Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Med Image Anal ; 99: 103344, 2024 Sep 11.
Artigo em Inglês | MEDLINE | ID: mdl-39265361

RESUMO

Significant diagnostic variability between and within observers persists in pathology, despite the fact that digital slide images provide the ability to measure and quantify features much more precisely compared to conventional methods. Automated and accurate segmentation of cancerous cell and tissue regions can streamline the diagnostic process, providing insights into the cancer progression, and helping experts decide on the most effective treatment. Here, we evaluate the performance of the proposed PathoSeg model, with an architecture comprising of a modified HRNet encoder and a UNet++ decoder integrated with a CBAM block to utilize attention mechanism for an improved segmentation capability. We demonstrate that PathoSeg outperforms the current state-of-the-art (SOTA) networks in both quantitative and qualitative assessment of instance and semantic segmentation. Notably, we leverage the use of synthetic data generated by PathopixGAN, which effectively addresses the data imbalance problem commonly encountered in histopathology datasets, further improving the performance of PathoSeg. It utilizes spatially adaptive normalization within a generative and discriminative mechanism to synthesize diverse histopathological environments dictated through semantic information passed through pixel-level annotated Ground Truth semantic masks.Besides, we contribute to the research community by providing an in-house dataset that includes semantically segmented masks for breast carcinoma tubules (BCT), micro/macrovesicular steatosis of the liver (MSL), and prostate carcinoma glands (PCG). In the first part of the dataset, we have a total of 14 whole slide images from 13 patients' liver, with fat cell segmented masks, totaling 951 masks of size 512 × 512 pixels. In the second part, it includes 17 whole slide images from 13 patients with prostate carcinoma gland segmentation masks, amounting to 30,000 masks of size 512 × 512 pixels. In the third part, the dataset contains 51 whole slides from 36 patients, with breast carcinoma tubule masks totaling 30,000 masks of size 512 × 512 pixels. To ensure transparency and encourage further research, we will make this dataset publicly available for non-commercial and academic purposes. To facilitate reproducibility and encourage further research, we will also make our code and pre-trained models publicly available at https://github.com/DeepMIALab/PathoSeg.

2.
Cell ; 187(10): 2502-2520.e17, 2024 May 09.
Artigo em Inglês | MEDLINE | ID: mdl-38729110

RESUMO

Human tissue, which is inherently three-dimensional (3D), is traditionally examined through standard-of-care histopathology as limited two-dimensional (2D) cross-sections that can insufficiently represent the tissue due to sampling bias. To holistically characterize histomorphology, 3D imaging modalities have been developed, but clinical translation is hampered by complex manual evaluation and lack of computational platforms to distill clinical insights from large, high-resolution datasets. We present TriPath, a deep-learning platform for processing tissue volumes and efficiently predicting clinical outcomes based on 3D morphological features. Recurrence risk-stratification models were trained on prostate cancer specimens imaged with open-top light-sheet microscopy or microcomputed tomography. By comprehensively capturing 3D morphologies, 3D volume-based prognostication achieves superior performance to traditional 2D slice-based approaches, including clinical/histopathological baselines from six certified genitourinary pathologists. Incorporating greater tissue volume improves prognostic performance and mitigates risk prediction variability from sampling bias, further emphasizing the value of capturing larger extents of heterogeneous morphology.


Assuntos
Imageamento Tridimensional , Neoplasias da Próstata , Aprendizado de Máquina Supervisionado , Humanos , Masculino , Aprendizado Profundo , Imageamento Tridimensional/métodos , Prognóstico , Neoplasias da Próstata/patologia , Neoplasias da Próstata/diagnóstico por imagem , Microtomografia por Raio-X/métodos
3.
Nat Med ; 30(4): 1174-1190, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38641744

RESUMO

Despite increasing numbers of regulatory approvals, deep learning-based computational pathology systems often overlook the impact of demographic factors on performance, potentially leading to biases. This concern is all the more important as computational pathology has leveraged large public datasets that underrepresent certain demographic groups. Using publicly available data from The Cancer Genome Atlas and the EBRAINS brain tumor atlas, as well as internal patient data, we show that whole-slide image classification models display marked performance disparities across different demographic groups when used to subtype breast and lung carcinomas and to predict IDH1 mutations in gliomas. For example, when using common modeling approaches, we observed performance gaps (in area under the receiver operating characteristic curve) between white and Black patients of 3.0% for breast cancer subtyping, 10.9% for lung cancer subtyping and 16.0% for IDH1 mutation prediction in gliomas. We found that richer feature representations obtained from self-supervised vision foundation models reduce performance variations between groups. These representations provide improvements upon weaker models even when those weaker models are combined with state-of-the-art bias mitigation strategies and modeling choices. Nevertheless, self-supervised vision foundation models do not fully eliminate these discrepancies, highlighting the continuing need for bias mitigation efforts in computational pathology. Finally, we demonstrate that our results extend to other demographic factors beyond patient race. Given these findings, we encourage regulatory and policy agencies to integrate demographic-stratified evaluation into their assessment guidelines.


Assuntos
Glioma , Neoplasias Pulmonares , Humanos , Viés , Negro ou Afro-Americano , População Negra , Demografia , Erros de Diagnóstico , Glioma/diagnóstico , Glioma/genética , Brancos
4.
Nat Med ; 30(3): 850-862, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38504018

RESUMO

Quantitative evaluation of tissue images is crucial for computational pathology (CPath) tasks, requiring the objective characterization of histopathological entities from whole-slide images (WSIs). The high resolution of WSIs and the variability of morphological features present significant challenges, complicating the large-scale annotation of data for high-performance applications. To address this challenge, current efforts have proposed the use of pretrained image encoders through transfer learning from natural image datasets or self-supervised learning on publicly available histopathology datasets, but have not been extensively developed and evaluated across diverse tissue types at scale. We introduce UNI, a general-purpose self-supervised model for pathology, pretrained using more than 100 million images from over 100,000 diagnostic H&E-stained WSIs (>77 TB of data) across 20 major tissue types. The model was evaluated on 34 representative CPath tasks of varying diagnostic difficulty. In addition to outperforming previous state-of-the-art models, we demonstrate new modeling capabilities in CPath such as resolution-agnostic tissue classification, slide classification using few-shot class prototypes, and disease subtyping generalization in classifying up to 108 cancer types in the OncoTree classification system. UNI advances unsupervised representation learning at scale in CPath in terms of both pretraining data and downstream evaluation, enabling data-efficient artificial intelligence models that can generalize and transfer to a wide range of diagnostically challenging tasks and clinical workflows in anatomic pathology.


Assuntos
Inteligência Artificial , Fluxo de Trabalho
5.
ArXiv ; 2023 Aug 29.
Artigo em Inglês | MEDLINE | ID: mdl-37693180

RESUMO

Tissue phenotyping is a fundamental computational pathology (CPath) task in learning objective characterizations of histopathologic biomarkers in anatomic pathology. However, whole-slide imaging (WSI) poses a complex computer vision problem in which the large-scale image resolutions of WSIs and the enormous diversity of morphological phenotypes preclude large-scale data annotation. Current efforts have proposed using pretrained image encoders with either transfer learning from natural image datasets or self-supervised pretraining on publicly-available histopathology datasets, but have not been extensively developed and evaluated across diverse tissue types at scale. We introduce UNI, a general-purpose self-supervised model for pathology, pretrained using over 100 million tissue patches from over 100,000 diagnostic haematoxylin and eosin-stained WSIs across 20 major tissue types, and evaluated on 33 representative CPath clinical tasks in CPath of varying diagnostic difficulties. In addition to outperforming previous state-of-the-art models, we demonstrate new modeling capabilities in CPath such as resolution-agnostic tissue classification, slide classification using few-shot class prototypes, and disease subtyping generalization in classifying up to 108 cancer types in the OncoTree code classification system. UNI advances unsupervised representation learning at scale in CPath in terms of both pretraining data and downstream evaluation, enabling data-efficient AI models that can generalize and transfer to a gamut of diagnostically-challenging tasks and clinical workflows in anatomic pathology.

6.
ArXiv ; 2023 Jul 27.
Artigo em Inglês | MEDLINE | ID: mdl-37547660

RESUMO

Human tissue consists of complex structures that display a diversity of morphologies, forming a tissue microenvironment that is, by nature, three-dimensional (3D). However, the current standard-of-care involves slicing 3D tissue specimens into two-dimensional (2D) sections and selecting a few for microscopic evaluation1,2, with concomitant risks of sampling bias and misdiagnosis3-6. To this end, there have been intense efforts to capture 3D tissue morphology and transition to 3D pathology, with the development of multiple high-resolution 3D imaging modalities7-18. However, these tools have had little translation to clinical practice as manual evaluation of such large data by pathologists is impractical and there is a lack of computational platforms that can efficiently process the 3D images and provide patient-level clinical insights. Here we present Modality-Agnostic Multiple instance learning for volumetric Block Analysis (MAMBA), a deep-learning-based platform for processing 3D tissue images from diverse imaging modalities and predicting patient outcomes. Archived prostate cancer specimens were imaged with open-top light-sheet microscopy12-14 or microcomputed tomography15,16 and the resulting 3D datasets were used to train risk-stratification networks based on 5-year biochemical recurrence outcomes via MAMBA. With the 3D block-based approach, MAMBA achieves an area under the receiver operating characteristic curve (AUC) of 0.86 and 0.74, superior to 2D traditional single-slice-based prognostication (AUC of 0.79 and 0.57), suggesting superior prognostication with 3D morphological features. Further analyses reveal that the incorporation of greater tissue volume improves prognostic performance and mitigates risk prediction variability from sampling bias, suggesting that there is value in capturing larger extents of spatially heterogeneous 3D morphology. With the rapid growth and adoption of 3D spatial biology and pathology techniques by researchers and clinicians, MAMBA provides a general and efficient framework for 3D weakly supervised learning for clinical decision support and can help to reveal novel 3D morphological biomarkers for prognosis and therapeutic response.

7.
Nat Biomed Eng ; 6(12): 1407-1419, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36564629

RESUMO

Histological artefacts in cryosectioned tissue can hinder rapid diagnostic assessments during surgery. Formalin-fixed and paraffin-embedded (FFPE) tissue provides higher quality slides, but the process for obtaining them is laborious (typically lasting 12-48 h) and hence unsuitable for intra-operative use. Here we report the development and performance of a deep-learning model that improves the quality of cryosectioned whole-slide images by transforming them into the style of whole-slide FFPE tissue within minutes. The model consists of a generative adversarial network incorporating an attention mechanism that rectifies cryosection artefacts and a self-regularization constraint between the cryosectioned and FFPE images for the preservation of clinically relevant features. Transformed FFPE-style images of gliomas and of non-small-cell lung cancers from a dataset independent from that used to train the model improved the rates of accurate tumour subtyping by pathologists.


Assuntos
Carcinoma Pulmonar de Células não Pequenas , Aprendizado Profundo , Neoplasias Pulmonares , Humanos , Formaldeído , Inclusão em Parafina/métodos
8.
Nat Biomed Eng ; 6(12): 1420-1434, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36217022

RESUMO

The adoption of digital pathology has enabled the curation of large repositories of gigapixel whole-slide images (WSIs). Computationally identifying WSIs with similar morphologic features within large repositories without requiring supervised training can have significant applications. However, the retrieval speeds of algorithms for searching similar WSIs often scale with the repository size, which limits their clinical and research potential. Here we show that self-supervised deep learning can be leveraged to search for and retrieve WSIs at speeds that are independent of repository size. The algorithm, which we named SISH (for self-supervised image search for histology) and provide as an open-source package, requires only slide-level annotations for training, encodes WSIs into meaningful discrete latent representations and leverages a tree data structure for fast searching followed by an uncertainty-based ranking algorithm for WSI retrieval. We evaluated SISH on multiple tasks (including retrieval tasks based on tissue-patch queries) and on datasets spanning over 22,000 patient cases and 56 disease subtypes. SISH can also be used to aid the diagnosis of rare cancer types for which the number of available WSIs is often insufficient to train supervised deep-learning models.


Assuntos
Aprendizado Profundo , Humanos , Algoritmos , Técnicas Histológicas
9.
Cancer Cell ; 40(10): 1095-1110, 2022 10 10.
Artigo em Inglês | MEDLINE | ID: mdl-36220072

RESUMO

In oncology, the patient state is characterized by a whole spectrum of modalities, ranging from radiology, histology, and genomics to electronic health records. Current artificial intelligence (AI) models operate mainly in the realm of a single modality, neglecting the broader clinical context, which inevitably diminishes their potential. Integration of different data modalities provides opportunities to increase robustness and accuracy of diagnostic and prognostic models, bringing AI closer to clinical practice. AI models are also capable of discovering novel patterns within and across modalities suitable for explaining differences in patient outcomes or treatment resistance. The insights gleaned from such models can guide exploration studies and contribute to the discovery of novel biomarkers and therapeutic targets. To support these advances, here we present a synopsis of AI methods and strategies for multimodal data fusion and association discovery. We outline approaches for AI interpretability and directions for AI-driven exploration through multimodal data interconnections. We examine challenges in clinical adoption and discuss emerging solutions.


Assuntos
Inteligência Artificial , Radiologia , Registros Eletrônicos de Saúde , Genômica , Humanos , Oncologia
10.
Cancer Cell ; 40(8): 865-878.e6, 2022 08 08.
Artigo em Inglês | MEDLINE | ID: mdl-35944502

RESUMO

The rapidly emerging field of computational pathology has demonstrated promise in developing objective prognostic models from histology images. However, most prognostic models are either based on histology or genomics alone and do not address how these data sources can be integrated to develop joint image-omic prognostic models. Additionally, identifying explainable morphological and molecular descriptors from these models that govern such prognosis is of interest. We use multimodal deep learning to jointly examine pathology whole-slide images and molecular profile data from 14 cancer types. Our weakly supervised, multimodal deep-learning algorithm is able to fuse these heterogeneous modalities to predict outcomes and discover prognostic features that correlate with poor and favorable outcomes. We present all analyses for morphological and molecular correlates of patient prognosis across the 14 cancer types at both a disease and a patient level in an interactive open-access database to allow for further exploration, biomarker discovery, and feature assessment.


Assuntos
Aprendizado Profundo , Neoplasias , Algoritmos , Genômica/métodos , Humanos , Neoplasias/genética , Neoplasias/patologia , Prognóstico
11.
Cancer Res ; 82(15): 2672-2673, 2022 08 03.
Artigo em Inglês | MEDLINE | ID: mdl-35919991

RESUMO

Despite the crucial role of phenotypic and genetic intratumoral heterogeneity in understanding and predicting clinical outcomes for patients with cancer, computational pathology studies have yet to make substantial steps in this area. The major limiting factor has been the bulk gene-sequencing practice that results in loss of spatial information of gene status, making the study of intratumoral heterogeneity difficult. In this issue of Cancer Research, Acosta and colleagues used deep learning to study if localized gene mutation status can be predicted from localized tumor morphology for clear cell renal cell carcinoma. The algorithm was developed using curated sets of matched hematoxylin and eosin and IHC images, which represent spatially resolved morphology and genotype, respectively. This study confirms the existence of a strong link between morphology and underlying genetics on a regional level, paving the way for further investigations into intratumoral heterogeneity. See related article by Acosta et al., p. 2792.


Assuntos
Aprendizado Profundo , Neoplasias Renais , Humanos , Neoplasias Renais/genética , Mutação
12.
PLoS One ; 17(2): e0264201, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35202431

RESUMO

Activating mutations in EGFR predict benefit from tyrosine kinase inhibitor therapy for patients with advanced non-small cell lung cancer. Directing patients to appropriate therapy depends on accurate and timely EGFR assessment in the molecular pathology laboratory. This article describes the analytical design, performance characteristics, and clinical implementation of an assay for the rapid detection of EGFR L858R and exon 19 deletion mutations. A droplet digital polymerase chain reaction (ddPCR) assay was implemented with probe hydrolysis-dependent signal detection. A mutation-specific probe was used to detect EGFR L858R. A loss of signal design was used to detect EGFR exon 19 deletion mutations. Analytical sensitivity was dependent on DNA input and was as low as 0.01% variant allele fraction for the EGFR L858R assay and 0.1% variant allele fraction for the EGFR exon 19 deletion assay. Correlation of 20 clinical specimens tested by ddPCR and next generation sequencing showed 100% concordance. ddPCR showed 53% clinical sensitivity in the detection of EGFR mutations in plasma cell-free DNA from patients with lung cancer. The median clinical turnaround time was 5 days for ddPCR compared to 13 days for next generation sequencing. The findings show that ddPCR is an accurate and rapid method for detecting EGFR mutations in patients with non-small cell lung cancer.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/genética , Análise Mutacional de DNA/métodos , Neoplasias Pulmonares/genética , Reação em Cadeia da Polimerase/métodos , Receptores ErbB/genética , Humanos , Mutação , Sensibilidade e Especificidade
13.
IEEE Trans Med Imaging ; 41(4): 757-770, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-32881682

RESUMO

Cancer diagnosis, prognosis, mymargin and therapeutic response predictions are based on morphological information from histology slides and molecular profiles from genomic data. However, most deep learning-based objective outcome prediction and grading paradigms are based on histology or genomics alone and do not make use of the complementary information in an intuitive manner. In this work, we propose Pathomic Fusion, an interpretable strategy for end-to-end multimodal fusion of histology image and genomic (mutations, CNV, RNA-Seq) features for survival outcome prediction. Our approach models pairwise feature interactions across modalities by taking the Kronecker product of unimodal feature representations, and controls the expressiveness of each representation via a gating-based attention mechanism. Following supervised learning, we are able to interpret and saliently localize features across each modality, and understand how feature importance shifts when conditioning on multimodal input. We validate our approach using glioma and clear cell renal cell carcinoma datasets from the Cancer Genome Atlas (TCGA), which contains paired whole-slide image, genotype, and transcriptome data with ground truth survival and histologic grade labels. In a 15-fold cross-validation, our results demonstrate that the proposed multimodal fusion paradigm improves prognostic determinations from ground truth grading and molecular subtyping, as well as unimodal deep networks trained on histology and genomic data alone. The proposed method establishes insight and theory on how to train deep networks on multimodal biomedical data in an intuitive manner, which will be useful for other problems in medicine that seek to combine heterogeneous data streams for understanding diseases and predicting response and resistance to treatment. Code and trained models are made available at: https://github.com/mahmoodlab/PathomicFusion.


Assuntos
Genômica , Glioma , Genômica/métodos , Glioma/patologia , Técnicas Histológicas , Humanos , Prognóstico
15.
Nature ; 594(7861): 106-110, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33953404

RESUMO

Cancer of unknown primary (CUP) origin is an enigmatic group of diagnoses in which the primary anatomical site of tumour origin cannot be determined1,2. This poses a considerable challenge, as modern therapeutics are predominantly specific to the primary tumour3. Recent research has focused on using genomics and transcriptomics to identify the origin of a tumour4-9. However, genomic testing is not always performed and lacks clinical penetration in low-resource settings. Here, to overcome these challenges, we present a deep-learning-based algorithm-Tumour Origin Assessment via Deep Learning (TOAD)-that can provide a differential diagnosis for the origin of the primary tumour using routinely acquired histology slides. We used whole-slide images of tumours with known primary origins to train a model that simultaneously identifies the tumour as primary or metastatic and predicts its site of origin. On our held-out test set of tumours with known primary origins, the model achieved a top-1 accuracy of 0.83 and a top-3 accuracy of 0.96, whereas on our external test set it achieved top-1 and top-3 accuracies of 0.80 and 0.93, respectively. We further curated a dataset of 317 cases of CUP for which a differential diagnosis was assigned. Our model predictions resulted in concordance for 61% of cases and a top-3 agreement of 82%. TOAD can be used as an assistive tool to assign a differential diagnosis to complicated cases of metastatic tumours and CUPs and could be used in conjunction with or in lieu of ancillary tests and extensive diagnostic work-ups to reduce the occurrence of CUP.


Assuntos
Inteligência Artificial , Simulação por Computador , Neoplasias Primárias Desconhecidas/patologia , Estudos de Coortes , Simulação por Computador/normas , Feminino , Humanos , Masculino , Metástase Neoplásica/patologia , Neoplasias Primárias Desconhecidas/diagnóstico , Reprodutibilidade dos Testes , Sensibilidade e Especificidade , Fluxo de Trabalho
16.
Histopathology ; 79(4): 491-498, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-33783022

RESUMO

BACKGROUND AND AIMS: Columnar cell variant (CCV) is a rare papillary thyroid carcinoma subtype. The majority of CCV occur in older patients and are large, invasive tumours that pursue an aggressive clinical course. Rare well-circumscribed CCV occur in younger female patients and are comparatively indolent. METHODS AND RESULTS: We retrospectively identified CCV with material available to perform targeted next-generation sequencing and correlated molecular results with clinicopathological features and outcome. Our cohort was comprised of nine CCV. Nearly all were aggressive tumours; however, one was predominantly well-circumscribed and arose in a thyroglossal duct cyst of a 26-year-old woman who had no evidence of disease at last follow-up. Seven (78%) cases demonstrated activating oncogenic driver alterations in BRAF, including BRAF V600E, an activating N486_P490del deletion, and BRAF-AGK fusions. Activating RAS mutations were seen in two (22%) cases. Additionally, three (33%) cases had TERT promoter mutations, four (44%) had loss of the tumour suppressor CDKN2A and one (11%) case had a loss of function TP53 mutation. Most cases (89%) also demonstrated copy number alterations, including recurrent gain of chromosome 1q (five cases) and losses of chromosome 9p (three cases) and 22q (four cases). The one case without secondary pathogenic mutations or copy number alterations was the tumour in the 26-year-old woman. CONCLUSIONS: We found that CCV is primarily a BRAF-driven tumour, with most also harbouring secondary oncogenic mutations and multiple chromosomal gains and losses. Moreover, our findings suggest that molecular analysis could potentially be used to help risk stratify CCV.


Assuntos
Câncer Papilífero da Tireoide/genética , Neoplasias da Glândula Tireoide/genética , Adulto , Idoso , Idoso de 80 Anos ou mais , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Estudos Retrospectivos , Transcriptoma
17.
Nat Biomed Eng ; 5(6): 555-570, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33649564

RESUMO

Deep-learning methods for computational pathology require either manual annotation of gigapixel whole-slide images (WSIs) or large datasets of WSIs with slide-level labels and typically suffer from poor domain adaptation and interpretability. Here we report an interpretable weakly supervised deep-learning method for data-efficient WSI processing and learning that only requires slide-level labels. The method, which we named clustering-constrained-attention multiple-instance learning (CLAM), uses attention-based learning to identify subregions of high diagnostic value to accurately classify whole slides and instance-level clustering over the identified representative regions to constrain and refine the feature space. By applying CLAM to the subtyping of renal cell carcinoma and non-small-cell lung cancer as well as the detection of lymph node metastasis, we show that it can be used to localize well-known morphological features on WSIs without the need for spatial labels, that it overperforms standard weakly supervised classification algorithms and that it is adaptable to independent test cohorts, smartphone microscopy and varying tissue content.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/diagnóstico por imagem , Carcinoma de Células Renais/diagnóstico por imagem , Aprendizado Profundo , Interpretação de Imagem Assistida por Computador/estatística & dados numéricos , Neoplasias Renais/diagnóstico por imagem , Neoplasias Pulmonares/diagnóstico por imagem , Área Sob a Curva , Carcinoma Pulmonar de Células não Pequenas/patologia , Carcinoma de Células Renais/patologia , Histocitoquímica/métodos , Histocitoquímica/estatística & dados numéricos , Humanos , Neoplasias Renais/patologia , Neoplasias Pulmonares/patologia , Metástase Linfática , Microscopia/métodos , Microscopia/estatística & dados numéricos , Smartphone
18.
JCO Clin Cancer Inform ; 3: 1-12, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30742484

RESUMO

PURPOSE: In an upcoming clinical trial at the Moffitt Cancer Center for women with stage 2/3 estrogen receptor-positive breast cancer, treatment with an aromatase inhibitor and a PD-L1 checkpoint inhibitor combination will be investigated to lower a preoperative endocrine prognostic index (PEPI) that correlates with relapse-free survival. PEPI is fundamentally a static index, measured at the end of neoadjuvant therapy before surgery. We have developed a mathematical model of the essential components of the PEPI score to identify successful combination therapy regimens that minimize tumor burden and metastatic potential, on the basis of time-dependent trade-offs in the system. METHODS: We considered two molecular traits, CCR7 and PD-L1, which correlate with treatment response and increased metastatic risk. We used a matrix game model with the four phenotypic strategies to examine the frequency-dependent interactions of cancer cells. This game was embedded in an ecological model of tumor population-growth dynamics. The resulting model predicts evolutionary and ecological dynamics that track with changes in the PEPI score. RESULTS: We considered various treatment regimens on the basis of combinations of the two therapies with drug holidays. By considering the trade off between tumor burden and metastatic potential, the optimal therapy plan was a 1-month kick start of the immune checkpoint inhibitor followed by 5 months of continuous combination therapy. Relative to a protocol giving both therapeutics together from the start, this delayed regimen resulted in transient suboptimal tumor regression while maintaining a phenotypic constitution that is more amenable to fast tumor regression for the final 5 months of therapy. CONCLUSION: The mathematical model provides a useful abstraction of clinical intuition, enabling hypothesis generation and testing of clinical assumptions.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/imunologia , Teoria dos Jogos , Imunoterapia/métodos , Inibidores da Aromatase/administração & dosagem , Antígeno B7-H1/antagonistas & inibidores , Biomarcadores Tumorais/metabolismo , Neoplasias da Mama/patologia , Feminino , Humanos , Imunoterapia/normas , Terapia Neoadjuvante , Receptores CCR7/antagonistas & inibidores
19.
J Open Source Softw ; 3(28)2018 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-33381678

RESUMO

High-dimensional datasets are becoming more common in a variety of scientific fields. Well-known examples include next-generation sequencing in biology, patient health status in medicine, and computer vision in deep learning. Dimension reduction, using methods like principal component analysis (PCA), is a common preprocessing step for such datasets. However, while dimension reduction can save computing and human resources, it comes with the cost of significant information loss. Topological data analysis (TDA) aims to analyze the "shape" of high-dimensional datasets, without dimension reduction, by extracting features that are robust to small perturbations in data. Persistent features of a dataset can be used to describe it, and to compare it to other datasets. Visualization of persistent features can be done using topological barcodes or persistence diagrams (Figure 1). Application of TDA methods has granted greater insight into high-dimensional data (Lakshmikanth et al., 2017); one prominent example of this is its use to characterize a clinically relevant subgroup of breast cancer patients (Nicolau, Levine, & Carlsson, 2011). This is a particularly salient study as Nicolau et al. (2011) used a topological method, termed Progression Analysis of Disease, to identify a patient subgroup with 100% survival using that remains invisible to other clustering methods.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA