantial improvement over traditional methods like Otsu thresholding and even advanced models like Segment Anything Model (SAM) when used in isolation. Our results demonstrate that Zenesis is a powerful tool for scientific applications, particularly in fields where high-quality annotated datasets are unavailable, accelerating accurate analysis of experimental imaging.

Mamba-FSCIL: Dynamic Adaptation with Selective State Space Model for Few-Shot Class-Incremental Learning

Few-shot class-incremental learning (FSCIL) confronts the challenge of integrating new classes into a model with minimal training samples while preserving the knowledge of previously learned classes. Traditional methods widely adopt static adaptation relying on a fixed parameter space to learn from data that arrive sequentially, prone to overfitting to the current session. Existing dynamic strategies require the expansion of the parameter space continually, leading to increased complexity. To address these challenges, we integrate the recently proposed selective state space model (SSM) into FSCIL. Concretely, we propose a dual selective SSM projector that dynamically adjusts the projection parameters based on the intermediate features for dynamic adaptation. The dual design enables the model to maintain the robust features of base classes, while adaptively learning distinctive feature shifts for novel classes. Additionally, we develop a class-sensitive selective scan mechanism to guide dynamic adaptation. It minimizes the disruption to base-class representations caused by training on novel data, and meanwhile, forces the selective scan to perform in distinct patterns between base and novel classes. Experiments on miniImageNet, CUB-200, and CIFAR-100 demonstrate that our framework outperforms the existing state-of-the-art methods. The code is available at https://github.com/xiaojieli0903/Mamba-FSCIL.

Machine learning applications to DNA subsequence and restriction site analysis

Based on the BioBricks standard, restriction synthesis is a novel catabolic iterative DNA synthesis method that utilizes endonucleases to synthesize a query sequence from a reference sequence. In this work, the reference sequence is built from shorter subsequences by classifying them as applicable or inapplicable for the synthesis method using three different machine learning methods: Support Vector Machines (SVMs), random forest, and Convolution Neural Networks (CNNs). Before applying these methods to the data, a series of feature selection, curation, and reduction steps are applied to create an accurate and representative feature space. Following these preprocessing steps, three different pipelines are proposed to classify subsequences based on their nucleotide sequence and other relevant features corresponding to the restriction sites of over 200 endonucleases. The sensitivity using SVMs, random forest, and CNNs are 94.9%, 92.7%, 91.4%, respectively. Moreover, each method scores lower in specificity with SVMs, random forest, and CNNs resulting in 77.4%, 85.7%, and 82.4%, respectively. In addition to analyzing these results, the misclassifications in SVMs and CNNs are investigated. Across these two models, different features with a derived nucleotide specificity visually contribute more to classification compared to other features. This observation is an important factor when considering new nucleotide sensitivity features for future studies.

Mixed Effects Deep Learning for the interpretable analysis of single cell RNA sequencing data by quantifying and visualizing batch effects

Single-cell RNA sequencing (scRNA-seq) data are often confounded by technical or biological batch effects. Existing deep learning models mitigate these effects but often discard batch-specific information, potentially losing valuable biological insights. We propose a Mixed Effects Deep Learning (MEDL) autoencoder framework that separately models batch-invariant (fixed effects) and batch-specific (random effects) components. By decoupling batch-invariant biological states from batch variations, our framework integrates both into predictive models. Our approach also generates 2D visualizations of how the same cell appears across batches, enhancing interpretability. Retaining both fixed and random effect latent spaces improves classification accuracy. We applied our framework to three datasets spanning the cardiovascular system (Healthy Heart), Autism Spectrum Disorder (ASD), and Acute Myeloid Leukemia (AML). With 147 batches in the Healthy Heart dataset, far exceeding typical numbers, we tested our framework's ability to handle many batches. In the ASD dataset, our approach captured donor heterogeneity between autistic and healthy individuals. In the AML dataset, it distinguished donor heterogeneity despite missing cell types and diseased donors exhibiting both healthy and malignant cells. These results highlight our framework's ability to characterize fixed and random effects, enhance batch effect visualization, and improve prediction accuracy across diverse datasets.

MedSegFactory: Text-Guided Generation of Medical Image-Mask Pairs

This paper presents MedSegFactory, a versatile medical synthesis framework that generates high-quality paired medical images and segmentation masks across modalities and tasks. It aims to serve as an unlimited data repository, supplying image-mask pairs to enhance existing segmentation tools. The core of MedSegFactory is a dual-stream diffusion model, where one stream synthesizes medical images and the other generates corresponding segmentation masks. To ensure precise alignment between image-mask pairs, we introduce Joint Cross-Attention (JCA), enabling a collaborative denoising paradigm by dynamic cross-conditioning between streams. This bidirectional interaction allows both representations to guide each other's generation, enhancing consistency between generated pairs. MedSegFactory unlocks on-demand generation of paired medical images and segmentation masks through user-defined prompts that specify the target labels, imaging modalities, anatomical regions, and pathological conditions, facilitating scalable and high-quality data generation. This new paradigm of medical image synthesis enables seamless integration into diverse medical imaging workflows, enhancing both efficiency and accuracy. Extensive experiments show that MedSegFactory generates data of superior quality and usability, achieving competitive or state-of-the-art performance in 2D and 3D segmentation tasks while addressing data scarcity and regulatory constraints.

Sequence Parallelism: Long Sequence Training from System Perspective

Transformer achieves promising results on various tasks. However, self-attention suffers from quadratic memory requirements with respect to the sequence length. Existing work focuses on reducing time and space complexity from an algorithm perspective. In this work, we propose sequence parallelism, a memory-efficient parallelism method to help us break input sequence length limitation and train with longer sequences on GPUs efficiently. Our approach is compatible with most existing parallelisms (e.g. data parallelism, pipeline parallelism and tensor parallelism), which means our sequence parallelism makes 4D parallelism possible. More importantly, we no longer require a single device to hold the whole sequence. That is, with sparse attention, our sequence parallelism enables us to train transformer with infinite long sequence. Specifically, we split the input sequence into multiple chunks and feed each chunk into its corresponding device (i.e. GPU). To compute the attention output, we integrated ring-style communication with self-attention calculation and proposed Ring Self-Attention (RSA). Experiments show that sequence parallelism performs well when scaling with batch size and sequence length. Compared with tensor parallelism, our approach achieved 13.7times and 3.0times maximum batch size and sequence length respectively when scaling up to 64 NVIDIA P100 GPUs. With sparse attention, sequence can handle sequence with over 114K tokens, which is over 27times longer than existing sparse attention works holding the whole sequence on a single device.

SeqPE: Transformer with Sequential Position Encoding

Since self-attention layers in Transformers are permutation invariant by design, positional encodings must be explicitly incorporated to enable spatial understanding. However, fixed-size lookup tables used in traditional learnable position embeddings (PEs) limit extrapolation capabilities beyond pre-trained sequence lengths. Expert-designed methods such as ALiBi and RoPE, mitigate this limitation but demand extensive modifications for adapting to new modalities, underscoring fundamental challenges in adaptability and scalability. In this work, we present SeqPE, a unified and fully learnable position encoding framework that represents each n-dimensional position index as a symbolic sequence and employs a lightweight sequential position encoder to learn their embeddings in an end-to-end manner. To regularize SeqPE's embedding space, we introduce two complementary objectives: a contrastive objective that aligns embedding distances with a predefined position-distance function, and a knowledge distillation loss that anchors out-of-distribution position embeddings to in-distribution teacher representations, further enhancing extrapolation performance. Experiments across language modeling, long-context question answering, and 2D image classification demonstrate that SeqPE not only surpasses strong baselines in perplexity, exact match (EM), and accuracy--particularly under context length extrapolation--but also enables seamless generalization to multi-dimensional inputs without requiring manual architectural redesign. We release our code, data, and checkpoints at https://github.com/ghrua/seqpe.

SGUQ: Staged Graph Convolution Neural Network for Alzheimer's Disease Diagnosis using Multi-Omics Data

Alzheimer's disease (AD) is a chronic neurodegenerative disorder and the leading cause of dementia, significantly impacting cost, mortality, and burden worldwide. The advent of high-throughput omics technologies, such as genomics, transcriptomics, proteomics, and epigenomics, has revolutionized the molecular understanding of AD. Conventional AI approaches typically require the completion of all omics data at the outset to achieve optimal AD diagnosis, which are inefficient and may be unnecessary. To reduce the clinical cost and improve the accuracy of AD diagnosis using multi-omics data, we propose a novel staged graph convolutional network with uncertainty quantification (SGUQ). SGUQ begins with mRNA and progressively incorporates DNA methylation and miRNA data only when necessary, reducing overall costs and exposure to harmful tests. Experimental results indicate that 46.23% of the samples can be reliably predicted using only single-modal omics data (mRNA), while an additional 16.04% of the samples can achieve reliable predictions when combining two omics data types (mRNA + DNA methylation). In addition, the proposed staged SGUQ achieved an accuracy of 0.858 on ROSMAP dataset, which outperformed existing methods significantly. The proposed SGUQ can not only be applied to AD diagnosis using multi-omics data but also has the potential for clinical decision-making using multi-viewed data. Our implementation is publicly available at https://github.com/chenzhao2023/multiomicsuncertainty.

A Large-Scale Benchmark of Cross-Modal Learning for Histology and Gene Expression in Spatial Transcriptomics

Spatial transcriptomics enables simultaneous measurement of gene expression and tissue morphology, offering unprecedented insights into cellular organization and disease mechanisms. However, the field lacks comprehensive benchmarks for evaluating multimodal learning methods that leverage both histology images and gene expression data. Here, we present HESCAPE, a large-scale benchmark for cross-modal contrastive pretraining in spatial transcriptomics, built on a curated pan-organ dataset spanning 6 different gene panels and 54 donors. We systematically evaluated state-of-the-art image and gene expression encoders across multiple pretraining strategies and assessed their effectiveness on two downstream tasks: gene mutation classification and gene expression prediction. Our benchmark demonstrates that gene expression encoders are the primary determinant of strong representational alignment, and that gene models pretrained on spatial transcriptomics data outperform both those trained without spatial data and simple baseline approaches. However, downstream task evaluation reveals a striking contradiction: while contrastive pretraining consistently improves gene mutation classification performance, it degrades direct gene expression prediction compared to baseline encoders trained without cross-modal objectives. We identify batch effects as a key factor that interferes with effective cross-modal alignment. Our findings highlight the critical need for batch-robust multimodal learning approaches in spatial transcriptomics. To accelerate progress in this direction, we release HESCAPE, providing standardized datasets, evaluation protocols, and benchmarking tools for the community

Protenix-Mini: Efficient Structure Predictor via Compact Architecture, Few-Step Diffusion and Switchable pLM

Lightweight inference is critical for biomolecular structure prediction and other downstream tasks, enabling efficient real-world deployment and inference-time scaling for large-scale applications. In this work, we address the challenge of balancing model efficiency and prediction accuracy by making several key modifications, 1) Multi-step AF3 sampler is replaced by a few-step ODE sampler, significantly reducing computational overhead for the diffusion module part during inference; 2) In the open-source Protenix framework, a subset of pairformer or diffusion transformer blocks doesn't make contributions to the final structure prediction, presenting opportunities for architectural pruning and lightweight redesign; 3) A model incorporating an ESM module is trained to substitute the conventional MSA module, reducing MSA preprocessing time. Building on these key insights, we present Protenix-Mini, a compact and optimized model designed for efficient protein structure prediction. This streamlined version incorporates a more efficient architectural design with a two-step Ordinary Differential Equation (ODE) sampling strategy. By eliminating redundant Transformer components and refining the sampling process, Protenix-Mini significantly reduces model complexity with slight accuracy drop. Evaluations on benchmark datasets demonstrate that it achieves high-fidelity predictions, with only a negligible 1 to 5 percent decrease in performance on benchmark datasets compared to its full-scale counterpart. This makes Protenix-Mini an ideal choice for applications where computational resources are limited but accurate structure prediction remains crucial.

NCL-SM: A Fully Annotated Dataset of Images from Human Skeletal Muscle Biopsies

Single cell analysis of human skeletal muscle (SM) tissue cross-sections is a fundamental tool for understanding many neuromuscular disorders. For this analysis to be reliable and reproducible, identification of individual fibres within microscopy images (segmentation) of SM tissue should be automatic and precise. Biomedical scientists in this field currently rely on custom tools and general machine learning (ML) models, both followed by labour intensive and subjective manual interventions to fine-tune segmentation. We believe that fully automated, precise, reproducible segmentation is possible by training ML models. However, in this important biomedical domain, there are currently no good quality, publicly available annotated imaging datasets available for ML model training. In this paper we release NCL-SM: a high quality bioimaging dataset of 46 human SM tissue cross-sections from both healthy control subjects and from patients with genetically diagnosed muscle pathology. These images include > 50k manually segmented muscle fibres (myofibres). In addition we also curated high quality myofibre segmentations, annotating reasons for rejecting low quality myofibres and low quality regions in SM tissue images, making these annotations completely ready for downstream analysis. This, we believe, will pave the way for development of a fully automatic pipeline that identifies individual myofibres within images of tissue sections and, in particular, also classifies individual myofibres that are fit for further analysis.

MMGenBench: Fully Automatically Evaluating LMMs from the Text-to-Image Generation Perspective

Large Multimodal Models (LMMs) demonstrate impressive capabilities. However, current benchmarks predominantly focus on image comprehension in specific domains, and these benchmarks are labor-intensive to construct. Moreover, their answers tend to be brief, making it difficult to assess the ability of LMMs to generate detailed descriptions of images. To address these limitations, we propose the MMGenBench-Pipeline, a straightforward and fully automated evaluation pipeline. This involves generating textual descriptions from input images, using these descriptions to create auxiliary images via text-to-image generative models, and then comparing the original and generated images. Furthermore, to ensure the effectiveness of MMGenBench-Pipeline, we design MMGenBench-Test, evaluating LMMs across 13 distinct image patterns, and MMGenBench-Domain, focusing on generative image performance. A thorough evaluation involving over 50 popular LMMs demonstrates the effectiveness and reliability of both the pipeline and benchmark. Our observations indicate that numerous LMMs excelling in existing benchmarks fail to adequately complete the basic tasks related to image understanding and description. This finding highlights the substantial potential for performance improvement in current LMMs and suggests avenues for future model optimization. Concurrently, MMGenBench-Pipeline can efficiently assess the performance of LMMs across diverse domains using only image inputs.

Integrating Biological Knowledge for Robust Microscopy Image Profiling on De Novo Cell Lines

High-throughput screening techniques, such as microscopy imaging of cellular responses to genetic and chemical perturbations, play a crucial role in drug discovery and biomedical research. However, robust perturbation screening for de novo cell lines remains challenging due to the significant morphological and biological heterogeneity across cell lines. To address this, we propose a novel framework that integrates external biological knowledge into existing pretraining strategies to enhance microscopy image profiling models. Our approach explicitly disentangles perturbation-specific and cell line-specific representations using external biological information. Specifically, we construct a knowledge graph leveraging protein interaction data from STRING and Hetionet databases to guide models toward perturbation-specific features during pretraining. Additionally, we incorporate transcriptomic features from single-cell foundation models to capture cell line-specific representations. By learning these disentangled features, our method improves the generalization of imaging models to de novo cell lines. We evaluate our framework on the RxRx database through one-shot fine-tuning on an RxRx1 cell line and few-shot fine-tuning on cell lines from the RxRx19a dataset. Experimental results demonstrate that our method enhances microscopy image profiling for de novo cell lines, highlighting its effectiveness in real-world phenotype-based drug discovery applications.

SeNMo: A Self-Normalizing Deep Learning Model for Enhanced Multi-Omics Data Analysis in Oncology

Multi-omics research has enhanced our understanding of cancer heterogeneity and progression. Investigating molecular data through multi-omics approaches is crucial for unraveling the complex biological mechanisms underlying cancer, thereby enabling effective diagnosis, treatment, and prevention strategies. However, predicting patient outcomes through integration of all available multi-omics data is an under-study research direction. Here, we present SeNMo (Self-normalizing Network for Multi-omics), a deep neural network trained on multi-omics data across 33 cancer types. SeNMo is efficient in handling multi-omics data characterized by high-width (many features) and low-length (fewer samples) attributes. We trained SeNMo for the task of overall survival using pan-cancer data involving 33 cancer sites from Genomics Data Commons (GDC). The training data includes gene expression, DNA methylation, miRNA expression, DNA mutations, protein expression modalities, and clinical data. We evaluated the model's performance in predicting overall survival using concordance index (C-Index). SeNMo performed consistently well in training regime, with the validation C-Index of 0.76 on GDC's public data. In the testing regime, SeNMo performed with a C-Index of 0.758 on a held-out test set. The model showed an average accuracy of 99.8% on the task of classifying the primary cancer type on the pan-cancer test cohort. SeNMo proved to be a mini-foundation model for multi-omics oncology data because it demonstrated robust performance, and adaptability not only across molecular data types but also on the classification task of predicting the primary cancer type of patients. SeNMo can be further scaled to any cancer site and molecular data type. We believe SeNMo and similar models are poised to transform the oncology landscape, offering hope for more effective, efficient, and patient-centric cancer care.

PixCell: A generative foundation model for digital histopathology images

The digitization of histology slides has revolutionized pathology, providing massive datasets for cancer diagnosis and research. Contrastive self-supervised and vision-language models have been shown to effectively mine large pathology datasets to learn discriminative representations. On the other hand, generative models, capable of synthesizing realistic and diverse images, present a compelling solution to address unique problems in pathology that involve synthesizing images; overcoming annotated data scarcity, enabling privacy-preserving data sharing, and performing inherently generative tasks, such as virtual staining. We introduce PixCell, the first diffusion-based generative foundation model for histopathology. We train PixCell on PanCan-30M, a vast, diverse dataset derived from 69,184 H\&E-stained whole slide images covering various cancer types. We employ a progressive training strategy and a self-supervision-based conditioning that allows us to scale up training without any annotated data. PixCell generates diverse and high-quality images across multiple cancer types, which we find can be used in place of real data to train a self-supervised discriminative model. Synthetic images shared between institutions are subject to fewer regulatory barriers than would be the case with real clinical images. Furthermore, we showcase the ability to precisely control image generation using a small set of annotated images, which can be used for both data augmentation and educational purposes. Testing on a cell segmentation task, a mask-guided PixCell enables targeted data augmentation, improving downstream performance. Finally, we demonstrate PixCell's ability to use H\&E structural staining to infer results from molecular marker studies; we use this capability to infer IHC staining from H\&E images. Our trained models are publicly released to accelerate research in computational pathology.

ChAda-ViT : Channel Adaptive Attention for Joint Representation Learning of Heterogeneous Microscopy Images

Unlike color photography images, which are consistently encoded into RGB channels, biological images encompass various modalities, where the type of microscopy and the meaning of each channel varies with each experiment. Importantly, the number of channels can range from one to a dozen and their correlation is often comparatively much lower than RGB, as each of them brings specific information content. This aspect is largely overlooked by methods designed out of the bioimage field, and current solutions mostly focus on intra-channel spatial attention, often ignoring the relationship between channels, yet crucial in most biological applications. Importantly, the variable channel type and count prevent the projection of several experiments to a unified representation for large scale pre-training. In this study, we propose ChAda-ViT, a novel Channel Adaptive Vision Transformer architecture employing an Inter-Channel Attention mechanism on images with an arbitrary number, order and type of channels. We also introduce IDRCell100k, a bioimage dataset with a rich set of 79 experiments covering 7 microscope modalities, with a multitude of channel types, and channel counts varying from 1 to 10 per experiment. Our proposed architecture, trained in a self-supervised manner, outperforms existing approaches in several biologically relevant downstream tasks. Additionally, it can be used to bridge the gap for the first time between assays with different microscopes, channel numbers or types by embedding various image and experimental modalities into a unified biological image representation. The latter should facilitate interdisciplinary studies and pave the way for better adoption of deep learning in biological image-based analyses. Code and Data to be released soon.

Fine-Tuning and Training of DenseNet for Histopathology Image Representation Using TCGA Diagnostic Slides

Feature vectors provided by pre-trained deep artificial neural networks have become a dominant source for image representation in recent literature. Their contribution to the performance of image analysis can be improved through finetuning. As an ultimate solution, one might even train a deep network from scratch with the domain-relevant images, a highly desirable option which is generally impeded in pathology by lack of labeled images and the computational expense. In this study, we propose a new network, namely KimiaNet, that employs the topology of the DenseNet with four dense blocks, fine-tuned and trained with histopathology images in different configurations. We used more than 240,000 image patches with 1000x1000 pixels acquired at 20x magnification through our proposed "highcellularity mosaic" approach to enable the usage of weak labels of 7,126 whole slide images of formalin-fixed paraffin-embedded human pathology samples publicly available through the The Cancer Genome Atlas (TCGA) repository. We tested KimiaNet using three public datasets, namely TCGA, endometrial cancer images, and colorectal cancer images by evaluating the performance of search and classification when corresponding features of different networks are used for image representation. As well, we designed and trained multiple convolutional batch-normalized ReLU (CBR) networks. The results show that KimiaNet provides superior results compared to the original DenseNet and smaller CBR networks when used as feature extractor to represent histopathology images.

DNABERT-2: Efficient Foundation Model and Benchmark For Multi-Species Genome

Decoding the linguistic intricacies of the genome is a crucial problem in biology, and pre-trained foundational models such as DNABERT and Nucleotide Transformer have made significant strides in this area. Existing works have largely hinged on k-mer, fixed-length permutations of A, T, C, and G, as the token of the genome language due to its simplicity. However, we argue that the computation and sample inefficiencies introduced by k-mer tokenization are primary obstacles in developing large genome foundational models. We provide conceptual and empirical insights into genome tokenization, building on which we propose to replace k-mer tokenization with Byte Pair Encoding (BPE), a statistics-based data compression algorithm that constructs tokens by iteratively merging the most frequent co-occurring genome segment in the corpus. We demonstrate that BPE not only overcomes the limitations of k-mer tokenization but also benefits from the computational efficiency of non-overlapping tokenization. Based on these insights, we introduce DNABERT-2, a refined genome foundation model that adapts an efficient tokenizer and employs multiple strategies to overcome input length constraints, reduce time and memory expenditure, and enhance model capability. Furthermore, we identify the absence of a comprehensive and standardized benchmark for genome understanding as another significant impediment to fair comparative analysis. In response, we propose the Genome Understanding Evaluation (GUE), a comprehensive multi-species genome classification dataset that amalgamates 28 distinct datasets across 7 tasks, with input lengths ranging from 70 to 1000. Through comprehensive experiments on the GUE benchmark, we demonstrate that DNABERT-2 achieves comparable performance to the state-of-the-art model with 21 times fewer parameters and approximately 56 times less GPU time in pre-training.

A Multimodal Knowledge-enhanced Whole-slide Pathology Foundation Model

Remarkable strides in computational pathology have been made in the task-agnostic foundation model that advances the performance of a wide array of downstream clinical tasks. Despite the promising performance, there are still several challenges. First, prior works have resorted to either vision-only or image-caption data, disregarding pathology reports with more clinically authentic information from pathologists and gene expression profiles which respectively offer distinct knowledge for versatile clinical applications. Second, the current progress in pathology FMs predominantly concentrates on the patch level, where the restricted context of patch-level pretraining fails to capture whole-slide patterns. Even recent slide-level FMs still struggle to provide whole-slide context for patch representation. In this study, for the first time, we develop a pathology foundation model incorporating three levels of modalities: pathology slides, pathology reports, and gene expression data, which resulted in 26,169 slide-level modality pairs from 10,275 patients across 32 cancer types, amounting to over 116 million pathological patch images. To leverage these data for CPath, we propose a novel whole-slide pretraining paradigm that injects the multimodal whole-slide context into the patch representation, called Multimodal Self-TAught PRetraining (mSTAR). The proposed paradigm revolutionizes the pretraining workflow for CPath, enabling the pathology FM to acquire the whole-slide context. To the best of our knowledge, this is the first attempt to incorporate three modalities at the whole-slide context for enhancing pathology FMs. To systematically evaluate the capabilities of mSTAR, we built the largest spectrum of oncological benchmark, spanning 7 categories of oncological applications in 15 types of 97 practical oncological tasks.

Meissonic: Revitalizing Masked Generative Transformers for Efficient High-Resolution Text-to-Image Synthesis

Diffusion models, such as Stable Diffusion, have made significant strides in visual generation, yet their paradigm remains fundamentally different from autoregressive language models, complicating the development of unified language-vision models. Recent efforts like LlamaGen have attempted autoregressive image generation using discrete VQVAE tokens, but the large number of tokens involved renders this approach inefficient and slow. In this work, we present Meissonic, which elevates non-autoregressive masked image modeling (MIM) text-to-image to a level comparable with state-of-the-art diffusion models like SDXL. By incorporating a comprehensive suite of architectural innovations, advanced positional encoding strategies, and optimized sampling conditions, Meissonic substantially improves MIM's performance and efficiency. Additionally, we leverage high-quality training data, integrate micro-conditions informed by human preference scores, and employ feature compression layers to further enhance image fidelity and resolution. Our model not only matches but often exceeds the performance of existing models like SDXL in generating high-quality, high-resolution images. Extensive experiments validate Meissonic's capabilities, demonstrating its potential as a new standard in text-to-image synthesis. We release a model checkpoint capable of producing 1024 times 1024 resolution images.

A versatile informative diffusion model for single-cell ATAC-seq data generation and analysis

The rapid advancement of single-cell ATAC sequencing (scATAC-seq) technologies holds great promise for investigating the heterogeneity of epigenetic landscapes at the cellular level. The amplification process in scATAC-seq experiments often introduces noise due to dropout events, which results in extreme sparsity that hinders accurate analysis. Consequently, there is a significant demand for the generation of high-quality scATAC-seq data in silico. Furthermore, current methodologies are typically task-specific, lacking a versatile framework capable of handling multiple tasks within a single model. In this work, we propose ATAC-Diff, a versatile framework, which is based on a latent diffusion model conditioned on the latent auxiliary variables to adapt for various tasks. ATAC-Diff is the first diffusion model for the scATAC-seq data generation and analysis, composed of auxiliary modules encoding the latent high-level variables to enable the model to learn the semantic information to sample high-quality data. Gaussian Mixture Model (GMM) as the latent prior and auxiliary decoder, the yield variables reserve the refined genomic information beneficial for downstream analyses. Another innovation is the incorporation of mutual information between observed and hidden variables as a regularization term to prevent the model from decoupling from latent variables. Through extensive experiments, we demonstrate that ATAC-Diff achieves high performance in both generation and analysis tasks, outperforming state-of-the-art models.

Instella-T2I: Pushing the Limits of 1D Discrete Latent Space Image Generation

Image tokenization plays a critical role in reducing the computational demands of modeling high-resolution images, significantly improving the efficiency of image and multimodal understanding and generation. Recent advances in 1D latent spaces have reduced the number of tokens required by eliminating the need for a 2D grid structure. In this paper, we further advance compact discrete image representation by introducing 1D binary image latents. By representing each image as a sequence of binary vectors, rather than using traditional one-hot codebook tokens, our approach preserves high-resolution details while maintaining the compactness of 1D latents. To the best of our knowledge, our text-to-image models are the first to achieve competitive performance in both diffusion and auto-regressive generation using just 128 discrete tokens for images up to 1024x1024, demonstrating up to a 32-fold reduction in token numbers compared to standard VQ-VAEs. The proposed 1D binary latent space, coupled with simple model architectures, achieves marked improvements in speed training and inference speed. Our text-to-image models allow for a global batch size of 4096 on a single GPU node with 8 AMD MI300X GPUs, and the training can be completed within 200 GPU days. Our models achieve competitive performance compared to modern image generation models without any in-house private training data or post-training refinements, offering a scalable and efficient alternative to conventional tokenization methods.

PixArt-Σ: Weak-to-Strong Training of Diffusion Transformer for 4K Text-to-Image Generation

In this paper, we introduce PixArt-\Sigma, a Diffusion Transformer model~(DiT) capable of directly generating images at 4K resolution. PixArt-\Sigma represents a significant advancement over its predecessor, PixArt-\alpha, offering images of markedly higher fidelity and improved alignment with text prompts. A key feature of PixArt-\Sigma is its training efficiency. Leveraging the foundational pre-training of PixArt-\alpha, it evolves from the `weaker' baseline to a `stronger' model via incorporating higher quality data, a process we term "weak-to-strong training". The advancements in PixArt-\Sigma are twofold: (1) High-Quality Training Data: PixArt-\Sigma incorporates superior-quality image data, paired with more precise and detailed image captions. (2) Efficient Token Compression: we propose a novel attention module within the DiT framework that compresses both keys and values, significantly improving efficiency and facilitating ultra-high-resolution image generation. Thanks to these improvements, PixArt-\Sigma achieves superior image quality and user prompt adherence capabilities with significantly smaller model size (0.6B parameters) than existing text-to-image diffusion models, such as SDXL (2.6B parameters) and SD Cascade (5.1B parameters). Moreover, PixArt-\Sigma's capability to generate 4K images supports the creation of high-resolution posters and wallpapers, efficiently bolstering the production of high-quality visual content in industries such as film and gaming.

DNA Sequence Classification with Compressors

Recent studies in DNA sequence classification have leveraged sophisticated machine learning techniques, achieving notable accuracy in categorizing complex genomic data. Among these, methods such as k-mer counting have proven effective in distinguishing sequences from varied species like chimpanzees, dogs, and humans, becoming a staple in contemporary genomic research. However, these approaches often demand extensive computational resources, posing a challenge in terms of scalability and efficiency. Addressing this issue, our study introduces a novel adaptation of Jiang et al.'s compressor-based, parameter-free classification method, specifically tailored for DNA sequence analysis. This innovative approach utilizes a variety of compression algorithms, such as Gzip, Brotli, and LZMA, to efficiently process and classify genomic sequences. Not only does this method align with the current state-of-the-art in terms of accuracy, but it also offers a more resource-efficient alternative to traditional machine learning methods. Our comprehensive evaluation demonstrates the proposed method's effectiveness in accurately classifying DNA sequences from multiple species. We present a detailed analysis of the performance of each algorithm used, highlighting the strengths and limitations of our approach in various genomic contexts. Furthermore, we discuss the broader implications of our findings for bioinformatics, particularly in genomic data processing and analysis. The results of our study pave the way for more efficient and scalable DNA sequence classification methods, offering significant potential for advancements in genomic research and applications.

PathGen-1.6M: 1.6 Million Pathology Image-text Pairs Generation through Multi-agent Collaboration

Vision Language Models (VLMs) like CLIP have attracted substantial attention in pathology, serving as backbones for applications such as zero-shot image classification and Whole Slide Image (WSI) analysis. Additionally, they can function as vision encoders when combined with large language models (LLMs) to support broader capabilities. Current efforts to train pathology VLMs rely on pathology image-text pairs from platforms like PubMed, YouTube, and Twitter, which provide limited, unscalable data with generally suboptimal image quality. In this work, we leverage large-scale WSI datasets like TCGA to extract numerous high-quality image patches. We then train a large multimodal model to generate captions for these images, creating PathGen-1.6M, a dataset containing 1.6 million high-quality image-caption pairs. Our approach involves multiple agent models collaborating to extract representative WSI patches, generating and refining captions to obtain high-quality image-text pairs. Extensive experiments show that integrating these generated pairs with existing datasets to train a pathology-specific CLIP model, PathGen-CLIP, significantly enhances its ability to analyze pathological images, with substantial improvements across nine pathology-related zero-shot image classification tasks and three whole-slide image tasks. Furthermore, we construct 200K instruction-tuning data based on PathGen-1.6M and integrate PathGen-CLIP with the Vicuna LLM to create more powerful multimodal models through instruction tuning. Overall, we provide a scalable pathway for high-quality data generation in pathology, paving the way for next-generation general pathology models.

TEDDY: A Family Of Foundation Models For Understanding Single Cell Biology

Understanding the biological mechanism of disease is critical for medicine, and in particular drug discovery. AI-powered analysis of genome-scale biological data hold great potential in this regard. The increasing availability of single-cell RNA sequencing data has enabled the development of large foundation models for disease biology. However, existing foundation models either do not improve or only modestly improve over task-specific models in downstream applications. Here, we explored two avenues for improving the state-of-the-art. First, we scaled the pre-training dataset to 116 million cells, which is larger than those used by previous models. Second, we leveraged the availability of large-scale biological annotations as a form of supervision during pre-training. We trained the TEDDY family of models comprising six transformer-based state-of-the-art single-cell foundation models with 70 million, 160 million, and 400 million parameters. We vetted our models on two downstream evaluation tasks -- identifying the underlying disease state of held-out donors not seen during training and distinguishing healthy cells from diseased ones for disease conditions and donors not seen during training. Scaling experiments showed that performance improved predictably with both data volume and parameter count. Our models showed substantial improvement over existing work on the first task and more muted improvements on the second.

ContriMix: Unsupervised disentanglement of content and attribute for domain generalization in microscopy image analysis

Domain generalization is critical for real-world applications of machine learning to microscopy images, including histopathology and fluorescence imaging. Artifacts in these modalities arise through a complex combination of factors relating to tissue collection and laboratory processing, as well as factors intrinsic to patient samples. In fluorescence imaging, these artifacts stem from variations across experimental batches. The complexity and subtlety of these artifacts make the enumeration of data domains intractable. Therefore, augmentation-based methods of domain generalization that require domain identifiers and manual fine-tuning are inadequate in this setting. To overcome this challenge, we introduce ContriMix, a domain generalization technique that learns to generate synthetic images by disentangling and permuting the biological content ("content") and technical variations ("attributes") in microscopy images. ContriMix does not rely on domain identifiers or handcrafted augmentations and makes no assumptions about the input characteristics of images. We assess the performance of ContriMix on two pathology datasets dealing with patch classification and Whole Slide Image label prediction tasks respectively (Camelyon17-WILDS and RCC subtyping), and one fluorescence microscopy dataset (RxRx1-WILDS). Without any access to domain identifiers at train or test time, ContriMix performs similar or better than current state-of-the-art methods in all these datasets, motivating its usage for microscopy image analysis in real-world settings where domain information is hard to come by. The code for ContriMix can be found at https://gitlab.com/huutan86/contrimix

The KiTS21 Challenge: Automatic segmentation of kidneys, renal tumors, and renal cysts in corticomedullary-phase CT

This paper presents the challenge report for the 2021 Kidney and Kidney Tumor Segmentation Challenge (KiTS21) held in conjunction with the 2021 international conference on Medical Image Computing and Computer Assisted Interventions (MICCAI). KiTS21 is a sequel to its first edition in 2019, and it features a variety of innovations in how the challenge was designed, in addition to a larger dataset. A novel annotation method was used to collect three separate annotations for each region of interest, and these annotations were performed in a fully transparent setting using a web-based annotation tool. Further, the KiTS21 test set was collected from an outside institution, challenging participants to develop methods that generalize well to new populations. Nonetheless, the top-performing teams achieved a significant improvement over the state of the art set in 2019, and this performance is shown to inch ever closer to human-level performance. An in-depth meta-analysis is presented describing which methods were used and how they faired on the leaderboard, as well as the characteristics of which cases generally saw good performance, and which did not. Overall KiTS21 facilitated a significant advancement in the state of the art in kidney tumor segmentation, and provides useful insights that are applicable to the field of semantic segmentation as a whole.

PhotoVerse: Tuning-Free Image Customization with Text-to-Image Diffusion Models

Personalized text-to-image generation has emerged as a powerful and sought-after tool, empowering users to create customized images based on their specific concepts and prompts. However, existing approaches to personalization encounter multiple challenges, including long tuning times, large storage requirements, the necessity for multiple input images per identity, and limitations in preserving identity and editability. To address these obstacles, we present PhotoVerse, an innovative methodology that incorporates a dual-branch conditioning mechanism in both text and image domains, providing effective control over the image generation process. Furthermore, we introduce facial identity loss as a novel component to enhance the preservation of identity during training. Remarkably, our proposed PhotoVerse eliminates the need for test time tuning and relies solely on a single facial photo of the target identity, significantly reducing the resource cost associated with image generation. After a single training phase, our approach enables generating high-quality images within only a few seconds. Moreover, our method can produce diverse images that encompass various scenes and styles. The extensive evaluation demonstrates the superior performance of our approach, which achieves the dual objectives of preserving identity and facilitating editability. Project page: https://photoverse2d.github.io/

Scaling Laws in Patchification: An Image Is Worth 50,176 Tokens And More

Since the introduction of Vision Transformer (ViT), patchification has long been regarded as a de facto image tokenization approach for plain visual architectures. By compressing the spatial size of images, this approach can effectively shorten the token sequence and reduce the computational cost of ViT-like plain architectures. In this work, we aim to thoroughly examine the information loss caused by this patchification-based compressive encoding paradigm and how it affects visual understanding. We conduct extensive patch size scaling experiments and excitedly observe an intriguing scaling law in patchification: the models can consistently benefit from decreased patch sizes and attain improved predictive performance, until it reaches the minimum patch size of 1x1, i.e., pixel tokenization. This conclusion is broadly applicable across different vision tasks, various input scales, and diverse architectures such as ViT and the recent Mamba models. Moreover, as a by-product, we discover that with smaller patches, task-specific decoder heads become less critical for dense prediction. In the experiments, we successfully scale up the visual sequence to an exceptional length of 50,176 tokens, achieving a competitive test accuracy of 84.6% with a base-sized model on the ImageNet-1k benchmark. We hope this study can provide insights and theoretical foundations for future works of building non-compressive vision models. Code is available at https://github.com/wangf3014/Patch_Scaling.

An Image is Worth 32 Tokens for Reconstruction and Generation

Recent advancements in generative models have highlighted the crucial role of image tokenization in the efficient synthesis of high-resolution images. Tokenization, which transforms images into latent representations, reduces computational demands compared to directly processing pixels and enhances the effectiveness and efficiency of the generation process. Prior methods, such as VQGAN, typically utilize 2D latent grids with fixed downsampling factors. However, these 2D tokenizations face challenges in managing the inherent redundancies present in images, where adjacent regions frequently display similarities. To overcome this issue, we introduce Transformer-based 1-Dimensional Tokenizer (TiTok), an innovative approach that tokenizes images into 1D latent sequences. TiTok provides a more compact latent representation, yielding substantially more efficient and effective representations than conventional techniques. For example, a 256 x 256 x 3 image can be reduced to just 32 discrete tokens, a significant reduction from the 256 or 1024 tokens obtained by prior methods. Despite its compact nature, TiTok achieves competitive performance to state-of-the-art approaches. Specifically, using the same generator framework, TiTok attains 1.97 gFID, outperforming MaskGIT baseline significantly by 4.21 at ImageNet 256 x 256 benchmark. The advantages of TiTok become even more significant when it comes to higher resolution. At ImageNet 512 x 512 benchmark, TiTok not only outperforms state-of-the-art diffusion model DiT-XL/2 (gFID 2.74 vs. 3.04), but also reduces the image tokens by 64x, leading to 410x faster generation process. Our best-performing variant can significantly surpasses DiT-XL/2 (gFID 2.13 vs. 3.04) while still generating high-quality samples 74x faster.

Wake Vision: A Large-scale, Diverse Dataset and Benchmark Suite for TinyML Person Detection

Machine learning applications on extremely low-power devices, commonly referred to as tiny machine learning (TinyML), promises a smarter and more connected world. However, the advancement of current TinyML research is hindered by the limited size and quality of pertinent datasets. To address this challenge, we introduce Wake Vision, a large-scale, diverse dataset tailored for person detection -- the canonical task for TinyML visual sensing. Wake Vision comprises over 6 million images, which is a hundredfold increase compared to the previous standard, and has undergone thorough quality filtering. Using Wake Vision for training results in a 2.41\% increase in accuracy compared to the established benchmark. Alongside the dataset, we provide a collection of five detailed benchmark sets that assess model performance on specific segments of the test data, such as varying lighting conditions, distances from the camera, and demographic characteristics of subjects. These novel fine-grained benchmarks facilitate the evaluation of model quality in challenging real-world scenarios that are often ignored when focusing solely on overall accuracy. Through an evaluation of a MobileNetV2 TinyML model on the benchmarks, we show that the input resolution plays a more crucial role than the model width in detecting distant subjects and that the impact of quantization on model robustness is minimal, thanks to the dataset quality. These findings underscore the importance of a detailed evaluation to identify essential factors for model development. The dataset, benchmark suite, code, and models are publicly available under the CC-BY 4.0 license, enabling their use for commercial use cases.

MetaShift: A Dataset of Datasets for Evaluating Contextual Distribution Shifts and Training Conflicts

Understanding the performance of machine learning models across diverse data distributions is critically important for reliable applications. Motivated by this, there is a growing focus on curating benchmark datasets that capture distribution shifts. While valuable, the existing benchmarks are limited in that many of them only contain a small number of shifts and they lack systematic annotation about what is different across different shifts. We present MetaShift--a collection of 12,868 sets of natural images across 410 classes--to address this challenge. We leverage the natural heterogeneity of Visual Genome and its annotations to construct MetaShift. The key construction idea is to cluster images using its metadata, which provides context for each image (e.g. "cats with cars" or "cats in bathroom") that represent distinct data distributions. MetaShift has two important benefits: first, it contains orders of magnitude more natural data shifts than previously available. Second, it provides explicit explanations of what is unique about each of its data sets and a distance score that measures the amount of distribution shift between any two of its data sets. We demonstrate the utility of MetaShift in benchmarking several recent proposals for training models to be robust to data shifts. We find that the simple empirical risk minimization performs the best when shifts are moderate and no method had a systematic advantage for large shifts. We also show how MetaShift can help to visualize conflicts between data subsets during model training.

Generative AI for Medical Imaging: extending the MONAI Framework

Recent advances in generative AI have brought incredible breakthroughs in several areas, including medical imaging. These generative models have tremendous potential not only to help safely share medical data via synthetic datasets but also to perform an array of diverse applications, such as anomaly detection, image-to-image translation, denoising, and MRI reconstruction. However, due to the complexity of these models, their implementation and reproducibility can be difficult. This complexity can hinder progress, act as a use barrier, and dissuade the comparison of new methods with existing works. In this study, we present MONAI Generative Models, a freely available open-source platform that allows researchers and developers to easily train, evaluate, and deploy generative models and related applications. Our platform reproduces state-of-art studies in a standardised way involving different architectures (such as diffusion models, autoregressive transformers, and GANs), and provides pre-trained models for the community. We have implemented these models in a generalisable fashion, illustrating that their results can be extended to 2D or 3D scenarios, including medical images with different modalities (like CT, MRI, and X-Ray data) and from different anatomical areas. Finally, we adopt a modular and extensible approach, ensuring long-term maintainability and the extension of current applications for future features.

Gen4Gen: Generative Data Pipeline for Generative Multi-Concept Composition

Recent text-to-image diffusion models are able to learn and synthesize images containing novel, personalized concepts (e.g., their own pets or specific items) with just a few examples for training. This paper tackles two interconnected issues within this realm of personalizing text-to-image diffusion models. First, current personalization techniques fail to reliably extend to multiple concepts -- we hypothesize this to be due to the mismatch between complex scenes and simple text descriptions in the pre-training dataset (e.g., LAION). Second, given an image containing multiple personalized concepts, there lacks a holistic metric that evaluates performance on not just the degree of resemblance of personalized concepts, but also whether all concepts are present in the image and whether the image accurately reflects the overall text description. To address these issues, we introduce Gen4Gen, a semi-automated dataset creation pipeline utilizing generative models to combine personalized concepts into complex compositions along with text-descriptions. Using this, we create a dataset called MyCanvas, that can be used to benchmark the task of multi-concept personalization. In addition, we design a comprehensive metric comprising two scores (CP-CLIP and TI-CLIP) for better quantifying the performance of multi-concept, personalized text-to-image diffusion methods. We provide a simple baseline built on top of Custom Diffusion with empirical prompting strategies for future researchers to evaluate on MyCanvas. We show that by improving data quality and prompting strategies, we can significantly increase multi-concept personalized image generation quality, without requiring any modifications to model architecture or training algorithms.

Transformer as Linear Expansion of Learngene

We propose expanding the shared Transformer module to produce and initialize Transformers of varying depths, enabling adaptation to diverse resource constraints. Drawing an analogy to genetic expansibility, we term such module as learngene. To identify the expansion mechanism, we delve into the relationship between the layer's position and its corresponding weight value, and find that linear function appropriately approximates this relationship. Building on this insight, we present Transformer as Linear Expansion of learnGene (TLEG), a novel approach for flexibly producing and initializing Transformers of diverse depths. Specifically, to learn learngene, we firstly construct an auxiliary Transformer linearly expanded from learngene, after which we train it through employing soft distillation. Subsequently, we can produce and initialize Transformers of varying depths via linearly expanding the well-trained learngene, thereby supporting diverse downstream scenarios. Extensive experiments on ImageNet-1K demonstrate that TLEG achieves comparable or better performance in contrast to many individual models trained from scratch, while reducing around 2x training cost. When transferring to several downstream classification datasets, TLEG surpasses existing initialization methods by a large margin (e.g., +6.87% on iNat 2019 and +7.66% on CIFAR-100). Under the situation where we need to produce models of varying depths adapting for different resource constraints, TLEG achieves comparable results while reducing around 19x parameters stored to initialize these models and around 5x pre-training costs, in contrast to the pre-training and fine-tuning approach. When transferring a fixed set of parameters to initialize different models, TLEG presents better flexibility and competitive performance while reducing around 2.9x parameters stored to initialize, compared to the pre-training approach.

ILLUME+: Illuminating Unified MLLM with Dual Visual Tokenization and Diffusion Refinement

We present ILLUME+ that leverages dual visual tokenization and a diffusion decoder to improve both deep semantic understanding and high-fidelity image generation. Existing unified models have struggled to simultaneously handle the three fundamental capabilities in a unified model: understanding, generation, and editing. Models like Chameleon and EMU3 utilize VQGAN for image discretization, due to the lack of deep semantic interaction, they lag behind specialist models like LLaVA in visual understanding tasks. To mitigate this, LaViT and ILLUME employ semantic encoders for tokenization, but they struggle with image editing due to poor texture preservation. Meanwhile, Janus series decouples the input and output image representation, limiting their abilities to seamlessly handle interleaved image-text understanding and generation. In contrast, ILLUME+ introduces a unified dual visual tokenizer, DualViTok, which preserves both fine-grained textures and text-aligned semantics while enabling a coarse-to-fine image representation strategy for multimodal understanding and generation. Additionally, we employ a diffusion model as the image detokenizer for enhanced generation quality and efficient super-resolution. ILLUME+ follows a continuous-input, discrete-output scheme within the unified MLLM and adopts a progressive training procedure that supports dynamic resolution across the vision tokenizer, MLLM, and diffusion decoder. This design allows for flexible and efficient context-aware image editing and generation across diverse tasks. ILLUME+ (3B) exhibits competitive performance against existing unified MLLMs and specialized models across multimodal understanding, generation, and editing benchmarks. With its strong performance, ILLUME+ provides a scalable and versatile foundation for future multimodal applications. Project Page: https://illume-unified-mllm.github.io/.

Multistain Pretraining for Slide Representation Learning in Pathology

Developing self-supervised learning (SSL) models that can learn universal and transferable representations of H&E gigapixel whole-slide images (WSIs) is becoming increasingly valuable in computational pathology. These models hold the potential to advance critical tasks such as few-shot classification, slide retrieval, and patient stratification. Existing approaches for slide representation learning extend the principles of SSL from small images (e.g., 224 x 224 patches) to entire slides, usually by aligning two different augmentations (or views) of the slide. Yet the resulting representation remains constrained by the limited clinical and biological diversity of the views. Instead, we postulate that slides stained with multiple markers, such as immunohistochemistry, can be used as different views to form a rich task-agnostic training signal. To this end, we introduce Madeleine, a multimodal pretraining strategy for slide representation learning. Madeleine is trained with a dual global-local cross-stain alignment objective on large cohorts of breast cancer samples (N=4,211 WSIs across five stains) and kidney transplant samples (N=12,070 WSIs across four stains). We demonstrate the quality of slide representations learned by Madeleine on various downstream evaluations, ranging from morphological and molecular classification to prognostic prediction, comprising 21 tasks using 7,299 WSIs from multiple medical centers. Code is available at https://github.com/mahmoodlab/MADELEINE.

Deep SNP: An End-to-end Deep Neural Network with Attention-based Localization for Break-point Detection in SNP Array Genomic data

Diagnosis and risk stratification of cancer and many other diseases require the detection of genomic breakpoints as a prerequisite of calling copy number alterations (CNA). This, however, is still challenging and requires time-consuming manual curation. As deep-learning methods outperformed classical state-of-the-art algorithms in various domains and have also been successfully applied to life science problems including medicine and biology, we here propose Deep SNP, a novel Deep Neural Network to learn from genomic data. Specifically, we used a manually curated dataset from 12 genomic single nucleotide polymorphism array (SNPa) profiles as truth-set and aimed at predicting the presence or absence of genomic breakpoints, an indicator of structural chromosomal variations, in windows of 40,000 probes. We compare our results with well-known neural network models as well as Rawcopy though this tool is designed to predict breakpoints and in addition genomic segments with high sensitivity. We show, that Deep SNP is capable of successfully predicting the presence or absence of a breakpoint in large genomic windows and outperforms state-of-the-art neural network models. Qualitative examples suggest that integration of a localization unit may enable breakpoint detection and prediction of genomic segments, even if the breakpoint coordinates were not provided for network training. These results warrant further evaluation of DeepSNP for breakpoint localization and subsequent calling of genomic segments.

MPFNet: A Multi-Prior Fusion Network with a Progressive Training Strategy for Micro-Expression Recognition

Micro-expression recognition (MER), a critical subfield of affective computing, presents greater challenges than macro-expression recognition due to its brief duration and low intensity. While incorporating prior knowledge has been shown to enhance MER performance, existing methods predominantly rely on simplistic, singular sources of prior knowledge, failing to fully exploit multi-source information. This paper introduces the Multi-Prior Fusion Network (MPFNet), leveraging a progressive training strategy to optimize MER tasks. We propose two complementary encoders: the Generic Feature Encoder (GFE) and the Advanced Feature Encoder (AFE), both based on Inflated 3D ConvNets (I3D) with Coordinate Attention (CA) mechanisms, to improve the model's ability to capture spatiotemporal and channel-specific features. Inspired by developmental psychology, we present two variants of MPFNet--MPFNet-P and MPFNet-C--corresponding to two fundamental modes of infant cognitive development: parallel and hierarchical processing. These variants enable the evaluation of different strategies for integrating prior knowledge. Extensive experiments demonstrate that MPFNet significantly improves MER accuracy while maintaining balanced performance across categories, achieving accuracies of 0.811, 0.924, and 0.857 on the SMIC, CASME II, and SAMM datasets, respectively. To the best of our knowledge, our approach achieves state-of-the-art performance on the SMIC and SAMM datasets.

Cross-Modal Translation and Alignment for Survival Analysis

With the rapid advances in high-throughput sequencing technologies, the focus of survival analysis has shifted from examining clinical indicators to incorporating genomic profiles with pathological images. However, existing methods either directly adopt a straightforward fusion of pathological features and genomic profiles for survival prediction, or take genomic profiles as guidance to integrate the features of pathological images. The former would overlook intrinsic cross-modal correlations. The latter would discard pathological information irrelevant to gene expression. To address these issues, we present a Cross-Modal Translation and Alignment (CMTA) framework to explore the intrinsic cross-modal correlations and transfer potential complementary information. Specifically, we construct two parallel encoder-decoder structures for multi-modal data to integrate intra-modal information and generate cross-modal representation. Taking the generated cross-modal representation to enhance and recalibrate intra-modal representation can significantly improve its discrimination for comprehensive survival analysis. To explore the intrinsic crossmodal correlations, we further design a cross-modal attention module as the information bridge between different modalities to perform cross-modal interactions and transfer complementary information. Our extensive experiments on five public TCGA datasets demonstrate that our proposed framework outperforms the state-of-the-art methods.

μ-Bench: A Vision-Language Benchmark for Microscopy Understanding

Recent advances in microscopy have enabled the rapid generation of terabytes of image data in cell biology and biomedical research. Vision-language models (VLMs) offer a promising solution for large-scale biological image analysis, enhancing researchers' efficiency, identifying new image biomarkers, and accelerating hypothesis generation and scientific discovery. However, there is a lack of standardized, diverse, and large-scale vision-language benchmarks to evaluate VLMs' perception and cognition capabilities in biological image understanding. To address this gap, we introduce {\mu}-Bench, an expert-curated benchmark encompassing 22 biomedical tasks across various scientific disciplines (biology, pathology), microscopy modalities (electron, fluorescence, light), scales (subcellular, cellular, tissue), and organisms in both normal and abnormal states. We evaluate state-of-the-art biomedical, pathology, and general VLMs on {\mu}-Bench and find that: i) current models struggle on all categories, even for basic tasks such as distinguishing microscopy modalities; ii) current specialist models fine-tuned on biomedical data often perform worse than generalist models; iii) fine-tuning in specific microscopy domains can cause catastrophic forgetting, eroding prior biomedical knowledge encoded in their base model. iv) weight interpolation between fine-tuned and pre-trained models offers one solution to forgetting and improves general performance across biomedical tasks. We release {\mu}-Bench under a permissive license to accelerate the research and development of microscopy foundation models.

Immunohistochemistry guided segmentation of benign epithelial cells, in situ lesions, and invasive epithelial cells in breast cancer slides

Digital pathology enables automatic analysis of histopathological sections using artificial intelligence (AI). Automatic evaluation could improve diagnostic efficiency and help find associations between morphological features and clinical outcome. For development of such prediction models, identifying invasive epithelial cells, and separating these from benign epithelial cells and in situ lesions would be the first step. In this study, we aimed to develop an AI model for segmentation of epithelial cells in sections from breast cancer. We generated epithelial ground truth masks by restaining hematoxylin and eosin (HE) sections with cytokeratin (CK) AE1/AE3, and by pathologists' annotations. HE/CK image pairs were used to train a convolutional neural network, and data augmentation was used to make the model more robust. Tissue microarrays (TMAs) from 839 patients, and whole slide images from two patients were used for training and evaluation of the models. The sections were derived from four cohorts of breast cancer patients. TMAs from 21 patients from a fifth cohort was used as a second test set. In quantitative evaluation, a mean Dice score of 0.70, 0.79, and 0.75 for invasive epithelial cells, benign epithelial cells, and in situ lesions, respectively, were achieved. In qualitative scoring (0-5) by pathologists, results were best for all epithelium and invasive epithelium, with scores of 4.7 and 4.4. Scores for benign epithelium and in situ lesions were 3.7 and 2.0. The proposed model segmented epithelial cells in HE stained breast cancer slides well, but further work is needed for accurate division between the classes. Immunohistochemistry, together with pathologists' annotations, enabled the creation of accurate ground truths. The model is made freely available in FastPathology and the code is available at https://github.com/AICAN-Research/breast-epithelium-segmentation

PathoHR: Breast Cancer Survival Prediction on High-Resolution Pathological Images

Breast cancer survival prediction in computational pathology presents a remarkable challenge due to tumor heterogeneity. For instance, different regions of the same tumor in the pathology image can show distinct morphological and molecular characteristics. This makes it difficult to extract representative features from whole slide images (WSIs) that truly reflect the tumor's aggressive potential and likely survival outcomes. In this paper, we present PathoHR, a novel pipeline for accurate breast cancer survival prediction that enhances any size of pathological images to enable more effective feature learning. Our approach entails (1) the incorporation of a plug-and-play high-resolution Vision Transformer (ViT) to enhance patch-wise WSI representation, enabling more detailed and comprehensive feature extraction, (2) the systematic evaluation of multiple advanced similarity metrics for comparing WSI-extracted features, optimizing the representation learning process to better capture tumor characteristics, (3) the demonstration that smaller image patches enhanced follow the proposed pipeline can achieve equivalent or superior prediction accuracy compared to raw larger patches, while significantly reducing computational overhead. Experimental findings valid that PathoHR provides the potential way of integrating enhanced image resolution with optimized feature learning to advance computational pathology, offering a promising direction for more accurate and efficient breast cancer survival prediction. Code will be available at https://github.com/AIGeeksGroup/PathoHR.

PLUTO: Pathology-Universal Transformer

Pathology is the study of microscopic inspection of tissue, and a pathology diagnosis is often the medical gold standard to diagnose disease. Pathology images provide a unique challenge for computer-vision-based analysis: a single pathology Whole Slide Image (WSI) is gigapixel-sized and often contains hundreds of thousands to millions of objects of interest across multiple resolutions. In this work, we propose PathoLogy Universal TransfOrmer (PLUTO): a light-weight pathology FM that is pre-trained on a diverse dataset of 195 million image tiles collected from multiple sites and extracts meaningful representations across multiple WSI scales that enable a large variety of downstream pathology tasks. In particular, we design task-specific adaptation heads that utilize PLUTO's output embeddings for tasks which span pathology scales ranging from subcellular to slide-scale, including instance segmentation, tile classification, and slide-level prediction. We compare PLUTO's performance to other state-of-the-art methods on a diverse set of external and internal benchmarks covering multiple biologically relevant tasks, tissue types, resolutions, stains, and scanners. We find that PLUTO matches or outperforms existing task-specific baselines and pathology-specific foundation models, some of which use orders-of-magnitude larger datasets and model sizes when compared to PLUTO. Our findings present a path towards a universal embedding to power pathology image analysis, and motivate further exploration around pathology foundation models in terms of data diversity, architectural improvements, sample efficiency, and practical deployability in real-world applications.

Planting a SEED of Vision in Large Language Model

We present SEED, an elaborate image tokenizer that empowers Large Language Models (LLMs) with the emergent ability to SEE and Draw at the same time. Research on image tokenizers has previously reached an impasse, as frameworks employing quantized visual tokens have lost prominence due to subpar performance and convergence in multimodal comprehension (compared to BLIP-2, etc.) or generation (compared to Stable Diffusion, etc.). Despite the limitations, we remain confident in its natural capacity to unify visual and textual representations, facilitating scalable multimodal training with LLM's original recipe. In this study, we identify two crucial principles for the architecture and training of SEED that effectively ease subsequent alignment with LLMs. (1) Image tokens should be independent of 2D physical patch positions and instead be produced with a 1D causal dependency, exhibiting intrinsic interdependence that aligns with the left-to-right autoregressive prediction mechanism in LLMs. (2) Image tokens should capture high-level semantics consistent with the degree of semantic abstraction in words, and be optimized for both discriminativeness and reconstruction during the tokenizer training phase. As a result, the off-the-shelf LLM is able to perform both image-to-text and text-to-image generation by incorporating our SEED through efficient LoRA tuning. Comprehensive multimodal pretraining and instruction tuning, which may yield improved results, are reserved for future investigation. This version of SEED was trained in 5.7 days using only 64 V100 GPUs and 5M publicly available image-text pairs. Our preliminary study emphasizes the great potential of discrete visual tokens in versatile multimodal LLMs and the importance of proper image tokenizers in broader research.

MAMMAL -- Molecular Aligned Multi-Modal Architecture and Language

Drug discovery typically consists of multiple steps, including identifying a target protein key to a disease's etiology, validating that interacting with this target could prevent symptoms or cure the disease, discovering a small molecule or biologic therapeutic to interact with it, and optimizing the candidate molecule through a complex landscape of required properties. Drug discovery related tasks often involve prediction and generation while considering multiple entities that potentially interact, which poses a challenge for typical AI models. For this purpose we present MAMMAL - Molecular Aligned Multi-Modal Architecture and Language - a method that we applied to create a versatile multi-task foundation model ibm/biomed.omics.bl.sm.ma-ted-458m that learns from large-scale biological datasets (2 billion samples) across diverse modalities, including proteins, small molecules, and genes. We introduce a prompt syntax that supports a wide range of classification, regression, and generation tasks. It allows combining different modalities and entity types as inputs and/or outputs. Our model handles combinations of tokens and scalars and enables the generation of small molecules and proteins, property prediction, and transcriptomic lab test predictions. We evaluated the model on 11 diverse downstream tasks spanning different steps within a typical drug discovery pipeline, where it reaches new SOTA in 9 tasks and is comparable to SOTA in 2 tasks. This performance is achieved while using a unified architecture serving all tasks, in contrast to the original SOTA performance achieved using tailored architectures. The model code and pretrained weights are publicly available at https://github.com/BiomedSciAI/biomed-multi-alignment and https://huggingface.co/ibm/biomed.omics.bl.sm.ma-ted-458m.

QuantNAS for super resolution: searching for efficient quantization-friendly architectures against quantization noise

There is a constant need for high-performing and computationally efficient neural network models for image super-resolution: computationally efficient models can be used via low-capacity devices and reduce carbon footprints. One way to obtain such models is to compress models, e.g. quantization. Another way is a neural architecture search that automatically discovers new, more efficient solutions. We propose a novel quantization-aware procedure, the QuantNAS that combines pros of these two approaches. To make QuantNAS work, the procedure looks for quantization-friendly super-resolution models. The approach utilizes entropy regularization, quantization noise, and Adaptive Deviation for Quantization (ADQ) module to enhance the search procedure. The entropy regularization technique prioritizes a single operation within each block of the search space. Adding quantization noise to parameters and activations approximates model degradation after quantization, resulting in a more quantization-friendly architectures. ADQ helps to alleviate problems caused by Batch Norm blocks in super-resolution models. Our experimental results show that the proposed approximations are better for search procedure than direct model quantization. QuantNAS discovers architectures with better PSNR/BitOps trade-off than uniform or mixed precision quantization of fixed architectures. We showcase the effectiveness of our method through its application to two search spaces inspired by the state-of-the-art SR models and RFDN. Thus, anyone can design a proper search space based on an existing architecture and apply our method to obtain better quality and efficiency. The proposed procedure is 30\% faster than direct weight quantization and is more stable.

MIPHEI-ViT: Multiplex Immunofluorescence Prediction from H&E Images using ViT Foundation Models

Histopathological analysis is a cornerstone of cancer diagnosis, with Hematoxylin and Eosin (H&E) staining routinely acquired for every patient to visualize cell morphology and tissue architecture. On the other hand, multiplex immunofluorescence (mIF) enables more precise cell type identification via proteomic markers, but has yet to achieve widespread clinical adoption due to cost and logistical constraints. To bridge this gap, we introduce MIPHEI (Multiplex Immunofluorescence Prediction from H&E), a U-Net-inspired architecture that integrates state-of-the-art ViT foundation models as encoders to predict mIF signals from H&E images. MIPHEI targets a comprehensive panel of markers spanning nuclear content, immune lineages (T cells, B cells, myeloid), epithelium, stroma, vasculature, and proliferation. We train our model using the publicly available ORION dataset of restained H&E and mIF images from colorectal cancer tissue, and validate it on two independent datasets. MIPHEI achieves accurate cell-type classification from H&E alone, with F1 scores of 0.88 for Pan-CK, 0.57 for CD3e, 0.56 for SMA, 0.36 for CD68, and 0.30 for CD20, substantially outperforming both a state-of-the-art baseline and a random classifier for most markers. Our results indicate that our model effectively captures the complex relationships between nuclear morphologies in their tissue context, as visible in H&E images and molecular markers defining specific cell types. MIPHEI offers a promising step toward enabling cell-type-aware analysis of large-scale H&E datasets, in view of uncovering relationships between spatial cellular organization and patient outcomes.

seq-JEPA: Autoregressive Predictive Learning of Invariant-Equivariant World Models

Current self-supervised algorithms commonly rely on transformations such as data augmentation and masking to learn visual representations. This is achieved by enforcing invariance or equivariance with respect to these transformations after encoding two views of an image. This dominant two-view paradigm often limits the flexibility of learned representations for downstream adaptation by creating performance trade-offs between high-level invariance-demanding tasks such as image classification and more fine-grained equivariance-related tasks. In this work, we proposes seq-JEPA, a world modeling framework that introduces architectural inductive biases into joint-embedding predictive architectures to resolve this trade-off. Without relying on dual equivariance predictors or loss terms, seq-JEPA simultaneously learns two architecturally segregated representations: one equivariant to specified transformations and another invariant to them. To do so, our model processes short sequences of different views (observations) of inputs. Each encoded view is concatenated with an embedding of the relative transformation (action) that produces the next observation in the sequence. These view-action pairs are passed through a transformer encoder that outputs an aggregate representation. A predictor head then conditions this aggregate representation on the upcoming action to predict the representation of the next observation. Empirically, seq-JEPA demonstrates strong performance on both equivariant and invariant benchmarks without sacrificing one for the other. Furthermore, it excels at tasks that inherently require aggregating a sequence of observations, such as path integration across actions and predictive learning across eye movements.

Robust Latent Matters: Boosting Image Generation with Sampling Error

Recent image generation schemes typically capture image distribution in a pre-constructed latent space relying on a frozen image tokenizer. Though the performance of tokenizer plays an essential role to the successful generation, its current evaluation metrics (e.g. rFID) fail to precisely assess the tokenizer and correlate its performance to the generation quality (e.g. gFID). In this paper, we comprehensively analyze the reason for the discrepancy of reconstruction and generation qualities in a discrete latent space, and, from which, we propose a novel plug-and-play tokenizer training scheme to facilitate latent space construction. Specifically, a latent perturbation approach is proposed to simulate sampling noises, i.e., the unexpected tokens sampled, from the generative process. With the latent perturbation, we further propose (1) a novel tokenizer evaluation metric, i.e., pFID, which successfully correlates the tokenizer performance to generation quality and (2) a plug-and-play tokenizer training scheme, which significantly enhances the robustness of tokenizer thus boosting the generation quality and convergence speed. Extensive benchmarking are conducted with 11 advanced discrete image tokenizers with 2 autoregressive generation models to validate our approach. The tokenizer trained with our proposed latent perturbation achieve a notable 1.60 gFID with classifier-free guidance (CFG) and 3.45 gFID without CFG with a sim400M generator. Code: https://github.com/lxa9867/ImageFolder.

Fast Certified Robust Training with Short Warmup

Recently, bound propagation based certified robust training methods have been proposed for training neural networks with certifiable robustness guarantees. Despite that state-of-the-art (SOTA) methods including interval bound propagation (IBP) and CROWN-IBP have per-batch training complexity similar to standard neural network training, they usually use a long warmup schedule with hundreds or thousands epochs to reach SOTA performance and are thus still costly. In this paper, we identify two important issues in existing methods, namely exploded bounds at initialization, and the imbalance in ReLU activation states and improve IBP training. These two issues make certified training difficult and unstable, and thereby long warmup schedules were needed in prior works. To mitigate these issues and conduct faster certified training with shorter warmup, we propose three improvements based on IBP training: 1) We derive a new weight initialization method for IBP training; 2) We propose to fully add Batch Normalization (BN) to each layer in the model, since we find BN can reduce the imbalance in ReLU activation states; 3) We also design regularization to explicitly tighten certified bounds and balance ReLU activation states during wamrup. We are able to obtain 65.03% verified error on CIFAR-10 (epsilon=8{255}) and 82.36% verified error on TinyImageNet (epsilon=1{255}) using very short training schedules (160 and 80 total epochs, respectively), outperforming literature SOTA trained with hundreds or thousands epochs under the same network architecture. The code is available at https://github.com/shizhouxing/Fast-Certified-Robust-Training.

Token-Shuffle: Towards High-Resolution Image Generation with Autoregressive Models

Autoregressive (AR) models, long dominant in language generation, are increasingly applied to image synthesis but are often considered less competitive than Diffusion-based models. A primary limitation is the substantial number of image tokens required for AR models, which constrains both training and inference efficiency, as well as image resolution. To address this, we present Token-Shuffle, a novel yet simple method that reduces the number of image tokens in Transformer. Our key insight is the dimensional redundancy of visual vocabularies in Multimodal Large Language Models (MLLMs), where low-dimensional visual codes from visual encoder are directly mapped to high-dimensional language vocabularies. Leveraging this, we consider two key operations: token-shuffle, which merges spatially local tokens along channel dimension to decrease the input token number, and token-unshuffle, which untangles the inferred tokens after Transformer blocks to restore the spatial arrangement for output. Jointly training with textual prompts, our strategy requires no additional pretrained text-encoder and enables MLLMs to support extremely high-resolution image synthesis in a unified next-token prediction way while maintaining efficient training and inference. For the first time, we push the boundary of AR text-to-image generation to a resolution of 2048x2048 with gratifying generation performance. In GenAI-benchmark, our 2.7B model achieves 0.77 overall score on hard prompts, outperforming AR models LlamaGen by 0.18 and diffusion models LDM by 0.15. Exhaustive large-scale human evaluations also demonstrate our prominent image generation ability in terms of text-alignment, visual flaw, and visual appearance. We hope that Token-Shuffle can serve as a foundational design for efficient high-resolution image generation within MLLMs.

FlexTok: Resampling Images into 1D Token Sequences of Flexible Length

Image tokenization has enabled major advances in autoregressive image generation by providing compressed, discrete representations that are more efficient to process than raw pixels. While traditional approaches use 2D grid tokenization, recent methods like TiTok have shown that 1D tokenization can achieve high generation quality by eliminating grid redundancies. However, these methods typically use a fixed number of tokens and thus cannot adapt to an image's inherent complexity. We introduce FlexTok, a tokenizer that projects 2D images into variable-length, ordered 1D token sequences. For example, a 256x256 image can be resampled into anywhere from 1 to 256 discrete tokens, hierarchically and semantically compressing its information. By training a rectified flow model as the decoder and using nested dropout, FlexTok produces plausible reconstructions regardless of the chosen token sequence length. We evaluate our approach in an autoregressive generation setting using a simple GPT-style Transformer. On ImageNet, this approach achieves an FID<2 across 8 to 128 tokens, outperforming TiTok and matching state-of-the-art methods with far fewer tokens. We further extend the model to support to text-conditioned image generation and examine how FlexTok relates to traditional 2D tokenization. A key finding is that FlexTok enables next-token prediction to describe images in a coarse-to-fine "visual vocabulary", and that the number of tokens to generate depends on the complexity of the generation task.

Optimizing Brain Tumor Segmentation with MedNeXt: BraTS 2024 SSA and Pediatrics

Identifying key pathological features in brain MRIs is crucial for the long-term survival of glioma patients. However, manual segmentation is time-consuming, requiring expert intervention and is susceptible to human error. Therefore, significant research has been devoted to developing machine learning methods that can accurately segment tumors in 3D multimodal brain MRI scans. Despite their progress, state-of-the-art models are often limited by the data they are trained on, raising concerns about their reliability when applied to diverse populations that may introduce distribution shifts. Such shifts can stem from lower quality MRI technology (e.g., in sub-Saharan Africa) or variations in patient demographics (e.g., children). The BraTS-2024 challenge provides a platform to address these issues. This study presents our methodology for segmenting tumors in the BraTS-2024 SSA and Pediatric Tumors tasks using MedNeXt, comprehensive model ensembling, and thorough postprocessing. Our approach demonstrated strong performance on the unseen validation set, achieving an average Dice Similarity Coefficient (DSC) of 0.896 on the BraTS-2024 SSA dataset and an average DSC of 0.830 on the BraTS Pediatric Tumor dataset. Additionally, our method achieved an average Hausdorff Distance (HD95) of 14.682 on the BraTS-2024 SSA dataset and an average HD95 of 37.508 on the BraTS Pediatric dataset. Our GitHub repository can be accessed here: Project Repository : https://github.com/python-arch/BioMbz-Optimizing-Brain-Tumor-Segmentation-with-MedNeXt-BraTS-2024-SSA-and-Pediatrics

On-device Online Learning and Semantic Management of TinyML Systems

Recent advances in Tiny Machine Learning (TinyML) empower low-footprint embedded devices for real-time on-device Machine Learning. While many acknowledge the potential benefits of TinyML, its practical implementation presents unique challenges. This study aims to bridge the gap between prototyping single TinyML models and developing reliable TinyML systems in production: (1) Embedded devices operate in dynamically changing conditions. Existing TinyML solutions primarily focus on inference, with models trained offline on powerful machines and deployed as static objects. However, static models may underperform in the real world due to evolving input data distributions. We propose online learning to enable training on constrained devices, adapting local models towards the latest field conditions. (2) Nevertheless, current on-device learning methods struggle with heterogeneous deployment conditions and the scarcity of labeled data when applied across numerous devices. We introduce federated meta-learning incorporating online learning to enhance model generalization, facilitating rapid learning. This approach ensures optimal performance among distributed devices by knowledge sharing. (3) Moreover, TinyML's pivotal advantage is widespread adoption. Embedded devices and TinyML models prioritize extreme efficiency, leading to diverse characteristics ranging from memory and sensors to model architectures. Given their diversity and non-standardized representations, managing these resources becomes challenging as TinyML systems scale up. We present semantic management for the joint management of models and devices at scale. We demonstrate our methods through a basic regression example and then assess them in three real-world TinyML applications: handwritten character image classification, keyword audio classification, and smart building presence detection, confirming our approaches' effectiveness.

Weakly Supervised Fine-grained Scene Graph Generation via Large Language Model

Weakly-Supervised Scene Graph Generation (WSSGG) research has recently emerged as an alternative to the fully-supervised approach that heavily relies on costly annotations. In this regard, studies on WSSGG have utilized image captions to obtain unlocalized triplets while primarily focusing on grounding the unlocalized triplets over image regions. However, they have overlooked the two issues involved in the triplet formation process from the captions: 1) Semantic over-simplification issue arises when extracting triplets from captions, where fine-grained predicates in captions are undesirably converted into coarse-grained predicates, resulting in a long-tailed predicate distribution, and 2) Low-density scene graph issue arises when aligning the triplets in the caption with entity/predicate classes of interest, where many triplets are discarded and not used in training, leading to insufficient supervision. To tackle the two issues, we propose a new approach, i.e., Large Language Model for weakly-supervised SGG (LLM4SGG), where we mitigate the two issues by leveraging the LLM's in-depth understanding of language and reasoning ability during the extraction of triplets from captions and alignment of entity/predicate classes with target data. To further engage the LLM in these processes, we adopt the idea of Chain-of-Thought and the in-context few-shot learning strategy. To validate the effectiveness of LLM4SGG, we conduct extensive experiments on Visual Genome and GQA datasets, showing significant improvements in both Recall@K and mean Recall@K compared to the state-of-the-art WSSGG methods. A further appeal is that LLM4SGG is data-efficient, enabling effective model training with a small amount of training images.

Molecular-driven Foundation Model for Oncologic Pathology

Foundation models are reshaping computational pathology by enabling transfer learning, where models pre-trained on vast datasets can be adapted for downstream diagnostic, prognostic, and therapeutic response tasks. Despite these advances, foundation models are still limited in their ability to encode the entire gigapixel whole-slide images without additional training and often lack complementary multimodal data. Here, we introduce Threads, a slide-level foundation model capable of generating universal representations of whole-slide images of any size. Threads was pre-trained using a multimodal learning approach on a diverse cohort of 47,171 hematoxylin and eosin (H&E)-stained tissue sections, paired with corresponding genomic and transcriptomic profiles - the largest such paired dataset to be used for foundation model development to date. This unique training paradigm enables Threads to capture the tissue's underlying molecular composition, yielding powerful representations applicable to a wide array of downstream tasks. In extensive benchmarking across 54 oncology tasks, including clinical subtyping, grading, mutation prediction, immunohistochemistry status determination, treatment response prediction, and survival prediction, Threads outperformed all baselines while demonstrating remarkable generalizability and label efficiency. It is particularly well suited for predicting rare events, further emphasizing its clinical utility. We intend to make the model publicly available for the broader community.

A Multi-Modal AI Copilot for Single-Cell Analysis with Instruction Following

Large language models excel at interpreting complex natural language instructions, enabling them to perform a wide range of tasks. In the life sciences, single-cell RNA sequencing (scRNA-seq) data serves as the "language of cellular biology", capturing intricate gene expression patterns at the single-cell level. However, interacting with this "language" through conventional tools is often inefficient and unintuitive, posing challenges for researchers. To address these limitations, we present InstructCell, a multi-modal AI copilot that leverages natural language as a medium for more direct and flexible single-cell analysis. We construct a comprehensive multi-modal instruction dataset that pairs text-based instructions with scRNA-seq profiles from diverse tissues and species. Building on this, we develop a multi-modal cell language architecture capable of simultaneously interpreting and processing both modalities. InstructCell empowers researchers to accomplish critical tasks-such as cell type annotation, conditional pseudo-cell generation, and drug sensitivity prediction-using straightforward natural language commands. Extensive evaluations demonstrate that InstructCell consistently meets or exceeds the performance of existing single-cell foundation models, while adapting to diverse experimental conditions. More importantly, InstructCell provides an accessible and intuitive tool for exploring complex single-cell data, lowering technical barriers and enabling deeper biological insights.

Unlasting: Unpaired Single-Cell Multi-Perturbation Estimation by Dual Conditional Diffusion Implicit Bridges

Estimating single-cell responses across various perturbations facilitates the identification of key genes and enhances drug screening, significantly boosting experimental efficiency. However, single-cell sequencing is a destructive process, making it impossible to capture the same cell's phenotype before and after perturbation. Consequently, data collected under perturbed and unperturbed conditions are inherently unpaired. Existing methods either attempt to forcibly pair unpaired data using random sampling, or neglect the inherent relationship between unperturbed and perturbed cells during the modeling. In this work, we propose a framework based on Dual Diffusion Implicit Bridges (DDIB) to learn the mapping between different data distributions, effectively addressing the challenge of unpaired data. We further interpret this framework as a form of data augmentation. We integrate gene regulatory network (GRN) information to propagate perturbation signals in a biologically meaningful way, and further incorporate a masking mechanism to predict silent genes, improving the quality of generated profiles. Moreover, gene expression under the same perturbation often varies significantly across cells, frequently exhibiting a bimodal distribution that reflects intrinsic heterogeneity. To capture this, we introduce a more suitable evaluation metric. We propose Unlasting, dual conditional diffusion models that overcome the problem of unpaired single-cell perturbation data and strengthen the model's insight into perturbations under the guidance of the GRN, with a dedicated mask model designed to improve generation quality by predicting silent genes. In addition, we introduce a biologically grounded evaluation metric that better reflects the inherent heterogeneity in single-cell responses.

MedITok: A Unified Tokenizer for Medical Image Synthesis and Interpretation

Advanced autoregressive models have reshaped multimodal AI. However, their transformative potential in medical imaging remains largely untapped due to the absence of a unified visual tokenizer -- one capable of capturing fine-grained visual structures for faithful image reconstruction and realistic image synthesis, as well as rich semantics for accurate diagnosis and image interpretation. To this end, we present MedITok, the first unified tokenizer tailored for medical images, encoding both low-level structural details and high-level clinical semantics within a unified latent space. To balance these competing objectives, we introduce a novel two-stage training framework: a visual representation alignment stage that cold-starts the tokenizer reconstruction learning with a visual semantic constraint, followed by a textual semantic representation alignment stage that infuses detailed clinical semantics into the latent space. Trained on the meticulously collected large-scale dataset with over 30 million medical images and 2 million image-caption pairs, MedITok achieves state-of-the-art performance on more than 30 datasets across 9 imaging modalities and 4 different tasks. By providing a unified token space for autoregressive modeling, MedITok supports a wide range of tasks in clinical diagnostics and generative healthcare applications. Model and code will be made publicly available at: https://github.com/Masaaki-75/meditok.

ADDP: Learning General Representations for Image Recognition and Generation with Alternating Denoising Diffusion Process

Image recognition and generation have long been developed independently of each other. With the recent trend towards general-purpose representation learning, the development of general representations for both recognition and generation tasks is also promoted. However, preliminary attempts mainly focus on generation performance, but are still inferior on recognition tasks. These methods are modeled in the vector-quantized (VQ) space, whereas leading recognition methods use pixels as inputs. Our key insights are twofold: (1) pixels as inputs are crucial for recognition tasks; (2) VQ tokens as reconstruction targets are beneficial for generation tasks. These observations motivate us to propose an Alternating Denoising Diffusion Process (ADDP) that integrates these two spaces within a single representation learning framework. In each denoising step, our method first decodes pixels from previous VQ tokens, then generates new VQ tokens from the decoded pixels. The diffusion process gradually masks out a portion of VQ tokens to construct the training samples. The learned representations can be used to generate diverse high-fidelity images and also demonstrate excellent transfer performance on recognition tasks. Extensive experiments show that our method achieves competitive performance on unconditional generation, ImageNet classification, COCO detection, and ADE20k segmentation. Importantly, our method represents the first successful development of general representations applicable to both generation and dense recognition tasks. Code shall be released.

PIE: Simulating Disease Progression via Progressive Image Editing

Disease progression simulation is a crucial area of research that has significant implications for clinical diagnosis, prognosis, and treatment. One major challenge in this field is the lack of continuous medical imaging monitoring of individual patients over time. To address this issue, we develop a novel framework termed Progressive Image Editing (PIE) that enables controlled manipulation of disease-related image features, facilitating precise and realistic disease progression simulation. Specifically, we leverage recent advancements in text-to-image generative models to simulate disease progression accurately and personalize it for each patient. We theoretically analyze the iterative refining process in our framework as a gradient descent with an exponentially decayed learning rate. To validate our framework, we conduct experiments in three medical imaging domains. Our results demonstrate the superiority of PIE over existing methods such as Stable Diffusion Walk and Style-Based Manifold Extrapolation based on CLIP score (Realism) and Disease Classification Confidence (Alignment). Our user study collected feedback from 35 veteran physicians to assess the generated progressions. Remarkably, 76.2% of the feedback agrees with the fidelity of the generated progressions. To our best knowledge, PIE is the first of its kind to generate disease progression images meeting real-world standards. It is a promising tool for medical research and clinical practice, potentially allowing healthcare providers to model disease trajectories over time, predict future treatment responses, and improve patient outcomes.

LEOPARD : A Vision Language Model For Text-Rich Multi-Image Tasks

Text-rich images, where text serves as the central visual element guiding the overall understanding, are prevalent in real-world applications, such as presentation slides, scanned documents, and webpage snapshots. Tasks involving multiple text-rich images are especially challenging, as they require not only understanding the content of individual images but reasoning about inter-relationships and logical flows across multiple visual inputs. Despite the importance of these scenarios, current multimodal large language models (MLLMs) struggle to handle such tasks due to two key challenges: (1) the scarcity of high-quality instruction tuning datasets for text-rich multi-image scenarios, and (2) the difficulty in balancing image resolution with visual feature sequence length. To address these challenges, we propose \OurMethod, a MLLM designed specifically for handling vision-language tasks involving multiple text-rich images. First, we curated about one million high-quality multimodal instruction-tuning data, tailored to text-rich, multi-image scenarios. Second, we developed an adaptive high-resolution multi-image encoding module to dynamically optimize the allocation of visual sequence length based on the original aspect ratios and resolutions of the input images. Experiments across a wide range of benchmarks demonstrate our model's superior capabilities in text-rich, multi-image evaluations and competitive performance in general domain evaluations.

FlashAttention: Fast and Memory-Efficient Exact Attention with IO-Awareness

Transformers are slow and memory-hungry on long sequences, since the time and memory complexity of self-attention are quadratic in sequence length. Approximate attention methods have attempted to address this problem by trading off model quality to reduce the compute complexity, but often do not achieve wall-clock speedup. We argue that a missing principle is making attention algorithms IO-aware -- accounting for reads and writes between levels of GPU memory. We propose FlashAttention, an IO-aware exact attention algorithm that uses tiling to reduce the number of memory reads/writes between GPU high bandwidth memory (HBM) and GPU on-chip SRAM. We analyze the IO complexity of FlashAttention, showing that it requires fewer HBM accesses than standard attention, and is optimal for a range of SRAM sizes. We also extend FlashAttention to block-sparse attention, yielding an approximate attention algorithm that is faster than any existing approximate attention method. FlashAttention trains Transformers faster than existing baselines: 15% end-to-end wall-clock speedup on BERT-large (seq. length 512) compared to the MLPerf 1.1 training speed record, 3times speedup on GPT-2 (seq. length 1K), and 2.4times speedup on long-range arena (seq. length 1K-4K). FlashAttention and block-sparse FlashAttention enable longer context in Transformers, yielding higher quality models (0.7 better perplexity on GPT-2 and 6.4 points of lift on long-document classification) and entirely new capabilities: the first Transformers to achieve better-than-chance performance on the Path-X challenge (seq. length 16K, 61.4% accuracy) and Path-256 (seq. length 64K, 63.1% accuracy).

Inducing High Energy-Latency of Large Vision-Language Models with Verbose Images

Large vision-language models (VLMs) such as GPT-4 have achieved exceptional performance across various multi-modal tasks. However, the deployment of VLMs necessitates substantial energy consumption and computational resources. Once attackers maliciously induce high energy consumption and latency time (energy-latency cost) during inference of VLMs, it will exhaust computational resources. In this paper, we explore this attack surface about availability of VLMs and aim to induce high energy-latency cost during inference of VLMs. We find that high energy-latency cost during inference of VLMs can be manipulated by maximizing the length of generated sequences. To this end, we propose verbose images, with the goal of crafting an imperceptible perturbation to induce VLMs to generate long sentences during inference. Concretely, we design three loss objectives. First, a loss is proposed to delay the occurrence of end-of-sequence (EOS) token, where EOS token is a signal for VLMs to stop generating further tokens. Moreover, an uncertainty loss and a token diversity loss are proposed to increase the uncertainty over each generated token and the diversity among all tokens of the whole generated sequence, respectively, which can break output dependency at token-level and sequence-level. Furthermore, a temporal weight adjustment algorithm is proposed, which can effectively balance these losses. Extensive experiments demonstrate that our verbose images can increase the length of generated sequences by 7.87 times and 8.56 times compared to original images on MS-COCO and ImageNet datasets, which presents potential challenges for various applications. Our code is available at https://github.com/KuofengGao/Verbose_Images.

DNABERT-S: Learning Species-Aware DNA Embedding with Genome Foundation Models

Effective DNA embedding remains crucial in genomic analysis, particularly in scenarios lacking labeled data for model fine-tuning, despite the significant advancements in genome foundation models. A prime example is metagenomics binning, a critical process in microbiome research that aims to group DNA sequences by their species from a complex mixture of DNA sequences derived from potentially thousands of distinct, often uncharacterized species. To fill the lack of effective DNA embedding models, we introduce DNABERT-S, a genome foundation model that specializes in creating species-aware DNA embeddings. To encourage effective embeddings to error-prone long-read DNA sequences, we introduce Manifold Instance Mixup (MI-Mix), a contrastive objective that mixes the hidden representations of DNA sequences at randomly selected layers and trains the model to recognize and differentiate these mixed proportions at the output layer. We further enhance it with the proposed Curriculum Contrastive Learning (C^2LR) strategy. Empirical results on 18 diverse datasets showed DNABERT-S's remarkable performance. It outperforms the top baseline's performance in 10-shot species classification with just a 2-shot training while doubling the Adjusted Rand Index (ARI) in species clustering and substantially increasing the number of correctly identified species in metagenomics binning. The code, data, and pre-trained model are publicly available at https://github.com/Zhihan1996/DNABERT_S.

Escaping the Big Data Paradigm with Compact Transformers

With the rise of Transformers as the standard for language processing, and their advancements in computer vision, there has been a corresponding growth in parameter size and amounts of training data. Many have come to believe that because of this, transformers are not suitable for small sets of data. This trend leads to concerns such as: limited availability of data in certain scientific domains and the exclusion of those with limited resource from research in the field. In this paper, we aim to present an approach for small-scale learning by introducing Compact Transformers. We show for the first time that with the right size, convolutional tokenization, transformers can avoid overfitting and outperform state-of-the-art CNNs on small datasets. Our models are flexible in terms of model size, and can have as little as 0.28M parameters while achieving competitive results. Our best model can reach 98% accuracy when training from scratch on CIFAR-10 with only 3.7M parameters, which is a significant improvement in data-efficiency over previous Transformer based models being over 10x smaller than other transformers and is 15% the size of ResNet50 while achieving similar performance. CCT also outperforms many modern CNN based approaches, and even some recent NAS-based approaches. Additionally, we obtain a new SOTA result on Flowers-102 with 99.76% top-1 accuracy, and improve upon the existing baseline on ImageNet (82.71% accuracy with 29% as many parameters as ViT), as well as NLP tasks. Our simple and compact design for transformers makes them more feasible to study for those with limited computing resources and/or dealing with small datasets, while extending existing research efforts in data efficient transformers. Our code and pre-trained models are publicly available at https://github.com/SHI-Labs/Compact-Transformers.

ZeroQ: A Novel Zero Shot Quantization Framework

Quantization is a promising approach for reducing the inference time and memory footprint of neural networks. However, most existing quantization methods require access to the original training dataset for retraining during quantization. This is often not possible for applications with sensitive or proprietary data, e.g., due to privacy and security concerns. Existing zero-shot quantization methods use different heuristics to address this, but they result in poor performance, especially when quantizing to ultra-low precision. Here, we propose ZeroQ , a novel zero-shot quantization framework to address this. ZeroQ enables mixed-precision quantization without any access to the training or validation data. This is achieved by optimizing for a Distilled Dataset, which is engineered to match the statistics of batch normalization across different layers of the network. ZeroQ supports both uniform and mixed-precision quantization. For the latter, we introduce a novel Pareto frontier based method to automatically determine the mixed-precision bit setting for all layers, with no manual search involved. We extensively test our proposed method on a diverse set of models, including ResNet18/50/152, MobileNetV2, ShuffleNet, SqueezeNext, and InceptionV3 on ImageNet, as well as RetinaNet-ResNet50 on the Microsoft COCO dataset. In particular, we show that ZeroQ can achieve 1.71\% higher accuracy on MobileNetV2, as compared to the recently proposed DFQ method. Importantly, ZeroQ has a very low computational overhead, and it can finish the entire quantization process in less than 30s (0.5\% of one epoch training time of ResNet50 on ImageNet). We have open-sourced the ZeroQ frameworkhttps://github.com/amirgholami/ZeroQ.

Mini-Monkey: Multi-Scale Adaptive Cropping for Multimodal Large Language Models

Recently, there has been significant interest in enhancing the capability of multimodal large language models (MLLMs) to process high-resolution images. Most existing methods focus on adopting a cropping strategy to improve the ability of multimodal large language models to understand image details. However, this cropping operation inevitably causes the segmentation of objects and connected areas, which impairs the MLLM's ability to recognize small or irregularly shaped objects or text. This issue is particularly evident in lightweight MLLMs. Addressing this issue, we propose Mini-Monkey, a lightweight MLLM that incorporates a plug-and-play method called multi-scale adaptive crop strategy (MSAC). Mini-Monkey adaptively generates multi-scale representations, allowing it to select non-segmented objects from various scales. To mitigate the computational overhead introduced by MSAC, we propose a Scale Compression Mechanism (SCM), which effectively compresses image tokens. Mini-Monkey achieves state-of-the-art performance among 2B-parameter MLLMs. It not only demonstrates leading performance on a variety of general multimodal understanding tasks but also shows consistent improvements in document understanding capabilities. On the OCRBench, Mini-Monkey achieves a score of 802, outperforming 8B-parameter state-of-the-art model InternVL2-8B. Besides, our model and training strategy are very efficient, which can be trained with only eight RTX 3090. The code is available at https://github.com/Yuliang-Liu/Monkey.

ProVision: Programmatically Scaling Vision-centric Instruction Data for Multimodal Language Models

With the rise of multimodal applications, instruction data has become critical for training multimodal language models capable of understanding complex image-based queries. Existing practices rely on powerful but costly large language models (LLMs) or multimodal language models (MLMs) to produce instruction data. These are often prone to hallucinations, licensing issues and the generation process is often hard to scale and interpret. In this work, we present a programmatic approach that employs scene graphs as symbolic representations of images and human-written programs to systematically synthesize vision-centric instruction data. Our approach ensures the interpretability and controllability of the data generation process and scales efficiently while maintaining factual accuracy. By implementing a suite of 24 single-image, 14 multi-image instruction generators, and a scene graph generation pipeline, we build a scalable, cost-effective system: ProVision which produces diverse question-answer pairs concerning objects, attributes, relations, depth, etc., for any given image. Applied to Visual Genome and DataComp datasets, we generate over 10 million instruction data points, ProVision-10M, and leverage them in both pretraining and instruction tuning stages of MLMs. When adopted in the instruction tuning stage, our single-image instruction data yields up to a 7% improvement on the 2D split and 8% on the 3D split of CVBench, along with a 3% increase in performance on QBench2, RealWorldQA, and MMMU. Our multi-image instruction data leads to an 8% improvement on Mantis-Eval. Incorporation of our data in both pre-training and fine-tuning stages of xGen-MM-4B leads to an averaged improvement of 1.6% across 11 benchmarks.

hist2RNA: An efficient deep learning architecture to predict gene expression from breast cancer histopathology images

Gene expression can be used to subtype breast cancer with improved prediction of risk of recurrence and treatment responsiveness over that obtained using routine immunohistochemistry (IHC). However, in the clinic, molecular profiling is primarily used for ER+ breast cancer, which is costly, tissue destructive, requires specialized platforms and takes several weeks to obtain a result. Deep learning algorithms can effectively extract morphological patterns in digital histopathology images to predict molecular phenotypes quickly and cost-effectively. We propose a new, computationally efficient approach called hist2RNA inspired by bulk RNA-sequencing techniques to predict the expression of 138 genes (incorporated from six commercially available molecular profiling tests), including luminal PAM50 subtype, from hematoxylin and eosin (H&E) stained whole slide images (WSIs). The training phase involves the aggregation of extracted features for each patient from a pretrained model to predict gene expression at the patient level using annotated H&E images from The Cancer Genome Atlas (TCGA, n=335). We demonstrate successful gene prediction on a held-out test set (n = 160, corr = 0.82 across patients, corr = 0.29 across genes) and perform exploratory analysis on an external tissue microarray (TMA) dataset (n = 498) with known IHC and survival information. Our model is able to predict gene expression and luminal PAM50 subtype (Luminal A versus Luminal B) on the TMA dataset with prognostic significance for overall survival in univariate analysis (c-index = 0.56, hazard ratio = 2.16 (95% CI 1.12-3.06), p < 5 x 10-3), and independent significance in multivariate analysis incorporating standard clinicopathological variables (c-index = 0.65, hazard ratio = 1.85 (95% CI 1.30-2.68), p < 5 x 10-3).

MCUNetV2: Memory-Efficient Patch-based Inference for Tiny Deep Learning

Tiny deep learning on microcontroller units (MCUs) is challenging due to the limited memory size. We find that the memory bottleneck is due to the imbalanced memory distribution in convolutional neural network (CNN) designs: the first several blocks have an order of magnitude larger memory usage than the rest of the network. To alleviate this issue, we propose a generic patch-by-patch inference scheduling, which operates only on a small spatial region of the feature map and significantly cuts down the peak memory. However, naive implementation brings overlapping patches and computation overhead. We further propose network redistribution to shift the receptive field and FLOPs to the later stage and reduce the computation overhead. Manually redistributing the receptive field is difficult. We automate the process with neural architecture search to jointly optimize the neural architecture and inference scheduling, leading to MCUNetV2. Patch-based inference effectively reduces the peak memory usage of existing networks by 4-8x. Co-designed with neural networks, MCUNetV2 sets a record ImageNet accuracy on MCU (71.8%), and achieves >90% accuracy on the visual wake words dataset under only 32kB SRAM. MCUNetV2 also unblocks object detection on tiny devices, achieving 16.9% higher mAP on Pascal VOC compared to the state-of-the-art result. Our study largely addressed the memory bottleneck in tinyML and paved the way for various vision applications beyond image classification.

scE^2TM: Toward Interpretable Single-Cell Embedding via Topic Modeling

Recent advances in sequencing technologies have enabled researchers to explore cellular heterogeneity at single-cell resolution. Meanwhile, interpretability has gained prominence parallel to the rapid increase in the complexity and performance of deep learning models. In recent years, topic models have been widely used for interpretable single-cell embedding learning and clustering analysis, which we refer to as single-cell embedded topic models. However, previous studies evaluated the interpretability of the models mainly through qualitative analysis, and these single-cell embedded topic models suffer from the potential problem of interpretation collapse. Furthermore, their neglect of external biological knowledge constrains analytical performance. Here, we present scE2TM, an external knowledge-guided single-cell embedded topic model that provides a high-quality cell embedding and strong interpretation, contributing to comprehensive scRNA-seq data analysis. Our comprehensive evaluation across 20 scRNA-seq datasets demonstrates that scE2TM achieves significant clustering performance gains compared to 7 state-of-the-art methods. In addition, we propose a new interpretability evaluation benchmark that introduces 10 metrics to quantitatively assess the interpretability of single-cell embedded topic models. The results show that the interpretation provided by scE2TM performs encouragingly in terms of diversity and consistency with the underlying biological signals, contributing to a better revealing of the underlying biological mechanisms.

HAWQ: Hessian AWare Quantization of Neural Networks with Mixed-Precision

Model size and inference speed/power have become a major challenge in the deployment of Neural Networks for many applications. A promising approach to address these problems is quantization. However, uniformly quantizing a model to ultra low precision leads to significant accuracy degradation. A novel solution for this is to use mixed-precision quantization, as some parts of the network may allow lower precision as compared to other layers. However, there is no systematic way to determine the precision of different layers. A brute force approach is not feasible for deep networks, as the search space for mixed-precision is exponential in the number of layers. Another challenge is a similar factorial complexity for determining block-wise fine-tuning order when quantizing the model to a target precision. Here, we introduce Hessian AWare Quantization (HAWQ), a novel second-order quantization method to address these problems. HAWQ allows for the automatic selection of the relative quantization precision of each layer, based on the layer's Hessian spectrum. Moreover, HAWQ provides a deterministic fine-tuning order for quantizing layers, based on second-order information. We show the results of our method on Cifar-10 using ResNet20, and on ImageNet using Inception-V3, ResNet50 and SqueezeNext models. Comparing HAWQ with state-of-the-art shows that we can achieve similar/better accuracy with 8times activation compression ratio on ResNet20, as compared to DNAS~wu2018mixed, and up to 1% higher accuracy with up to 14% smaller models on ResNet50 and Inception-V3, compared to recently proposed methods of RVQuant~park2018value and HAQ~wang2018haq. Furthermore, we show that we can quantize SqueezeNext to just 1MB model size while achieving above 68% top1 accuracy on ImageNet.

CellForge: Agentic Design of Virtual Cell Models

Virtual cell modeling represents an emerging frontier at the intersection of artificial intelligence and biology, aiming to predict quantities such as responses to diverse perturbations quantitatively. However, autonomously building computational models for virtual cells is challenging due to the complexity of biological systems, the heterogeneity of data modalities, and the need for domain-specific expertise across multiple disciplines. Here, we introduce CellForge, an agentic system that leverages a multi-agent framework that transforms presented biological datasets and research objectives directly into optimized computational models for virtual cells. More specifically, given only raw single-cell multi-omics data and task descriptions as input, CellForge outputs both an optimized model architecture and executable code for training virtual cell models and inference. The framework integrates three core modules: Task Analysis for presented dataset characterization and relevant literature retrieval, Method Design, where specialized agents collaboratively develop optimized modeling strategies, and Experiment Execution for automated generation of code. The agents in the Design module are separated into experts with differing perspectives and a central moderator, and have to collaboratively exchange solutions until they achieve a reasonable consensus. We demonstrate CellForge's capabilities in single-cell perturbation prediction, using six diverse datasets that encompass gene knockouts, drug treatments, and cytokine stimulations across multiple modalities. CellForge consistently outperforms task-specific state-of-the-art methods. Overall, CellForge demonstrates how iterative interaction between LLM agents with differing perspectives provides better solutions than directly addressing a modeling challenge. Our code is publicly available at https://github.com/gersteinlab/CellForge.

TinySAM: Pushing the Envelope for Efficient Segment Anything Model

Recently segment anything model (SAM) has shown powerful segmentation capability and has drawn great attention in computer vision fields. Massive following works have developed various applications based on the pretrained SAM and achieved impressive performance on downstream vision tasks. However, SAM consists of heavy architectures and requires massive computational capacity, which hinders the further application of SAM on computation constrained edge devices. To this end, in this paper we propose a framework to obtain a tiny segment anything model (TinySAM) while maintaining the strong zero-shot performance. We first propose a full-stage knowledge distillation method with online hard prompt sampling strategy to distill a lightweight student model. We also adapt the post-training quantization to the promptable segmentation task and further reduce the computational cost. Moreover, a hierarchical segmenting everything strategy is proposed to accelerate the everything inference by 2times with almost no performance degradation. With all these proposed methods, our TinySAM leads to orders of magnitude computational reduction and pushes the envelope for efficient segment anything task. Extensive experiments on various zero-shot transfer tasks demonstrate the significantly advantageous performance of our TinySAM against counterpart methods. Pre-trained models and codes will be available at https://github.com/xinghaochen/TinySAM and https://gitee.com/mindspore/models/tree/master/research/cv/TinySAM.

UniGenX: Unified Generation of Sequence and Structure with Autoregressive Diffusion

Unified generation of sequence and structure for scientific data (e.g., materials, molecules, proteins) is a critical task. Existing approaches primarily rely on either autoregressive sequence models or diffusion models, each offering distinct advantages and facing notable limitations. Autoregressive models, such as GPT, Llama, and Phi-4, have demonstrated remarkable success in natural language generation and have been extended to multimodal tasks (e.g., image, video, and audio) using advanced encoders like VQ-VAE to represent complex modalities as discrete sequences. However, their direct application to scientific domains is challenging due to the high precision requirements and the diverse nature of scientific data. On the other hand, diffusion models excel at generating high-dimensional scientific data, such as protein, molecule, and material structures, with remarkable accuracy. Yet, their inability to effectively model sequences limits their potential as general-purpose multimodal foundation models. To address these challenges, we propose UniGenX, a unified framework that combines autoregressive next-token prediction with conditional diffusion models. This integration leverages the strengths of autoregressive models to ease the training of conditional diffusion models, while diffusion-based generative heads enhance the precision of autoregressive predictions. We validate the effectiveness of UniGenX on material and small molecule generation tasks, achieving a significant leap in state-of-the-art performance for material crystal structure prediction and establishing new state-of-the-art results for small molecule structure prediction, de novo design, and conditional generation. Notably, UniGenX demonstrates significant improvements, especially in handling long sequences for complex structures, showcasing its efficacy as a versatile tool for scientific data generation.

StRegA: Unsupervised Anomaly Detection in Brain MRIs using a Compact Context-encoding Variational Autoencoder

Expert interpretation of anatomical images of the human brain is the central part of neuro-radiology. Several machine learning-based techniques have been proposed to assist in the analysis process. However, the ML models typically need to be trained to perform a specific task, e.g., brain tumour segmentation or classification. Not only do the corresponding training data require laborious manual annotations, but a wide variety of abnormalities can be present in a human brain MRI - even more than one simultaneously, which renders representation of all possible anomalies very challenging. Hence, a possible solution is an unsupervised anomaly detection (UAD) system that can learn a data distribution from an unlabelled dataset of healthy subjects and then be applied to detect out of distribution samples. Such a technique can then be used to detect anomalies - lesions or abnormalities, for example, brain tumours, without explicitly training the model for that specific pathology. Several Variational Autoencoder (VAE) based techniques have been proposed in the past for this task. Even though they perform very well on controlled artificially simulated anomalies, many of them perform poorly while detecting anomalies in clinical data. This research proposes a compact version of the "context-encoding" VAE (ceVAE) model, combined with pre and post-processing steps, creating a UAD pipeline (StRegA), which is more robust on clinical data, and shows its applicability in detecting anomalies such as tumours in brain MRIs. The proposed pipeline achieved a Dice score of 0.642pm0.101 while detecting tumours in T2w images of the BraTS dataset and 0.859pm0.112 while detecting artificially induced anomalies, while the best performing baseline achieved 0.522pm0.135 and 0.783pm0.111, respectively.

Elucidating the design space of language models for image generation

The success of autoregressive (AR) language models in text generation has inspired the computer vision community to adopt Large Language Models (LLMs) for image generation. However, considering the essential differences between text and image modalities, the design space of language models for image generation remains underexplored. We observe that image tokens exhibit greater randomness compared to text tokens, which presents challenges when training with token prediction. Nevertheless, AR models demonstrate their potential by effectively learning patterns even from a seemingly suboptimal optimization problem. Our analysis also reveals that while all models successfully grasp the importance of local information in image generation, smaller models struggle to capture the global context. In contrast, larger models showcase improved capabilities in this area, helping to explain the performance gains achieved when scaling up model size. We further elucidate the design space of language models for vision generation, including tokenizer choice, model choice, model scalability, vocabulary design, and sampling strategy through extensive comparative experiments. Our work is the first to analyze the optimization behavior of language models in vision generation, and we believe it can inspire more effective designs when applying LMs to other domains. Finally, our elucidated language model for image generation, termed as ELM, achieves state-of-the-art performance on the ImageNet 256*256 benchmark. The code is available at https://github.com/Pepperlll/LMforImageGeneration.git.

On-Device Training Under 256KB Memory

On-device training enables the model to adapt to new data collected from the sensors by fine-tuning a pre-trained model. Users can benefit from customized AI models without having to transfer the data to the cloud, protecting the privacy. However, the training memory consumption is prohibitive for IoT devices that have tiny memory resources. We propose an algorithm-system co-design framework to make on-device training possible with only 256KB of memory. On-device training faces two unique challenges: (1) the quantized graphs of neural networks are hard to optimize due to low bit-precision and the lack of normalization; (2) the limited hardware resource does not allow full back-propagation. To cope with the optimization difficulty, we propose Quantization-Aware Scaling to calibrate the gradient scales and stabilize 8-bit quantized training. To reduce the memory footprint, we propose Sparse Update to skip the gradient computation of less important layers and sub-tensors. The algorithm innovation is implemented by a lightweight training system, Tiny Training Engine, which prunes the backward computation graph to support sparse updates and offload the runtime auto-differentiation to compile time. Our framework is the first solution to enable tiny on-device training of convolutional neural networks under 256KB SRAM and 1MB Flash without auxiliary memory, using less than 1/1000 of the memory of PyTorch and TensorFlow while matching the accuracy on tinyML application VWW. Our study enables IoT devices not only to perform inference but also to continuously adapt to new data for on-device lifelong learning. A video demo can be found here: https://youtu.be/XaDCO8YtmBw.

SANSformers: Self-Supervised Forecasting in Electronic Health Records with Attention-Free Models

Despite the proven effectiveness of Transformer neural networks across multiple domains, their performance with Electronic Health Records (EHR) can be nuanced. The unique, multidimensional sequential nature of EHR data can sometimes make even simple linear models with carefully engineered features more competitive. Thus, the advantages of Transformers, such as efficient transfer learning and improved scalability are not always fully exploited in EHR applications. Addressing these challenges, we introduce SANSformer, an attention-free sequential model designed with specific inductive biases to cater for the unique characteristics of EHR data. In this work, we aim to forecast the demand for healthcare services, by predicting the number of patient visits to healthcare facilities. The challenge amplifies when dealing with divergent patient subgroups, like those with rare diseases, which are characterized by unique health trajectories and are typically smaller in size. To address this, we employ a self-supervised pretraining strategy, Generative Summary Pretraining (GSP), which predicts future summary statistics based on past health records of a patient. Our models are pretrained on a health registry of nearly one million patients, then fine-tuned for specific subgroup prediction tasks, showcasing the potential to handle the multifaceted nature of EHR data. In evaluation, SANSformer consistently surpasses robust EHR baselines, with our GSP pretraining method notably amplifying model performance, particularly within smaller patient subgroups. Our results illuminate the promising potential of tailored attention-free models and self-supervised pretraining in refining healthcare utilization predictions across various patient demographics.

MaIR: A Locality- and Continuity-Preserving Mamba for Image Restoration

Recent advancements in Mamba have shown promising results in image restoration. These methods typically flatten 2D images into multiple distinct 1D sequences along rows and columns, process each sequence independently using selective scan operation, and recombine them to form the outputs. However, such a paradigm overlooks two vital aspects: i) the local relationships and spatial continuity inherent in natural images, and ii) the discrepancies among sequences unfolded through totally different ways. To overcome the drawbacks, we explore two problems in Mamba-based restoration methods: i) how to design a scanning strategy preserving both locality and continuity while facilitating restoration, and ii) how to aggregate the distinct sequences unfolded in totally different ways. To address these problems, we propose a novel Mamba-based Image Restoration model (MaIR), which consists of Nested S-shaped Scanning strategy (NSS) and Sequence Shuffle Attention block (SSA). Specifically, NSS preserves locality and continuity of the input images through the stripe-based scanning region and the S-shaped scanning path, respectively. SSA aggregates sequences through calculating attention weights within the corresponding channels of different sequences. Thanks to NSS and SSA, MaIR surpasses 40 baselines across 14 challenging datasets, achieving state-of-the-art performance on the tasks of image super-resolution, denoising, deblurring and dehazing. The code is available at https://github.com/XLearning-SCU/2025-CVPR-MaIR.

Accelerating Auto-regressive Text-to-Image Generation with Training-free Speculative Jacobi Decoding

The current large auto-regressive models can generate high-quality, high-resolution images, but these models require hundreds or even thousands of steps of next-token prediction during inference, resulting in substantial time consumption. In existing studies, Jacobi decoding, an iterative parallel decoding algorithm, has been used to accelerate the auto-regressive generation and can be executed without training. However, the Jacobi decoding relies on a deterministic criterion to determine the convergence of iterations. Thus, it works for greedy decoding but is incompatible with sampling-based decoding which is crucial for visual quality and diversity in the current auto-regressive text-to-image generation. In this paper, we propose a training-free probabilistic parallel decoding algorithm, Speculative Jacobi Decoding (SJD), to accelerate auto-regressive text-to-image generation. By introducing a probabilistic convergence criterion, our SJD accelerates the inference of auto-regressive text-to-image generation while maintaining the randomness in sampling-based token decoding and allowing the model to generate diverse images. Specifically, SJD facilitates the model to predict multiple tokens at each step and accepts tokens based on the probabilistic criterion, enabling the model to generate images with fewer steps than the conventional next-token-prediction paradigm. We also investigate the token initialization strategies that leverage the spatial locality of visual data to further improve the acceleration ratio under specific scenarios. We conduct experiments for our proposed SJD on multiple auto-regressive text-to-image generation models, showing the effectiveness of model acceleration without sacrificing the visual quality.

Plug-and-Play Context Feature Reuse for Efficient Masked Generation

Masked generative models (MGMs) have emerged as a powerful framework for image synthesis, combining parallel decoding with strong bidirectional context modeling. However, generating high-quality samples typically requires many iterative decoding steps, resulting in high inference costs. A straightforward way to speed up generation is by decoding more tokens in each step, thereby reducing the total number of steps. However, when many tokens are decoded simultaneously, the model can only estimate the univariate marginal distributions independently, failing to capture the dependency among them. As a result, reducing the number of steps significantly compromises generation fidelity. In this work, we introduce ReCAP (Reused Context-Aware Prediction), a plug-and-play module that accelerates inference in MGMs by constructing low-cost steps via reusing feature embeddings from previously decoded context tokens. ReCAP interleaves standard full evaluations with lightweight steps that cache and reuse context features, substantially reducing computation while preserving the benefits of fine-grained, iterative generation. We demonstrate its effectiveness on top of three representative MGMs (MaskGIT, MAGE, and MAR), including both discrete and continuous token spaces and covering diverse architectural designs. In particular, on ImageNet256 class-conditional generation, ReCAP achieves up to 2.4x faster inference than the base model with minimal performance drop, and consistently delivers better efficiency-fidelity trade-offs under various generation settings.

FreeCustom: Tuning-Free Customized Image Generation for Multi-Concept Composition

Benefiting from large-scale pre-trained text-to-image (T2I) generative models, impressive progress has been achieved in customized image generation, which aims to generate user-specified concepts. Existing approaches have extensively focused on single-concept customization and still encounter challenges when it comes to complex scenarios that involve combining multiple concepts. These approaches often require retraining/fine-tuning using a few images, leading to time-consuming training processes and impeding their swift implementation. Furthermore, the reliance on multiple images to represent a singular concept increases the difficulty of customization. To this end, we propose FreeCustom, a novel tuning-free method to generate customized images of multi-concept composition based on reference concepts, using only one image per concept as input. Specifically, we introduce a new multi-reference self-attention (MRSA) mechanism and a weighted mask strategy that enables the generated image to access and focus more on the reference concepts. In addition, MRSA leverages our key finding that input concepts are better preserved when providing images with context interactions. Experiments show that our method's produced images are consistent with the given concepts and better aligned with the input text. Our method outperforms or performs on par with other training-based methods in terms of multi-concept composition and single-concept customization, but is simpler. Codes can be found at https://github.com/aim-uofa/FreeCustom.

Deep Learning architectures for generalized immunofluorescence based nuclear image segmentation

Separating and labeling each instance of a nucleus (instance-aware segmentation) is the key challenge in segmenting single cell nuclei on fluorescence microscopy images. Deep Neural Networks can learn the implicit transformation of a nuclear image into a probability map indicating the class membership of each pixel (nucleus or background), but the use of post-processing steps to turn the probability map into a labeled object mask is error-prone. This especially accounts for nuclear images of tissue sections and nuclear images across varying tissue preparations. In this work, we aim to evaluate the performance of state-of-the-art deep learning architectures to segment nuclei in fluorescence images of various tissue origins and sample preparation types without post-processing. We compare architectures that operate on pixel to pixel translation and an architecture that operates on object detection and subsequent locally applied segmentation. In addition, we propose a novel strategy to create artificial images to extend the training set. We evaluate the influence of ground truth annotation quality, image scale and segmentation complexity on segmentation performance. Results show that three out of four deep learning architectures (U-Net, U-Net with ResNet34 backbone, Mask R-CNN) can segment fluorescent nuclear images on most of the sample preparation types and tissue origins with satisfactory segmentation performance. Mask R-CNN, an architecture designed to address instance aware segmentation tasks, outperforms other architectures. Equal nuclear mean size, consistent nuclear annotations and the use of artificially generated images result in overall acceptable precision and recall across different tissues and sample preparation types.

ImageFlowNet: Forecasting Multiscale Image-Level Trajectories of Disease Progression with Irregularly-Sampled Longitudinal Medical Images

Advances in medical imaging technologies have enabled the collection of longitudinal images, which involve repeated scanning of the same patients over time, to monitor disease progression. However, predictive modeling of such data remains challenging due to high dimensionality, irregular sampling, and data sparsity. To address these issues, we propose ImageFlowNet, a novel model designed to forecast disease trajectories from initial images while preserving spatial details. ImageFlowNet first learns multiscale joint representation spaces across patients and time points, then optimizes deterministic or stochastic flow fields within these spaces using a position-parameterized neural ODE/SDE framework. The model leverages a UNet architecture to create robust multiscale representations and mitigates data scarcity by combining knowledge from all patients. We provide theoretical insights that support our formulation of ODEs, and motivate our regularizations involving high-level visual features, latent space organization, and trajectory smoothness. We validate ImageFlowNet on three longitudinal medical image datasets depicting progression in geographic atrophy, multiple sclerosis, and glioblastoma, demonstrating its ability to effectively forecast disease progression and outperform existing methods. Our contributions include the development of ImageFlowNet, its theoretical underpinnings, and empirical validation on real-world datasets. The official implementation is available at https://github.com/KrishnaswamyLab/ImageFlowNet.

Phikon-v2, A large and public feature extractor for biomarker prediction

Gathering histopathology slides from over 100 publicly available cohorts, we compile a diverse dataset of 460 million pathology tiles covering more than 30 cancer sites. Using this dataset, we train a large self-supervised vision transformer using DINOv2 and publicly release one iteration of this model for further experimentation, coined Phikon-v2. While trained on publicly available histology slides, Phikon-v2 surpasses our previously released model (Phikon) and performs on par with other histopathology foundation models (FM) trained on proprietary data. Our benchmarks include eight slide-level tasks with results reported on external validation cohorts avoiding any data contamination between pre-training and evaluation datasets. Our downstream training procedure follows a simple yet robust ensembling strategy yielding a +1.75 AUC increase across tasks and models compared to one-shot retraining (p<0.001). We compare Phikon (ViT-B) and Phikon-v2 (ViT-L) against 14 different histology feature extractors, making our evaluation the most comprehensive to date. Our result support evidences that DINOv2 handles joint model and data scaling better than iBOT. Also, we show that recent scaling efforts are overall beneficial to downstream performance in the context of biomarker prediction with GigaPath and H-Optimus-0 (two ViT-g with 1.1B parameters each) standing out. However, the statistical margins between the latest top-performing FMs remain mostly non-significant; some even underperform on specific indications or tasks such as MSI prediction - deposed by a 13x smaller model developed internally. While latest foundation models may exhibit limitations for clinical deployment, they nonetheless offer excellent grounds for the development of more specialized and cost-efficient histology encoders fueling AI-guided diagnostic tools.

Text-Driven Tumor Synthesis

Tumor synthesis can generate examples that AI often misses or over-detects, improving AI performance by training on these challenging cases. However, existing synthesis methods, which are typically unconditional -- generating images from random variables -- or conditioned only by tumor shapes, lack controllability over specific tumor characteristics such as texture, heterogeneity, boundaries, and pathology type. As a result, the generated tumors may be overly similar or duplicates of existing training data, failing to effectively address AI's weaknesses. We propose a new text-driven tumor synthesis approach, termed TextoMorph, that provides textual control over tumor characteristics. This is particularly beneficial for examples that confuse the AI the most, such as early tumor detection (increasing Sensitivity by +8.5%), tumor segmentation for precise radiotherapy (increasing DSC by +6.3%), and classification between benign and malignant tumors (improving Sensitivity by +8.2%). By incorporating text mined from radiology reports into the synthesis process, we increase the variability and controllability of the synthetic tumors to target AI's failure cases more precisely. Moreover, TextoMorph uses contrastive learning across different texts and CT scans, significantly reducing dependence on scarce image-report pairs (only 141 pairs used in this study) by leveraging a large corpus of 34,035 radiology reports. Finally, we have developed rigorous tests to evaluate synthetic tumors, including Text-Driven Visual Turing Test and Radiomics Pattern Analysis, showing that our synthetic tumors is realistic and diverse in texture, heterogeneity, boundaries, and pathology.

Towards Accurate Image Coding: Improved Autoregressive Image Generation with Dynamic Vector Quantization

Existing vector quantization (VQ) based autoregressive models follow a two-stage generation paradigm that first learns a codebook to encode images as discrete codes, and then completes generation based on the learned codebook. However, they encode fixed-size image regions into fixed-length codes and ignore their naturally different information densities, which results in insufficiency in important regions and redundancy in unimportant ones, and finally degrades the generation quality and speed. Moreover, the fixed-length coding leads to an unnatural raster-scan autoregressive generation. To address the problem, we propose a novel two-stage framework: (1) Dynamic-Quantization VAE (DQ-VAE) which encodes image regions into variable-length codes based on their information densities for an accurate and compact code representation. (2) DQ-Transformer which thereby generates images autoregressively from coarse-grained (smooth regions with fewer codes) to fine-grained (details regions with more codes) by modeling the position and content of codes in each granularity alternately, through a novel stacked-transformer architecture and shared-content, non-shared position input layers designs. Comprehensive experiments on various generation tasks validate our superiorities in both effectiveness and efficiency. Code will be released at https://github.com/CrossmodalGroup/DynamicVectorQuantization.

SqueezeSAM: User friendly mobile interactive segmentation

Segment Anything Model (SAM) is a foundation model for interactive segmentation, and it has catalyzed major advances in generative AI, computational photography, and medical imaging. This model takes in an arbitrary user input and provides segmentation masks of the corresponding objects. It is our goal to develop a version of SAM that is appropriate for use in a photography app. The original SAM model has a few challenges in this setting. First, original SAM a 600 million parameter based on ViT-H, and its high computational cost and large model size that are not suitable for todays mobile hardware. We address this by proposing the SqueezeSAM model architecture, which is 50x faster and 100x smaller than SAM. Next, when a user takes a photo on their phone, it might not occur to them to click on the image and get a mask. Our solution is to use salient object detection to generate the first few clicks. This produces an initial segmentation mask that the user can interactively edit. Finally, when a user clicks on an object, they typically expect all related pieces of the object to be segmented. For instance, if a user clicks on a person t-shirt in a photo, they expect the whole person to be segmented, but SAM typically segments just the t-shirt. We address this with a new data augmentation scheme, and the end result is that if the user clicks on a person holding a basketball, the person and the basketball are all segmented together.

MedImageInsight: An Open-Source Embedding Model for General Domain Medical Imaging

In this work, we present MedImageInsight, an open-source medical imaging embedding model. MedImageInsight is trained on medical images with associated text and labels across a diverse collection of domains, including X-Ray, CT, MRI, dermoscopy, OCT, fundus photography, ultrasound, histopathology, and mammography. Rigorous evaluations demonstrate MedImageInsight's ability to achieve state-of-the-art (SOTA) or human expert level performance across classification, image-image search, and fine-tuning tasks. Specifically, on public datasets, MedImageInsight achieves SOTA in CT 3D medical image retrieval, as well as SOTA in disease classification and search for chest X-ray, dermatology, and OCT imaging. Furthermore, MedImageInsight achieves human expert performance in bone age estimation (on both public and partner data), as well as AUC above 0.9 in most other domains. When paired with a text decoder, MedImageInsight achieves near SOTA level single image report findings generation with less than 10\% the parameters of other models. Compared to fine-tuning GPT-4o with only MIMIC-CXR data for the same task, MedImageInsight outperforms in clinical metrics, but underperforms on lexical metrics where GPT-4o sets a new SOTA. Importantly for regulatory purposes, MedImageInsight can generate ROC curves, adjust sensitivity and specificity based on clinical need, and provide evidence-based decision support through image-image search (which can also enable retrieval augmented generation). In an independent clinical evaluation of image-image search in chest X-ray, MedImageInsight outperformed every other publicly available foundation model evaluated by large margins (over 6 points AUC), and significantly outperformed other models in terms of AI fairness (across age and gender). We hope releasing MedImageInsight will help enhance collective progress in medical imaging AI research and development.

Cached Multi-Lora Composition for Multi-Concept Image Generation

Low-Rank Adaptation (LoRA) has emerged as a widely adopted technique in text-to-image models, enabling precise rendering of multiple distinct elements, such as characters and styles, in multi-concept image generation. However, current approaches face significant challenges when composing these LoRAs for multi-concept image generation, resulting in diminished generated image quality. In this paper, we initially investigate the role of LoRAs in the denoising process through the lens of the Fourier frequency domain. Based on the hypothesis that applying multiple LoRAs could lead to "semantic conflicts", we find that certain LoRAs amplify high-frequency features such as edges and textures, whereas others mainly focus on low-frequency elements, including the overall structure and smooth color gradients. Building on these insights, we devise a frequency domain based sequencing strategy to determine the optimal order in which LoRAs should be integrated during inference. This strategy offers a methodical and generalizable solution compared to the naive integration commonly found in existing LoRA fusion techniques. To fully leverage our proposed LoRA order sequence determination method in multi-LoRA composition tasks, we introduce a novel, training-free framework, Cached Multi-LoRA (CMLoRA), designed to efficiently integrate multiple LoRAs while maintaining cohesive image generation. With its flexible backbone for multi-LoRA fusion and a non-uniform caching strategy tailored to individual LoRAs, CMLoRA has the potential to reduce semantic conflicts in LoRA composition and improve computational efficiency. Our experimental evaluations demonstrate that CMLoRA outperforms state-of-the-art training-free LoRA fusion methods by a significant margin -- it achieves an average improvement of 2.19% in CLIPScore, and 11.25% in MLLM win rate compared to LoraHub, LoRA Composite, and LoRA Switch.

ITI-GEN: Inclusive Text-to-Image Generation

Text-to-image generative models often reflect the biases of the training data, leading to unequal representations of underrepresented groups. This study investigates inclusive text-to-image generative models that generate images based on human-written prompts and ensure the resulting images are uniformly distributed across attributes of interest. Unfortunately, directly expressing the desired attributes in the prompt often leads to sub-optimal results due to linguistic ambiguity or model misrepresentation. Hence, this paper proposes a drastically different approach that adheres to the maxim that "a picture is worth a thousand words". We show that, for some attributes, images can represent concepts more expressively than text. For instance, categories of skin tones are typically hard to specify by text but can be easily represented by example images. Building upon these insights, we propose a novel approach, ITI-GEN, that leverages readily available reference images for Inclusive Text-to-Image GENeration. The key idea is learning a set of prompt embeddings to generate images that can effectively represent all desired attribute categories. More importantly, ITI-GEN requires no model fine-tuning, making it computationally efficient to augment existing text-to-image models. Extensive experiments demonstrate that ITI-GEN largely improves over state-of-the-art models to generate inclusive images from a prompt. Project page: https://czhang0528.github.io/iti-gen.

Computation-Efficient Era: A Comprehensive Survey of State Space Models in Medical Image Analysis

Sequence modeling plays a vital role across various domains, with recurrent neural networks being historically the predominant method of performing these tasks. However, the emergence of transformers has altered this paradigm due to their superior performance. Built upon these advances, transformers have conjoined CNNs as two leading foundational models for learning visual representations. However, transformers are hindered by the O(N^2) complexity of their attention mechanisms, while CNNs lack global receptive fields and dynamic weight allocation. State Space Models (SSMs), specifically the \textbf{Mamba} model with selection mechanisms and hardware-aware architecture, have garnered immense interest lately in sequential modeling and visual representation learning, challenging the dominance of transformers by providing infinite context lengths and offering substantial efficiency maintaining linear complexity in the input sequence. Capitalizing on the advances in computer vision, medical imaging has heralded a new epoch with Mamba models. Intending to help researchers navigate the surge, this survey seeks to offer an encyclopedic review of Mamba models in medical imaging. Specifically, we start with a comprehensive theoretical review forming the basis of SSMs, including Mamba architecture and its alternatives for sequence modeling paradigms in this context. Next, we offer a structured classification of Mamba models in the medical field and introduce a diverse categorization scheme based on their application, imaging modalities, and targeted organs. Finally, we summarize key challenges, discuss different future research directions of the SSMs in the medical domain, and propose several directions to fulfill the demands of this field. In addition, we have compiled the studies discussed in this paper along with their open-source implementations on our GitHub repository.

Don't Play Favorites: Minority Guidance for Diffusion Models

We explore the problem of generating minority samples using diffusion models. The minority samples are instances that lie on low-density regions of a data manifold. Generating a sufficient number of such minority instances is important, since they often contain some unique attributes of the data. However, the conventional generation process of the diffusion models mostly yields majority samples (that lie on high-density regions of the manifold) due to their high likelihoods, making themselves ineffective and time-consuming for the minority generating task. In this work, we present a novel framework that can make the generation process of the diffusion models focus on the minority samples. We first highlight that Tweedie's denoising formula yields favorable results for majority samples. The observation motivates us to introduce a metric that describes the uniqueness of a given sample. To address the inherent preference of the diffusion models w.r.t. the majority samples, we further develop minority guidance, a sampling technique that can guide the generation process toward regions with desired likelihood levels. Experiments on benchmark real datasets demonstrate that our minority guidance can greatly improve the capability of generating high-quality minority samples over existing generative samplers. We showcase that the performance benefit of our framework persists even in demanding real-world scenarios such as medical imaging, further underscoring the practical significance of our work. Code is available at https://github.com/soobin-um/minority-guidance.

Negative Token Merging: Image-based Adversarial Feature Guidance

Text-based adversarial guidance using a negative prompt has emerged as a widely adopted approach to push the output features away from undesired concepts. While useful, performing adversarial guidance using text alone can be insufficient to capture complex visual concepts and avoid undesired visual elements like copyrighted characters. In this paper, for the first time we explore an alternate modality in this direction by performing adversarial guidance directly using visual features from a reference image or other images in a batch. In particular, we introduce negative token merging (NegToMe), a simple but effective training-free approach which performs adversarial guidance by selectively pushing apart matching semantic features (between reference and output generation) during the reverse diffusion process. When used w.r.t. other images in the same batch, we observe that NegToMe significantly increases output diversity (racial, gender, visual) without sacrificing output image quality. Similarly, when used w.r.t. a reference copyrighted asset, NegToMe helps reduce visual similarity with copyrighted content by 34.57%. NegToMe is simple to implement using just few-lines of code, uses only marginally higher (<4%) inference times and generalizes to different diffusion architectures like Flux, which do not natively support the use of a separate negative prompt. Code is available at https://negtome.github.io

TokenFlow: Unified Image Tokenizer for Multimodal Understanding and Generation

We present TokenFlow, a novel unified image tokenizer that bridges the long-standing gap between multimodal understanding and generation. Prior research attempt to employ a single reconstruction-targeted Vector Quantization (VQ) encoder for unifying these two tasks. We observe that understanding and generation require fundamentally different granularities of visual information. This leads to a critical trade-off, particularly compromising performance in multimodal understanding tasks. TokenFlow addresses this challenge through an innovative dual-codebook architecture that decouples semantic and pixel-level feature learning while maintaining their alignment via a shared mapping mechanism. This design enables direct access to both high-level semantic representations crucial for understanding tasks and fine-grained visual features essential for generation through shared indices. Our extensive experiments demonstrate TokenFlow's superiority across multiple dimensions. Leveraging TokenFlow, we demonstrate for the first time that discrete visual input can surpass LLaVA-1.5 13B in understanding performance, achieving a 7.2\% average improvement. For image reconstruction, we achieve a strong FID score of 0.63 at 384*384 resolution. Moreover, TokenFlow establishes state-of-the-art performance in autoregressive image generation with a GenEval score of 0.55 at 256*256 resolution, achieving comparable results to SDXL.