Dale Webster

Dale Webster

Dale Webster is Director of Research at Google Health working to improve patient outcomes in healthcare using Deep Learning and Medical Imaging. His recent work leverages AI to screen for Diabetic Retinopathy in India and Thailand, predict Cardiovascular health factors from fundus photos, differential diagnosis of skin disease, and applications of medically tuned LLMs. Prior to Google he was a Software Engineer at Pacific Biosciences working on direct sequencing of methylation state and rapid sequencing and assembly of microbial pathogens during global outbreaks. His PhD work in Bioinformatics at the University of California San Francisco focused on viral evolution, and he received his Bachelor of Science in Computer Science from Rice University.
Authored Publications
Sort By
  • Title
  • Title, descending
  • Year
  • Year, descending
    Preview abstract Background Health datasets from clinical sources do not reflect the breadth and diversity of disease in the real world, impacting research, medical education and artificial intelligence (AI) tool development. Dermatology is a suitable area to develop and test a new and scalable method to create representative health datasets. Methods We used Google Search advertisements to solicit contributions of images of dermatology conditions, demographic and symptom information from internet users in the United States (US) over 265 days starting March 2023. With informed contributor consent, we described and released this dataset containing 10,106 images from 5058 contributions, with dermatologist labels as well as Fitzpatrick Skin Type and Monk Skin Tone labels for the images. Results We received 22 ± 14 submissions/day over 265 days. Female contributors (66.04%) and younger individuals (52.3% < age 40) had a higher representation in the dataset compared to the US population, and 36.6% of contributors had a non-White racial or ethnic identity. Over 97.5% of contributions were genuine images of skin conditions. Image quality had no impact on dermatologist confidence in assigning a differential diagnosis. The dataset consists largely of short duration (54% with onset < 7 days ago) allergic, infectious, and inflammatory conditions. Fitzpatrick skin type distribution is well-balanced, considering the geographical origin of the dataset and the absence of enrichment for population groups or skin tones. Interpretation Search ads are effective at crowdsourcing images of health conditions. The SCIN dataset bridges important gaps in the availability of representative images of common skin conditions. View details
    Differences between Patient and Clinician Submitted Images: Implications for Virtual Care of Skin Conditions
    Rajeev Rikhye
    Grace Eunhae Hong
    Margaret Ann Smith
    Aaron Loh
    Vijaytha Muralidharan
    Doris Wong
    Michelle Phung
    Nicolas Betancourt
    Bradley Fong
    Rachna Sahasrabudhe
    Khoban Nasim
    Alec Eschholz
    Kat Chou
    Peggy Bui
    Justin Ko
    Steven Lin
    Mayo Clinic Proceedings: Digital Health(2024)
    Preview abstract Objective: To understand and highlight the differences in clinical, demographic, and image quality characteristics between patient-taken (PAT) and clinic-taken (CLIN) photographs of skin conditions. Patients and Methods: This retrospective study applied logistic regression to data from 2500 deidentified cases in Stanford Health Care’s eConsult system, from November 2015 to January 2021. Cases with undiagnosable or multiple conditions or cases with both patient and clinician image sources were excluded, leaving 628 PAT cases and 1719 CLIN cases. Demographic characteristic factors, such as age and sex were self-reported, whereas anatomic location, estimated skin type, clinical signs and symptoms, condition duration, and condition frequency were summarized from patient health records. Image quality variables such as blur, lighting issues and whether the image contained skin, hair, or nails were estimated through a deep learning model. Results: Factors that were positively associated with CLIN photographs, post-2020 were as follows: age 60 years or older, darker skin types (eFST V/VI), and presence of skin growths. By contrast, factors that were positively associated with PAT photographs include conditions appearing intermittently, cases with blurry photographs, photographs with substantial nonskin (or nail/hair) regions and cases with more than 3 photographs. Within the PAT cohort, older age was associated with blurry photographs. Conclusion: There are various demographic, clinical, and image quality characteristic differences between PAT and CLIN photographs of skin concerns. The demographic characteristic differences present important considerations for improving digital literacy or access, whereas the image quality differences point to the need for improved patient education and better image capture workflows, particularly among elderly patients. View details
    Conversational AI in health: Design considerations from a Wizard-of-Oz dermatology case study with users, clinicians and a medical LLM
    Brenna Li
    Amy Wang
    Patricia Strachan
    Julie Anne Seguin
    Sami Lachgar
    Karyn Schroeder
    Renee Wong
    Extended Abstracts of the 2024 CHI Conference on Human Factors in Computing Systems, Association for Computing Machinery, pp. 10
    Preview abstract Although skin concerns are common, access to specialist care is limited. Artificial intelligence (AI)-assisted tools to support medical decisions may provide patients with feedback on their concerns while also helping ensure the most urgent cases are routed to dermatologists. Although AI-based conversational agents have been explored recently, how they are perceived by patients and clinicians is not well understood. We conducted a Wizard-of-Oz study involving 18 participants with real skin concerns. Participants were randomly assigned to interact with either a clinician agent (portrayed by a dermatologist) or an LLM agent (supervised by a dermatologist) via synchronous multimodal chat. In both conditions, participants found the conversation to be helpful in understanding their medical situation and alleviate their concerns. Through qualitative coding of the conversation transcripts, we provide insight on the importance of empathy and effective information-seeking. We conclude with design considerations for future AI-based conversational agents in healthcare settings. View details
    Preview abstract Advances in machine learning for health care have brought concerns about bias from the research community; specifically, the introduction, perpetuation, or exacerbation of care disparities. Reinforcing these concerns is the finding that medical images often reveal signals about sensitive attributes in ways that are hard to pinpoint by both algorithms and people. This finding raises a question about how to best design general purpose pretrained embeddings (GPPEs, defined as embeddings meant to support a broad array of use cases) for building downstream models that are free from particular types of bias. The downstream model should be carefully evaluated for bias, and audited and improved as appropriate. However, in our view, well intentioned attempts to prevent the upstream components—GPPEs—from learning sensitive attributes can have unintended consequences on the downstream models. Despite producing a veneer of technical neutrality, the resultant end-to-end system might still be biased or poorly performing. We present reasons, by building on previously published data, to support the reasoning that GPPEs should ideally contain as much information as the original data contain, and highlight the perils of trying to remove sensitive attributes from a GPPE. We also emphasise that downstream prediction models trained for specific tasks and settings, whether developed using GPPEs or not, should be carefully designed and evaluated to avoid bias that makes models vulnerable to issues such as distributional shift. These evaluations should be done by a diverse team, including social scientists, on a diverse cohort representing the full breadth of the patient population for which the final model is intended. View details
    Towards Generalist Biomedical AI
    Danny Driess
    Andrew Carroll
    Chuck Lau
    Ryutaro Tanno
    Ira Ktena
    Anil Palepu
    Basil Mustafa
    Aakanksha Chowdhery
    Simon Kornblith
    Philip Mansfield
    Sushant Prakash
    Renee Wong
    Sunny Virmani
    Sara Mahdavi
    Bradley Green
    Ewa Dominowska
    Joelle Barral
    Karan Singhal
    Pete Florence
    NEJM AI(2024)
    Preview abstract BACKGROUND: Medicine is inherently multimodal, requiring the simultaneous interpretation and integration of insights between many data modalities spanning text, imaging, genomics, and more. Generalist biomedical artificial intelligence systems that flexibly encode, integrate, and interpret these data might better enable impactful applications ranging from scientific discovery to care delivery. METHODS: To catalyze development of these models, we curated MultiMedBench, a new multimodal biomedical benchmark. MultiMedBench encompasses 14 diverse tasks, such as medical question answering, mammography and dermatology image interpretation, radiology report generation and summarization, and genomic variant calling. We then introduced Med-PaLM Multimodal (Med-PaLM M), our proof of concept for a generalist biomedical AI system that flexibly encodes and interprets biomedical data including clinical language, imaging, and genomics with the same set of model weights. To further probe the capabilities and limitations of Med-PaLM M, we conducted a radiologist evaluation of model-generated (and human) chest x-ray reports. RESULTS: We observed encouraging performance across model scales. Med-PaLM M reached performance competitive with or exceeding the state of the art on all MultiMedBench tasks, often surpassing specialist models by a wide margin. In a side-by-side ranking on 246 retrospective chest x-rays, clinicians expressed a pairwise preference for Med-PaLM Multimodal reports over those produced by radiologists in up to 40.50% of cases, suggesting potential clinical utility. CONCLUSIONS: Although considerable work is needed to validate these models in real-world cases and understand if cross-modality generalization is possible, our results represent a milestone toward the development of generalist biomedical artificial intelligence systems. View details
    Health equity assessment of machine learning performance (HEAL): a framework and dermatology AI model case study
    Terry Spitz
    Malcolm Chelliah
    Heather Cole-Lewis
    Stephanie Farquhar
    Qinghan Xue
    Jenna Lester
    Cían Hughes
    Patricia Strachan
    Fraser Tan
    Peggy Bui
    Craig Mermel
    Lily Peng
    Sunny Virmani
    Ivor Horn
    Cameron Chen
    The Lancet eClinicalMedicine(2024)
    Preview abstract Background Artificial intelligence (AI) has repeatedly been shown to encode historical inequities in healthcare. We aimed to develop a framework to quantitatively assess the performance equity of health AI technologies and to illustrate its utility via a case study. Methods Here, we propose a methodology to assess whether health AI technologies prioritise performance for patient populations experiencing worse outcomes, that is complementary to existing fairness metrics. We developed the Health Equity Assessment of machine Learning performance (HEAL) framework designed to quantitatively assess the performance equity of health AI technologies via a four-step interdisciplinary process to understand and quantify domain-specific criteria, and the resulting HEAL metric. As an illustrative case study (analysis conducted between October 2022 and January 2023), we applied the HEAL framework to a dermatology AI model. A set of 5420 teledermatology cases (store-and-forward cases from patients of 20 years or older, submitted from primary care providers in the USA and skin cancer clinics in Australia), enriched for diversity in age, sex and race/ethnicity, was used to retrospectively evaluate the AI model's HEAL metric, defined as the likelihood that the AI model performs better for subpopulations with worse average health outcomes as compared to others. The likelihood that AI performance was anticorrelated to pre-existing health outcomes was estimated using bootstrap methods as the probability that the negated Spearman's rank correlation coefficient (i.e., “R”) was greater than zero. Positive values of R suggest that subpopulations with poorer health outcomes have better AI model performance. Thus, the HEAL metric, defined as p (R >0), measures how likely the AI technology is to prioritise performance for subpopulations with worse average health outcomes as compared to others (presented as a percentage below). Health outcomes were quantified as disability-adjusted life years (DALYs) when grouping by sex and age, and years of life lost (YLLs) when grouping by race/ethnicity. AI performance was measured as top-3 agreement with the reference diagnosis from a panel of 3 dermatologists per case. Findings Across all dermatologic conditions, the HEAL metric was 80.5% for prioritizing AI performance of racial/ethnic subpopulations based on YLLs, and 92.1% and 0.0% respectively for prioritizing AI performance of sex and age subpopulations based on DALYs. Certain dermatologic conditions were significantly associated with greater AI model performance compared to a reference category of less common conditions. For skin cancer conditions, the HEAL metric was 73.8% for prioritizing AI performance of age subpopulations based on DALYs. Interpretation Analysis using the proposed HEAL framework showed that the dermatology AI model prioritised performance for race/ethnicity, sex (all conditions) and age (cancer conditions) subpopulations with respect to pre-existing health disparities. More work is needed to investigate ways of promoting equitable AI performance across age for non-cancer conditions and to better understand how AI models can contribute towards improving equity in health outcomes. View details
    Large Language Models Encode Clinical Knowledge
    Karan Singhal
    Sara Mahdavi
    Jason Wei
    Hyung Won Chung
    Nathan Scales
    Ajay Tanwani
    Heather Cole-Lewis
    Perry Payne
    Martin Seneviratne
    Paul Gamble
    Abubakr Abdelrazig Hassan Babiker
    Nathanael Schaerli
    Aakanksha Chowdhery
    Philip Mansfield
    Dina Demner-Fushman
    Katherine Chou
    Juraj Gottweis
    Nenad Tomašev
    Alvin Rajkomar
    Joelle Barral
    Nature(2023)
    Preview abstract Large language models (LLMs) have demonstrated impressive capabilities, but the bar for clinical applications is high. Attempts to assess the clinical knowledge of models typically rely on automated evaluations based on limited benchmarks. Here, to address these limitations, we present MultiMedQA, a benchmark combining six existing medical question answering datasets spanning professional medicine, research and consumer queries and a new dataset of medical questions searched online, HealthSearchQA. We propose a human evaluation framework for model answers along multiple axes including factuality, comprehension, reasoning, possible harm and bias. In addition, we evaluate Pathways Language Model (PaLM, a 540-billion parameter LLM) and its instruction-tuned variant, Flan-PaLM on MultiMedQA. Using a combination of prompting strategies, Flan-PaLM achieves state-of-the-art accuracy on every MultiMedQA multiple-choice dataset (MedQA, MedMCQA, PubMedQA and Measuring Massive Multitask Language Understanding (MMLU) clinical topics), including 67.6% accuracy on MedQA (US Medical Licensing Exam-style questions), surpassing the prior state of the art by more than 17%. However, human evaluation reveals key gaps. To resolve this, we introduce instruction prompt tuning, a parameter-efficient approach for aligning LLMs to new domains using a few exemplars. The resulting model, Med-PaLM, performs encouragingly, but remains inferior to clinicians. We show that comprehension, knowledge recall and reasoning improve with model scale and instruction prompt tuning, suggesting the potential utility of LLMs in medicine. Our human evaluations reveal limitations of today’s models, reinforcing the importance of both evaluation frameworks and method development in creating safe, helpful LLMs for clinical applications. View details
    Towards Accurate Differential Diagnosis with Large Language Models
    Daniel McDuff
    Anil Palepu
    Amy Wang
    Karan Singhal
    Yash Sharma
    Kavita Kulkarni
    Le Hou
    Sara Mahdavi
    Sushant Prakash
    Anupam Pathak
    Shwetak Patel
    Ewa Dominowska
    Juro Gottweis
    Joelle Barral
    Kat Chou
    Jake Sunshine
    Arxiv(2023)
    Preview abstract An accurate differential diagnosis (DDx) is a cornerstone of medical care, often reached through an iterative process of interpretation that combines clinical history, physical examination, investigations and procedures. Interactive interfaces powered by Large Language Models (LLMs) present new opportunities to both assist and automate aspects of this process. In this study, we introduce an LLM optimized for diagnostic reasoning, and evaluate its ability to generate a DDx alone or as an aid to clinicians. 20 clinicians evaluated 302 challenging, real-world medical cases sourced from the New England Journal of Medicine (NEJM) case reports. Each case report was read by two clinicians, who were randomized to one of two assistive conditions: either assistance from search engines and standard medical resources, or LLM assistance in addition to these tools. All clinicians provided a baseline, unassisted DDx prior to using the respective assistive tools. Our LLM for DDx exhibited standalone performance that exceeded that of unassisted clinicians (top-10 accuracy 59.1% vs 33.6%, [p = 0.04]). Comparing the two assisted study arms, the DDx quality score was higher for clinicians assisted by our LLM (top-10 accuracy 51.7%) compared to clinicians without its assistance (36.1%) (McNemar's Test: 45.7, p < 0.01) and clinicians with search (44.4%) (4.75, p = 0.03). Further, clinicians assisted by our LLM arrived at more comprehensive differential lists than those without its assistance. Our study suggests that our LLM for DDx has potential to improve clinicians' diagnostic reasoning and accuracy in challenging cases, meriting further real-world evaluation for its ability to empower physicians and widen patients' access to specialist-level expertise. View details
    Preview abstract The application of an artificial intelligence (AI)-based screening tool for retinal disease in India and Thailand highlighted the myths and reality of introducing medical AI, which may form a framework for subsequent tools. View details
    Risk Stratification for Diabetic Retinopathy Screening Order Using Deep Learning: A Multicenter Prospective Study
    Ashish Bora
    Sunny Virmani
    Rayman Huang
    Ilana Traynis
    Lily Peng
    Avinash Varadarajan
    Warisara Pattanapongpaiboon
    Reena Chopra
    Dr. Paisan Raumviboonsuk
    Translational Vision Science & Technology(2023)
    Preview abstract Purpose: Real-world evaluation of a deep learning model that prioritizes patients based on risk of progression to moderate or worse (MOD+) diabetic retinopathy (DR). Methods: This nonrandomized, single-arm, prospective, interventional study included patients attending DR screening at four centers across Thailand from September 2019 to January 2020, with mild or no DR. Fundus photographs were input into the model, and patients were scheduled for their subsequent screening from September 2020 to January 2021 in order of predicted risk. Evaluation focused on model sensitivity, defined as correctly ranking patients that developed MOD+ within the first 50% of subsequent screens. Results: We analyzed 1,757 patients, of which 52 (3.0%) developed MOD+. Using the model-proposed order, the model's sensitivity was 90.4%. Both the model-proposed order and mild/no DR plus HbA1c had significantly higher sensitivity than the random order (P < 0.001). Excluding one major (rural) site that had practical implementation challenges, the remaining sites included 567 patients and 15 (2.6%) developed MOD+. Here, the model-proposed order achieved 86.7% versus 73.3% for the ranking that used DR grade and hemoglobin A1c. Conclusions: The model can help prioritize follow-up visits for the largest subgroups of DR patients (those with no or mild DR). Further research is needed to evaluate the impact on clinical management and outcomes. Translational Relevance: Deep learning demonstrated potential for risk stratification in DR screening. However, real-world practicalities must be resolved to fully realize the benefit. View details