1
|
Zhang Z, Zhou X, Fang Y, Xiong Z, Zhang T. AI-driven 3D bioprinting for regenerative medicine: From bench to bedside. Bioact Mater 2025; 45:201-230. [PMID: 39651398 PMCID: PMC11625302 DOI: 10.1016/j.bioactmat.2024.11.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 11/01/2024] [Accepted: 11/16/2024] [Indexed: 12/11/2024] Open
Abstract
In recent decades, 3D bioprinting has garnered significant research attention due to its ability to manipulate biomaterials and cells to create complex structures precisely. However, due to technological and cost constraints, the clinical translation of 3D bioprinted products (BPPs) from bench to bedside has been hindered by challenges in terms of personalization of design and scaling up of production. Recently, the emerging applications of artificial intelligence (AI) technologies have significantly improved the performance of 3D bioprinting. However, the existing literature remains deficient in a methodological exploration of AI technologies' potential to overcome these challenges in advancing 3D bioprinting toward clinical application. This paper aims to present a systematic methodology for AI-driven 3D bioprinting, structured within the theoretical framework of Quality by Design (QbD). This paper commences by introducing the QbD theory into 3D bioprinting, followed by summarizing the technology roadmap of AI integration in 3D bioprinting, including multi-scale and multi-modal sensing, data-driven design, and in-line process control. This paper further describes specific AI applications in 3D bioprinting's key elements, including bioink formulation, model structure, printing process, and function regulation. Finally, the paper discusses current prospects and challenges associated with AI technologies to further advance the clinical translation of 3D bioprinting.
Collapse
Affiliation(s)
- Zhenrui Zhang
- Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, PR China
- Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Beijing, 100084, PR China
- “Biomanufacturing and Engineering Living Systems” Innovation International Talents Base (111 Base), Beijing, 100084, PR China
| | - Xianhao Zhou
- Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, PR China
- Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Beijing, 100084, PR China
- “Biomanufacturing and Engineering Living Systems” Innovation International Talents Base (111 Base), Beijing, 100084, PR China
| | - Yongcong Fang
- Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, PR China
- Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Beijing, 100084, PR China
- “Biomanufacturing and Engineering Living Systems” Innovation International Talents Base (111 Base), Beijing, 100084, PR China
- State Key Laboratory of Tribology in Advanced Equipment, Tsinghua University, Beijing, 100084, PR China
| | - Zhuo Xiong
- Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, PR China
- Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Beijing, 100084, PR China
- “Biomanufacturing and Engineering Living Systems” Innovation International Talents Base (111 Base), Beijing, 100084, PR China
| | - Ting Zhang
- Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, PR China
- Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Beijing, 100084, PR China
- “Biomanufacturing and Engineering Living Systems” Innovation International Talents Base (111 Base), Beijing, 100084, PR China
- State Key Laboratory of Tribology in Advanced Equipment, Tsinghua University, Beijing, 100084, PR China
| |
Collapse
|
2
|
To A, Yu Z, Sugimura R. Recent advancement in the spatial immuno-oncology. Semin Cell Dev Biol 2025; 166:22-28. [PMID: 39705969 DOI: 10.1016/j.semcdb.2024.12.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 12/11/2024] [Indexed: 12/23/2024]
Abstract
Recent advancements in spatial transcriptomics and spatial proteomics enabled the high-throughput profiling of single or multi-cell types and cell states with spatial information. They transformed our understanding of the higher-order architectures and paired cell-cell interactions within a tumor microenvironment (TME). Within less than a decade, this rapidly emerging field has discovered much crucial fundamental knowledge and significantly improved clinical diagnosis in the field of immuno-oncology. This review summarizes the conceptual frameworks to understand spatial omics data and highlights the updated knowledge of spatial immuno-oncology.
Collapse
Affiliation(s)
- Alex To
- School of Biomedical Sciences, University of Hong Kong, Hong Kong
| | - Zou Yu
- School of Biomedical Sciences, University of Hong Kong, Hong Kong
| | - Ryohichi Sugimura
- School of Biomedical Sciences, University of Hong Kong, Hong Kong; Centre for Translational Stem Cell Biology, Hong Kong.
| |
Collapse
|
3
|
Nia HT, Munn LL, Jain RK. Probing the physical hallmarks of cancer. Nat Methods 2025:10.1038/s41592-024-02564-4. [PMID: 39815103 DOI: 10.1038/s41592-024-02564-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Accepted: 11/11/2024] [Indexed: 01/18/2025]
Abstract
The physical microenvironment plays a crucial role in tumor development, progression, metastasis and treatment. Recently, we proposed four physical hallmarks of cancer, with distinct origins and consequences, to characterize abnormalities in the physical tumor microenvironment: (1) elevated compressive-tensile solid stresses, (2) elevated interstitial fluid pressure and the resulting interstitial fluid flow, (3) altered material properties (for example, increased tissue stiffness) and (4) altered physical micro-architecture. As this emerging field of physical oncology is being advanced by tumor biologists, cell and developmental biologists, engineers, physicists and oncologists, there is a critical need for model systems and measurement tools to mechanistically probe these physical hallmarks. Here, after briefly defining these physical hallmarks, we discuss the tools and model systems available for probing each hallmark in vitro, ex vivo, in vivo and in clinical settings. We finally review the unmet needs for mechanistic probing of the physical hallmarks of tumors and discuss the challenges and unanswered questions associated with each hallmark.
Collapse
Affiliation(s)
- Hadi T Nia
- Department of Biomedical Engineering, Boston University, Boston, MA, USA.
| | - Lance L Munn
- Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
| | - Rakesh K Jain
- Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
4
|
Wang R, Hastings WJ, Saliba JG, Bao D, Huang Y, Maity S, Kamal Ahmad OM, Hu L, Wang S, Fan J, Ning B. Applications of Nanotechnology for Spatial Omics: Biological Structures and Functions at Nanoscale Resolution. ACS NANO 2025; 19:73-100. [PMID: 39704725 PMCID: PMC11752498 DOI: 10.1021/acsnano.4c11505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 11/30/2024] [Accepted: 12/10/2024] [Indexed: 12/21/2024]
Abstract
Spatial omics methods are extensions of traditional histological methods that can illuminate important biomedical mechanisms of physiology and disease by examining the distribution of biomolecules, including nucleic acids, proteins, lipids, and metabolites, at microscale resolution within tissues or individual cells. Since, for some applications, the desired resolution for spatial omics approaches the nanometer scale, classical tools have inherent limitations when applied to spatial omics analyses, and they can measure only a limited number of targets. Nanotechnology applications have been instrumental in overcoming these bottlenecks. When nanometer-level resolution is needed for spatial omics, super resolution microscopy or detection imaging techniques, such as mass spectrometer imaging, are required to generate precise spatial images of target expression. DNA nanostructures are widely used in spatial omics for purposes such as nucleic acid detection, signal amplification, and DNA barcoding for target molecule labeling, underscoring advances in spatial omics. Other properties of nanotechnologies include advanced spatial omics methods, such as microfluidic chips and DNA barcodes. In this review, we describe how nanotechnologies have been applied to the development of spatial transcriptomics, proteomics, metabolomics, epigenomics, and multiomics approaches. We focus on how nanotechnology supports improved resolution and throughput of spatial omics, surpassing traditional techniques. We also summarize future challenges and opportunities for the application of nanotechnology to spatial omics methods.
Collapse
Affiliation(s)
- Ruixuan Wang
- Center
for Cellular and Molecular Diagnostics, Tulane University School of Medicine, New Orleans, Louisiana 70112, United States
- Department
of Biochemistry and Molecular Biology, Tulane
University School of Medicine, New Orleans, Louisiana 70112, United States
| | - Waylon J. Hastings
- Department
of Psychiatry and Behavioral Science, Tulane
University School of Medicine, New Orleans, Louisiana 70112, United States
| | - Julian G. Saliba
- Center
for Cellular and Molecular Diagnostics, Tulane University School of Medicine, New Orleans, Louisiana 70112, United States
- Department
of Biochemistry and Molecular Biology, Tulane
University School of Medicine, New Orleans, Louisiana 70112, United States
| | - Duran Bao
- Center
for Cellular and Molecular Diagnostics, Tulane University School of Medicine, New Orleans, Louisiana 70112, United States
- Department
of Biochemistry and Molecular Biology, Tulane
University School of Medicine, New Orleans, Louisiana 70112, United States
| | - Yuanyu Huang
- Center
for Cellular and Molecular Diagnostics, Tulane University School of Medicine, New Orleans, Louisiana 70112, United States
- Department
of Biochemistry and Molecular Biology, Tulane
University School of Medicine, New Orleans, Louisiana 70112, United States
| | - Sudipa Maity
- Center
for Cellular and Molecular Diagnostics, Tulane University School of Medicine, New Orleans, Louisiana 70112, United States
- Department
of Biochemistry and Molecular Biology, Tulane
University School of Medicine, New Orleans, Louisiana 70112, United States
| | - Omar Mustafa Kamal Ahmad
- Center
for Cellular and Molecular Diagnostics, Tulane University School of Medicine, New Orleans, Louisiana 70112, United States
- Department
of Biochemistry and Molecular Biology, Tulane
University School of Medicine, New Orleans, Louisiana 70112, United States
| | - Logan Hu
- Groton
School, 282 Farmers Row, Groton, Massachusetts 01450, United States
| | - Shengyu Wang
- St.
Margaret’s Episcopal School, 31641 La Novia Avenue, San
Juan Capistrano, California92675, United States
| | - Jia Fan
- Center
for Cellular and Molecular Diagnostics, Tulane University School of Medicine, New Orleans, Louisiana 70112, United States
- Department
of Biochemistry and Molecular Biology, Tulane
University School of Medicine, New Orleans, Louisiana 70112, United States
| | - Bo Ning
- Center
for Cellular and Molecular Diagnostics, Tulane University School of Medicine, New Orleans, Louisiana 70112, United States
- Department
of Biochemistry and Molecular Biology, Tulane
University School of Medicine, New Orleans, Louisiana 70112, United States
| |
Collapse
|
5
|
Ravichandran A, Mahajan V, van de Kemp T, Taubenberger A, Bray LJ. Phenotypic analysis of complex bioengineered 3D models. Trends Cell Biol 2025:S0962-8924(24)00257-5. [PMID: 39794253 DOI: 10.1016/j.tcb.2024.12.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 12/09/2024] [Accepted: 12/10/2024] [Indexed: 01/13/2025]
Abstract
With advances in underlying technologies such as complex multicellular systems, synthetic materials, and bioengineering techniques, we can now generate in vitro miniaturized human tissues that recapitulate the organotypic features of normal or diseased tissues. Importantly, these 3D culture models have increasingly provided experimental access to diverse and complex tissues architectures and their morphogenic assembly in vitro. This review presents an analytical toolbox for biological researchers using 3D modeling technologies through which they can find a collation of currently available methods to phenotypically assess their 3D models in their normal state as well as their response to therapeutic or pathological agents.
Collapse
Affiliation(s)
- Akhilandeshwari Ravichandran
- Centre for Biomedical Technologies, Queensland University of Technology (QUT), Brisbane, QLD 4059, Australia; School of Mechanical, Medical, and Process Engineering, Queensland University of Technology (QUT), Brisbane, QLD 4059, Australia
| | - Vaibhav Mahajan
- Biotechnology Center, Center for Molecular and Cellular Bioengineering (CMCB), Technische Universität Dresden, 01307 Dresden, Germany
| | - Tom van de Kemp
- Centre for Biomedical Technologies, Queensland University of Technology (QUT), Brisbane, QLD 4059, Australia; School of Mechanical, Medical, and Process Engineering, Queensland University of Technology (QUT), Brisbane, QLD 4059, Australia
| | - Anna Taubenberger
- Biotechnology Center, Center for Molecular and Cellular Bioengineering (CMCB), Technische Universität Dresden, 01307 Dresden, Germany
| | - Laura J Bray
- Centre for Biomedical Technologies, Queensland University of Technology (QUT), Brisbane, QLD 4059, Australia; School of Mechanical, Medical, and Process Engineering, Queensland University of Technology (QUT), Brisbane, QLD 4059, Australia; Australian Research Council (ARC) Training Centre for Cell and Tissue Engineering Technologies, Queensland University of Technology (QUT), Brisbane, QLD 4059, Australia.
| |
Collapse
|
6
|
Hong SM, Qian X, Deshpande V, Kulkarni S. Optimization of protocols for immunohistochemical assessment of enteric nervous system in formalin fixed human tissue. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.15.628584. [PMID: 39763767 PMCID: PMC11702535 DOI: 10.1101/2024.12.15.628584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/31/2025]
Abstract
Gastrointestinal (GI) motility is regulated in a large part by the cells of the enteric nervous system (ENS), suggesting that ENS dysfunctions either associate with, or drive GI dysmotility in patients. However, except for select diseases such as Hirschsprung's Disease or Achalasia that show a significant loss of all neurons or a subset of neurons, our understanding of human ENS histopathology is extremely limited. Recent endoscopic advances allow biopsying patient's full thickness gut tissues, which makes capturing ENS tissues simpler than biopsying other neuronal tissues, such as the brain. Yet, our understanding of ENS aberrations observed in GI dysmotility patients lags behind our understanding of central nervous system aberrations observed in patients with neurological disease. Paucity of optimized methods for histopathological assessment of ENS in pathological specimens represent an important bottleneck in ascertaining how the ENS is altered in diverse GI dysmotility conditions. While recent studies have interrogated ENS structure in surgically resected whole mount human gut, most pathological specimens are banked as formalin fixed paraffin embedded (FFPE) tissue blocks - suggesting that methods to interrogate ENS in FFPE tissue blocks would provide the biggest impetus for ENS histopathology in a clinical setting. In this report, we present optimized methods for immunohistochemical interrogation of the human ENS tissue on the basis of >25 important protein markers that include proteins expressed by all neurons, subset of neurons, hormones, and neurotransmitter receptors. This report provides a resource which will help pathologists and investigators assess ENS aberrations in patients with various GI dysmotility conditions.
Collapse
Affiliation(s)
- Su Min Hong
- Division of Gastroenterology, Dept of Medicine, Beth Israel Deaconess Medical Center, Boston, MA 02115
| | - Xia Qian
- Dept of Pathology, Beth Israel Deaconess Medical Center, Boston, MA 02115
| | - Vikram Deshpande
- Dept of Pathology, Beth Israel Deaconess Medical Center, Boston, MA 02115
| | - Subhash Kulkarni
- Division of Gastroenterology, Dept of Medicine, Beth Israel Deaconess Medical Center, Boston, MA 02115
- Division of Medical Sciences, Harvard Medical School, Boston, MA 02115
- Graduate program in Neuroscience, Harvard Medical School, Boston, MA 02115
| |
Collapse
|
7
|
Fayzullin A, Ivanova E, Grinin V, Ermilov D, Solovyeva S, Balyasin M, Bakulina A, Nikitin P, Valieva Y, Kalinichenko A, Arutyunyan A, Lychagin A, Timashev P. Towards accurate and efficient diagnoses in nephropathology: An AI-based approach for assessing kidney transplant rejection. Comput Struct Biotechnol J 2024; 24:571-582. [PMID: 39258238 PMCID: PMC11385065 DOI: 10.1016/j.csbj.2024.08.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 08/11/2024] [Accepted: 08/11/2024] [Indexed: 09/12/2024] Open
Abstract
The Banff classification is useful for diagnosing renal transplant rejection. However, it has limitations due to subjectivity and varying concordance in physicians' assessments. Artificial intelligence (AI) can help standardize research, increase objectivity and accurately quantify morphological characteristics, improving reproducibility in clinical practice. This study aims to develop an AI-based solutions for diagnosing acute kidney transplant rejection by introducing automated evaluation of prognostic morphological patterns. The proposed approach aims to help accurately distinguish borderline changes from rejection. We trained a deep-learning model utilizing a fine-tuned Mask R-CNN architecture which achieved a mean Average Precision value of 0.74 for the segmentation of renal tissue structures. A strong positive nonlinear correlation was found between the measured infiltration areas and fibrosis, indicating the model's potential for assessing these parameters in kidney biopsies. The ROC analysis showed a high predictive ability for distinguishing between ci and i scores based on infiltration area and fibrosis area measurements. The AI model demonstrated high precision in predicting clinical scores which makes it a promising AI assisting tool for pathologists. The application of AI in nephropathology has a potential for advancements, including automated morphometric evaluation, 3D histological models and faster processing to enhance diagnostic accuracy and efficiency.
Collapse
Affiliation(s)
- Alexey Fayzullin
- Institute for Regenerative Medicine, Sechenov First Moscow State Medical University (Sechenov University), 8-2 Trubetskaya st., Moscow 119991, Russia
- World-Class Research Center "Digital Biodesign and Personalized Healthcare, Sechenov First Moscow State Medical University (Sechenov University), 8-2 Trubetskaya st., Moscow 119991, Russia
| | - Elena Ivanova
- Institute for Regenerative Medicine, Sechenov First Moscow State Medical University (Sechenov University), 8-2 Trubetskaya st., Moscow 119991, Russia
- B.V.Petrovsky Russian Research Center of Surgery, 2 Abrikosovskiy lane, Moscow 119991, Russia
| | - Victor Grinin
- PJSC VimpelCom, 10 8th March Street, Moscow 127083, Russia
| | - Dmitry Ermilov
- PJSC VimpelCom, 10 8th March Street, Moscow 127083, Russia
| | - Svetlana Solovyeva
- B.V.Petrovsky Russian Research Center of Surgery, 2 Abrikosovskiy lane, Moscow 119991, Russia
| | - Maxim Balyasin
- Scientific and Educational Resource Center, Peoples' Friendship University of Russia, 6 Miklukho-Maklaya st., Moscow 117198, Russia
| | - Alesia Bakulina
- Institute for Regenerative Medicine, Sechenov First Moscow State Medical University (Sechenov University), 8-2 Trubetskaya st., Moscow 119991, Russia
| | - Pavel Nikitin
- Institute for Regenerative Medicine, Sechenov First Moscow State Medical University (Sechenov University), 8-2 Trubetskaya st., Moscow 119991, Russia
| | - Yana Valieva
- Institute for Regenerative Medicine, Sechenov First Moscow State Medical University (Sechenov University), 8-2 Trubetskaya st., Moscow 119991, Russia
- World-Class Research Center "Digital Biodesign and Personalized Healthcare, Sechenov First Moscow State Medical University (Sechenov University), 8-2 Trubetskaya st., Moscow 119991, Russia
| | - Alina Kalinichenko
- Institute for Regenerative Medicine, Sechenov First Moscow State Medical University (Sechenov University), 8-2 Trubetskaya st., Moscow 119991, Russia
| | | | - Aleksey Lychagin
- Department of Trauma, Orthopedics and Disaster Surgery, Sechenov First Moscow State Medical University (Sechenov University), 8-2 Trubetskaya st., Moscow 119991, Russia
| | - Peter Timashev
- Institute for Regenerative Medicine, Sechenov First Moscow State Medical University (Sechenov University), 8-2 Trubetskaya st., Moscow 119991, Russia
- World-Class Research Center "Digital Biodesign and Personalized Healthcare, Sechenov First Moscow State Medical University (Sechenov University), 8-2 Trubetskaya st., Moscow 119991, Russia
| |
Collapse
|
8
|
Chen JH, Elmelech L, Tang AL, Hacohen N. Powerful microscopy technologies decode spatially organized cellular networks that drive response to immunotherapy in humans. Curr Opin Immunol 2024; 91:102463. [PMID: 39277910 PMCID: PMC11609032 DOI: 10.1016/j.coi.2024.102463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Revised: 08/22/2024] [Accepted: 08/24/2024] [Indexed: 09/17/2024]
Abstract
In tumors, immune cells organize into networks of different sizes and composition, including complex tertiary lymphoid structures and recently identified networks centered around the chemokines CXCL9/10/11 and CCL19. New commercially available highly multiplexed microscopy using cyclical RNA in situ hybridization and antibody-based approaches have the potential to establish the organization of the immune response in human tissue and serve as a foundation for future immunology research.
Collapse
Affiliation(s)
- Jonathan H Chen
- Northwestern University, Feinberg School of Medicine, Department of Pathology, Chicago, IL, USA; Northwestern University, Feinberg School of Medicine, Center for Human Immunobiology, Chicago, IL, USA; Krantz Family Center for Cancer Research, Massachusetts General Hospital (MGH) Cancer Center, Harvard Medical School (HMS), Boston, MA, USA; Department of Pathology, MGH, Boston, MA, USA; Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA; Harvard Medical School, Boston, MA, USA.
| | - Liad Elmelech
- Krantz Family Center for Cancer Research, Massachusetts General Hospital (MGH) Cancer Center, Harvard Medical School (HMS), Boston, MA, USA; Department of Pathology, MGH, Boston, MA, USA; Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
| | - Alexander L Tang
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
| | - Nir Hacohen
- Krantz Family Center for Cancer Research, Massachusetts General Hospital (MGH) Cancer Center, Harvard Medical School (HMS), Boston, MA, USA; Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA; Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
9
|
Quail DF, Walsh LA. Revolutionizing cancer research with spatial proteomics and visual intelligence. Nat Methods 2024; 21:2216-2219. [PMID: 39643688 DOI: 10.1038/s41592-024-02542-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/09/2024]
Affiliation(s)
- Daniela F Quail
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, Quebec, Canada.
- Department of Physiology, McGill University, Montreal, Quebec, Canada.
- Division of Experimental Medicine, McGill University, Montreal, Quebec, Canada.
| | - Logan A Walsh
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, Quebec, Canada.
- Department of Human Genetics, McGill University, Montreal, Quebec, Canada.
| |
Collapse
|
10
|
Iyer MK, Fletcher A, Shi C, Chen F, Kanu E, Eckhoff AM, Bao M, Frankel TL, Chinnaiyan AM, Nussbaum DP, Allen PJ. Spatial Transcriptomics of IPMN Reveals Divergent Indolent and Malignant Lineages. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.29.620810. [PMID: 39554015 PMCID: PMC11565728 DOI: 10.1101/2024.10.29.620810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
Purpose Intraductal papillary mucinous neoplasms (IPMN) occur in 5-10% of the population, but only a small minority progress to pancreatic ductal adenocarcinoma (PDAC). The lack of accurate predictors of high-risk disease leads both to unnecessary operations for indolent neoplasms as well as missed diagnoses of PDAC. Digital spatial RNA profiling (DSP-RNA) provides an opportunity to define and associate transcriptomic states with cancer risk. Experimental Design Whole-transcriptome DSP-RNA profiling was performed on 10 IPMN specimens encompassing the spectrum of dysplastic changes from normal duct to cancer. Ductal epithelial regions within each tissue were annotated as normal duct (NL), low-grade dysplasia (LGD), high-grade dysplasia (HGD), or invasive carcinoma (INV). Gene expression count data was generated by Illumina sequencing and analyzed with R/Bioconductor. Results Dimension reduction analysis exposed three clusters reflecting IPMN transcriptomic states denoted "normal-like" ( cNL ), "low-risk" ( cLR ) and "high-risk" ( cHR ). In addition to specific marker genes, the three states exhibited significant enrichment for the exocrine, classical, and basal-like programs in PDAC. Specifically, exocrine function diminished in cHR , classical activation distinguished neoplasia from cNL , and basal-like genes were specifically upregulated in cHR . Intriguingly, markers of cHR were detected in NL and LGD regions from specimens with PDAC but not low-grade IPMN. Conclusions DSP-RNA of IPMN revealed low-risk (indolent) and high-risk (malignant) expression programs that correlated with the activity of exocrine and basal-like PDAC signatures, respectively, and distinguished pathologically low-grade from malignant specimens. These findings contextualize IPMN pathogenesis and have the potential to transform existing risk stratification models. Statement of translational relevance Current consensus guidelines for management of intraductal papillary mucinous neoplasms (IPMN) of the pancreas utilize clinical and radiographic criteria for risk stratification. Unfortunately, the estimated positive predictive value of these criteria for IPMN-associated pancreatic ductal adenocarcinoma (PDAC) is under 50%, indicating that over half of pancreatectomies are performed for benign disease. Moreover, nearly 15% of patients who were deemed "low risk" by the same criteria harbored PDAC. Surgical resection of IPMN has maximal benefit when performed prior to the development of PDAC, as evidence of carcinoma has been associated with a high rate of recurrence and poor overall survival. Thus, the development of molecular diagnostics that improve the accuracy of IPMN risk classification would have immediate relevance for patient care, both in terms of better selecting patients for potentially curative operations, as well as sparing patients with low-risk lesions from invasive procedures.
Collapse
|
11
|
Bao R, Hutson A, Madabhushi A, Jonsson VD, Rosario SR, Barnholtz-Sloan JS, Fertig EJ, Marathe H, Harris L, Altreuter J, Chen Q, Dignam J, Gentles AJ, Gonzalez-Kozlova E, Gnjatic S, Kim E, Long M, Morgan M, Ruppin E, Valen DV, Zhang H, Vokes N, Meerzaman D, Liu S, Van Allen EM, Xing Y. Ten challenges and opportunities in computational immuno-oncology. J Immunother Cancer 2024; 12:e009721. [PMID: 39461879 PMCID: PMC11529678 DOI: 10.1136/jitc-2024-009721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 09/23/2024] [Indexed: 10/29/2024] Open
Abstract
Immuno-oncology has transformed the treatment of cancer, with several immunotherapies becoming the standard treatment across histologies. Despite these advancements, the majority of patients do not experience durable clinical benefits, highlighting the imperative for ongoing advancement in immuno-oncology. Computational immuno-oncology emerges as a forefront discipline that draws on biomedical data science and intersects with oncology, immunology, and clinical research, with the overarching goal to accelerate the development of effective and safe immuno-oncology treatments from the laboratory to the clinic. In this review, we outline 10 critical challenges and opportunities in computational immuno-oncology, emphasizing the importance of robust computational strategies and interdisciplinary collaborations amid the constantly evolving interplay between clinical needs and technological innovation.
Collapse
Affiliation(s)
- Riyue Bao
- UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Alan Hutson
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Anant Madabhushi
- Emory University, Atlanta, Georgia, USA
- Georgia Institute of Technology, Atlanta, Georgia, USA
- Atlanta Veterans Affairs Medical Center, Atlanta, Georgia, USA
| | - Vanessa D Jonsson
- Department of Biomolecular Engineering, University of California Santa Cruz, Santa Cruz, California, USA
- Genomics Institute, University of California Santa Cruz, Santa Cruz, California, USA
| | - Spencer R Rosario
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Jill S Barnholtz-Sloan
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland, USA
- Center for Biomedical Informatics & Information Technology, National Cancer Institute, Bethesda, Maryland, USA
| | - Elana J Fertig
- Department of Oncology, Johns Hopkins University, Baltimore, Maryland, USA
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland, USA
- Department of Applied Mathematics and Statistics, Johns Hopkins University, Baltimore, Maryland, USA
| | - Himangi Marathe
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Lyndsay Harris
- Cancer Diagnosis Program, National Cancer Institute Division of Cancer Treatment and Diagnosis, Bethesda, Maryland, USA
| | | | - Qingrong Chen
- Center for Biomedical Informatics and Information Technology, National Cancer Institute, Rockville, Maryland, USA
| | - James Dignam
- Department of Public Health Sciences, University of Chicago Division of the Biological Sciences, Chicago, Illinois, USA
| | - Andrew J Gentles
- Department of Pathology, Stanford University, Stanford, California, USA
| | - Edgar Gonzalez-Kozlova
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Immunology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Sacha Gnjatic
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Immunology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Erika Kim
- Informatics and Data Science Program, Center for Biomedical Informatics and Information Technology, National Cancer Institute, Bethesda, Maryland, USA
| | - Mark Long
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Martin Morgan
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Eytan Ruppin
- Cancer Data Science Laboratory, National Cancer Institute, Bethesda, Maryland, USA
| | - David Van Valen
- Division of Computing and Mathematical Science, Caltech, Pasadena, California, USA
- Howard Hughes Medical Institute, Chevy Chase, Maryland, USA
| | - Hong Zhang
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Natalie Vokes
- Department of Thoracic and Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Daoud Meerzaman
- Center for Biomedical Informatics and Information Technology, National Cancer Institute, Rockville, Maryland, USA
| | - Song Liu
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Eliezer M Van Allen
- Harvard Medical School, Boston, Massachusetts, USA
- Division of Population Sciences, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Yi Xing
- Center for Computational and Genomic Medicine, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Biomedical and Health Informatics, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| |
Collapse
|
12
|
O’Brien J, Niehaus P, Chang K, Remark J, Barrett J, Dasgupta A, Adenegan M, Salimian M, Kevas Y, Chandrasekaran K, Kristian T, Chellappan R, Rubin S, Kiemen A, Lu CPJ, Russell JW, Ho CY. Skin keratinocyte-derived SIRT1 and BDNF modulate mechanical allodynia in mouse models of diabetic neuropathy. Brain 2024; 147:3471-3486. [PMID: 38554393 PMCID: PMC11449144 DOI: 10.1093/brain/awae100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 03/06/2024] [Accepted: 03/08/2024] [Indexed: 04/01/2024] Open
Abstract
Diabetic neuropathy is a debilitating disorder characterized by spontaneous and mechanical allodynia. The role of skin mechanoreceptors in the development of mechanical allodynia is unclear. We discovered that mice with diabetic neuropathy had decreased sirtuin 1 (SIRT1) deacetylase activity in foot skin, leading to reduced expression of brain-derived neurotrophic factor (BDNF) and subsequent loss of innervation in Meissner corpuscles, a mechanoreceptor expressing the BDNF receptor TrkB. When SIRT1 was depleted from skin, the mechanical allodynia worsened in diabetic neuropathy mice, likely due to retrograde degeneration of the Meissner-corpuscle innervating Aβ axons and aberrant formation of Meissner corpuscles which may have increased the mechanosensitivity. The same phenomenon was also noted in skin-keratinocyte specific BDNF knockout mice. Furthermore, overexpression of SIRT1 in skin induced Meissner corpuscle reinnervation and regeneration, resulting in significant improvement of diabetic mechanical allodynia. Overall, the findings suggested that skin-derived SIRT1 and BDNF function in the same pathway in skin sensory apparatus regeneration and highlighted the potential of developing topical SIRT1-activating compounds as a novel treatment for diabetic mechanical allodynia.
Collapse
Affiliation(s)
- Jennifer O’Brien
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Peter Niehaus
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Koping Chang
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
- Department of Pathology, National Taiwan University, Taipei, 100, Taiwan
| | - Juliana Remark
- Hansjörg Wyss Department of Plastic Surgery, Department of Cell Biology, New York University School of Medicine, New York, NY 10016, USA
| | - Joy Barrett
- Hansjörg Wyss Department of Plastic Surgery, Department of Cell Biology, New York University School of Medicine, New York, NY 10016, USA
| | - Abhishikta Dasgupta
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Morayo Adenegan
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Mohammad Salimian
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Yanni Kevas
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Krish Chandrasekaran
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Baltimore Veterans Affairs Medical Center, Baltimore, MD 21201, USA
| | - Tibor Kristian
- Department of Anesthesiology, University of Maryland School of Medicine, Baltimore, MD 21021, USA
| | - Rajeshwari Chellappan
- Department of Pathology, University of Alabama Birmingham, Birmingham, AL 35233, USA
| | - Samuel Rubin
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
- Department of Chemistry, College of William and Mary, Williamsburg, VA 23187, USA
| | - Ashley Kiemen
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Catherine Pei-Ju Lu
- Hansjörg Wyss Department of Plastic Surgery, Department of Cell Biology, New York University School of Medicine, New York, NY 10016, USA
| | - James W Russell
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Baltimore Veterans Affairs Medical Center, Baltimore, MD 21201, USA
| | - Cheng-Ying Ho
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| |
Collapse
|
13
|
Bareham B, Dibble M, Parsons M. Defining and modeling dynamic spatial heterogeneity within tumor microenvironments. Curr Opin Cell Biol 2024; 90:102422. [PMID: 39216233 DOI: 10.1016/j.ceb.2024.102422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 08/05/2024] [Accepted: 08/07/2024] [Indexed: 09/04/2024]
Abstract
Many solid tumors exhibit significant genetic, cellular, and biophysical heterogeneity which dynamically evolves during disease progression and after treatment. This constant flux in cell composition, phenotype, spatial relationships, and tissue properties poses significant challenges in accurately diagnosing and treating patients. Much of the complexity lies in unraveling the molecular changes in different tumor compartments, how they influence one another in space and time and where vulnerabilities exist that might be appropriate to target therapeutically. Recent advances in spatial profiling tools and technologies are enabling new insight into the underlying biology of complex tumors, creating a greater understanding of the intricate relationship between cell types, states, and the microenvironment. Here we reflect on some recent discoveries in this area, where the key knowledge and technology gaps lie, and the advancements in spatial measurements and in vitro models for the study of spatial intratumoral heterogeneity.
Collapse
Affiliation(s)
- Bethany Bareham
- Randall Centre for Cell and Molecular Biophysics, King's College London, New Hunt's House, Guy's Campus, London, SE1 1UL, UK
| | - Matthew Dibble
- Randall Centre for Cell and Molecular Biophysics, King's College London, New Hunt's House, Guy's Campus, London, SE1 1UL, UK
| | - Maddy Parsons
- Randall Centre for Cell and Molecular Biophysics, King's College London, New Hunt's House, Guy's Campus, London, SE1 1UL, UK.
| |
Collapse
|
14
|
Crawford AJ, Forjaz A, Bons J, Bhorkar I, Roy T, Schell D, Queiroga V, Ren K, Kramer D, Huang W, Russo GC, Lee MH, Wu PH, Shih IM, Wang TL, Atkinson MA, Schilling B, Kiemen AL, Wirtz D. Combined assembloid modeling and 3D whole-organ mapping captures the microanatomy and function of the human fallopian tube. SCIENCE ADVANCES 2024; 10:eadp6285. [PMID: 39331707 PMCID: PMC11430475 DOI: 10.1126/sciadv.adp6285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 08/22/2024] [Indexed: 09/29/2024]
Abstract
The fallopian tubes play key roles in processes from pregnancy to ovarian cancer where three-dimensional (3D) cellular and extracellular interactions are important to their pathophysiology. Here, we develop a 3D multicompartment assembloid model of the fallopian tube that molecularly, functionally, and architecturally resembles the organ. Global label-free proteomics, innovative assays capturing physiological functions of the fallopian tube (i.e., oocyte transport), and whole-organ single-cell resolution mapping are used to validate these assembloids through a multifaceted platform with direct comparisons to fallopian tube tissue. These techniques converge at a unique combination of assembloid parameters with the highest similarity to the reference fallopian tube. This work establishes (i) an optimized model of the human fallopian tubes for in vitro studies of their pathophysiology and (ii) an iterative platform for customized 3D in vitro models of human organs that are molecularly, functionally, and microanatomically accurate by combining tunable assembloid and tissue mapping methods.
Collapse
Affiliation(s)
- Ashleigh J Crawford
- Johns Hopkins Institute for Nanobiotechnology, The Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Chemical and Biomolecular Engineering, Johns Hopkins Physical Sciences-Oncology Center, The Johns Hopkins University, Baltimore, MD 21218, USA
| | - André Forjaz
- Johns Hopkins Institute for Nanobiotechnology, The Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Chemical and Biomolecular Engineering, Johns Hopkins Physical Sciences-Oncology Center, The Johns Hopkins University, Baltimore, MD 21218, USA
| | - Joanna Bons
- Buck Institute for Research on Aging, Novato, CA 94945, USA
| | - Isha Bhorkar
- Johns Hopkins Institute for Nanobiotechnology, The Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Biomedical Engineering, The Johns Hopkins University, Baltimore, MD 21218, USA
| | - Triya Roy
- Johns Hopkins Institute for Nanobiotechnology, The Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Chemical and Biomolecular Engineering, Johns Hopkins Physical Sciences-Oncology Center, The Johns Hopkins University, Baltimore, MD 21218, USA
| | - David Schell
- Johns Hopkins Institute for Nanobiotechnology, The Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Chemical and Biomolecular Engineering, Johns Hopkins Physical Sciences-Oncology Center, The Johns Hopkins University, Baltimore, MD 21218, USA
| | - Vasco Queiroga
- Johns Hopkins Institute for Nanobiotechnology, The Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Chemical and Biomolecular Engineering, Johns Hopkins Physical Sciences-Oncology Center, The Johns Hopkins University, Baltimore, MD 21218, USA
| | - Kehan Ren
- Johns Hopkins Institute for Nanobiotechnology, The Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Biomedical Engineering, The Johns Hopkins University, Baltimore, MD 21218, USA
| | - Donald Kramer
- Johns Hopkins Institute for Nanobiotechnology, The Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Biotechnology, Johns Hopkins Advanced Academic Programs, The Johns Hopkins University, Baltimore, MD 21218, USA
| | - Wilson Huang
- Johns Hopkins Institute for Nanobiotechnology, The Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Biology, The Johns Hopkins University, Baltimore, MD 21218, USA
| | - Gabriella C Russo
- Johns Hopkins Institute for Nanobiotechnology, The Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Chemical and Biomolecular Engineering, Johns Hopkins Physical Sciences-Oncology Center, The Johns Hopkins University, Baltimore, MD 21218, USA
| | - Meng-Horng Lee
- Johns Hopkins Institute for Nanobiotechnology, The Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Chemical and Biomolecular Engineering, Johns Hopkins Physical Sciences-Oncology Center, The Johns Hopkins University, Baltimore, MD 21218, USA
| | - Pei-Hsun Wu
- Johns Hopkins Institute for Nanobiotechnology, The Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Chemical and Biomolecular Engineering, Johns Hopkins Physical Sciences-Oncology Center, The Johns Hopkins University, Baltimore, MD 21218, USA
| | - Ie-Ming Shih
- Department of Gynecology and Obstetrics, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Tian-Li Wang
- Department of Gynecology and Obstetrics, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Mark A Atkinson
- Departments of Pathology, Immunology, and Laboratory Medicine, College of Medicine, University of Florida Diabetes Institute, Gainesville, FL 32610, USA
- Departments of Pediatrics, College of Medicine, University of Florida Diabetes Institute, Gainesville, FL 32610, USA
| | | | - Ashley L Kiemen
- Johns Hopkins Institute for Nanobiotechnology, The Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Chemical and Biomolecular Engineering, Johns Hopkins Physical Sciences-Oncology Center, The Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Functional Anatomy and Evolution, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Denis Wirtz
- Johns Hopkins Institute for Nanobiotechnology, The Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Chemical and Biomolecular Engineering, Johns Hopkins Physical Sciences-Oncology Center, The Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Biomedical Engineering, The Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
15
|
Sidiropoulos DN, Shin SM, Wetzel M, Girgis AA, Bergman D, Danilova L, Perikala S, Shu DH, Montagne JM, Deshpande A, Leatherman J, Dequiedt L, Jacobs V, Ogurtsova A, Mo G, Yuan X, Lvovs D, Stein-O'Brien G, Yarchoan M, Zhu Q, Harper EI, Weeraratna AT, Kiemen AL, Jaffee EM, Zheng L, Ho WJ, Anders RA, Fertig EJ, Kagohara LT. Spatial multi-omics reveal intratumoral humoral immunity niches associated with tertiary lymphoid structures in pancreatic cancer immunotherapy pathologic responders. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.22.613714. [PMID: 39386736 PMCID: PMC11463490 DOI: 10.1101/2024.09.22.613714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Pancreatic adenocarcinoma (PDAC) is a rapidly progressing cancer that responds poorly to immunotherapies. Intratumoral tertiary lymphoid structures (TLS) have been associated with rare long-term PDAC survivors, but the role of TLS in PDAC and their spatial relationships within the context of the broader tumor microenvironment remain unknown. We generated a spatial multi-omics atlas encompassing 26 PDAC tumors from patients treated with combination immunotherapies. Using machine learning-enabled H&E image classification models and unsupervised gene expression matrix factorization methods for spatial transcriptomics, we characterized cellular states within TLS niches spanning across distinct morphologies and immunotherapies. Unsupervised learning generated a TLS-specific spatial gene expression signature that significantly associates with improved survival in PDAC patients. These analyses demonstrate TLS-associated intratumoral B cell maturation in pathological responders, confirmed with spatial proteomics and BCR profiling. Our study also identifies spatial features of pathologic immune responses, revealing TLS maturation colocalizing with IgG/IgA distribution and extracellular matrix remodeling. GRAPHICAL ABSTRACT HIGHLIGHTS Integrated multi-modal spatial profiling of human PDAC tumors from neoadjuvant immunotherapy clinical trials reveal diverse spatial niches enriched in TLS.TLS maturity is influenced by tumor location and the cellular neighborhoods in which TLS immune cells are recruited.Unsupervised machine learning of genome-wide signatures on spatial transcriptomics data characterizes the TLS-enriched TME and associates TLS transcriptomes with survival outcomes in PDAC.Interactions of spatially variable gene expression patterns showed TLS maturation is coupled with immunoglobulin distribution and ECM remodeling in pathologic responders.Intratumoral plasma cell and immunoglobin gene expression spatial dynamics demonstrate trafficking of TLS-driven humoral immunity in the PDAC TME. Significance We report a spatial multi-omics atlas of PDAC tumors from a series of immunotherapy neoadjuvant clinical trials. Intratumorally, pathologic responders exhibit mature TLS that propagate plasma cells into malignant niches. Our findings offer insights on the role of TLS-associated humoral immunity and stromal remodeling during immunotherapy treatment.
Collapse
|
16
|
Redekop E, Pleasure M, Wang Z, Sisk A, Zong Y, Flores K, Speier W, Arnold CW. Digital Volumetric Biopsy Cores Improve Gleason Grading of Prostate Cancer Using Deep Learning. ARXIV 2024:arXiv:2409.08331v1. [PMID: 39314499 PMCID: PMC11419188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
Prostate cancer (PCa) was the most frequently diagnosed cancer among American men in 2023 [1]. The histological grading of biopsies is essential for diagnosis, and various deep learning-based solutions have been developed to assist with this task. Existing deep learning frameworks are typically applied to individual 2D cross-sections sliced from 3D biopsy tissue specimens. This process impedes the analysis of complex tissue structures such as glands, which can vary depending on the tissue slice examined. We propose a novel digital pathology data source called a "volumetric core," obtained via the extraction and co-alignment of serially sectioned tissue sections using a novel morphology-preserving alignment framework. We trained an attention-based multiple-instance learning (ABMIL) framework on deep features extracted from volumetric patches to automatically classify the Gleason Grade Group (GGG). To handle volumetric patches, we used a modified video transformer with a deep feature extractor pretrained using self-supervised learning. We ran our morphology preserving alignment framework to construct 10,210 volumetric cores, leaving out 30% for pretraining. The rest of the dataset was used to train ABMIL, which resulted in a 0.958 macro-average AUC, 0.671 F1 score, 0.661 precision, and 0.695 recall averaged across all five GGG significantly outperforming the 2D baselines.
Collapse
Affiliation(s)
- Ekaterina Redekop
- Department of Bioengineering, University of California, Los Angeles, USA
| | - Mara Pleasure
- Medical Informatics, University of California, Los Angeles, USA
| | - Zichen Wang
- Department of Bioengineering, University of California, Los Angeles, USA
| | - Anthony Sisk
- Department of Pathology, University of California, Los Angeles, USA
| | - Yang Zong
- Department of Pathology, University of California, Los Angeles, USA
| | - Kimberly Flores
- Department of Pathology, University of California, Los Angeles, USA
| | - William Speier
- Department of Bioengineering, University of California, Los Angeles, USA
- Medical Informatics, University of California, Los Angeles, USA
- Department of Radiological Sciences, University of California, Los Angeles, USA
| | - Corey W. Arnold
- Department of Bioengineering, University of California, Los Angeles, USA
- Medical Informatics, University of California, Los Angeles, USA
- Department of Pathology, University of California, Los Angeles, USA
- Department of Radiological Sciences, University of California, Los Angeles, USA
| |
Collapse
|
17
|
Chen J, Larsson L, Swarbrick A, Lundeberg J. Spatial landscapes of cancers: insights and opportunities. Nat Rev Clin Oncol 2024; 21:660-674. [PMID: 39043872 DOI: 10.1038/s41571-024-00926-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/28/2024] [Indexed: 07/25/2024]
Abstract
Solid tumours comprise many different cell types organized in spatially structured arrangements, with substantial intratumour and intertumour heterogeneity. Advances in spatial profiling technologies over the past decade hold promise to capture the complexity of these cellular architectures to build a holistic view of the intricate molecular mechanisms that shape the tumour ecosystem. Some of these mechanisms act at the cellular scale and are controlled by cell-autonomous programmes or communication between nearby cells, whereas other mechanisms result from coordinated efforts between large networks of cells and extracellular molecules organized into tissues and organs. In this Review we provide insights into the application of single-cell and spatial profiling tools, with a focus on spatially resolved transcriptomic tools developed to understand the cellular architecture of the tumour microenvironment and identify opportunities to use them to improve clinical management of cancers.
Collapse
Affiliation(s)
- Julia Chen
- Cancer Ecosystems Program, Garvan Institute of Medical Research, Sydney, New South Wales, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Sydney, New South Wales, Australia
- Department of Medical Oncology, St George Hospital, Sydney, New South Wales, Australia
| | - Ludvig Larsson
- Department of Gene Technology, KTH Royal Institute of Technology, Science for Life Laboratory, Stockholm, Sweden
| | - Alexander Swarbrick
- Cancer Ecosystems Program, Garvan Institute of Medical Research, Sydney, New South Wales, Australia.
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Sydney, New South Wales, Australia.
| | - Joakim Lundeberg
- Department of Gene Technology, KTH Royal Institute of Technology, Science for Life Laboratory, Stockholm, Sweden.
| |
Collapse
|
18
|
Liu L, Chen A, Li Y, Mulder J, Heyn H, Xu X. Spatiotemporal omics for biology and medicine. Cell 2024; 187:4488-4519. [PMID: 39178830 DOI: 10.1016/j.cell.2024.07.040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 07/05/2024] [Accepted: 07/23/2024] [Indexed: 08/26/2024]
Abstract
The completion of the Human Genome Project has provided a foundational blueprint for understanding human life. Nonetheless, understanding the intricate mechanisms through which our genetic blueprint is involved in disease or orchestrates development across temporal and spatial dimensions remains a profound scientific challenge. Recent breakthroughs in cellular omics technologies have paved new pathways for understanding the regulation of genomic elements and the relationship between gene expression, cellular functions, and cell fate determination. The advent of spatial omics technologies, encompassing both imaging and sequencing-based methodologies, has enabled a comprehensive understanding of biological processes from a cellular ecosystem perspective. This review offers an updated overview of how spatial omics has advanced our understanding of the translation of genetic information into cellular heterogeneity and tissue structural organization and their dynamic changes over time. It emphasizes the discovery of various biological phenomena, related to organ functionality, embryogenesis, species evolution, and the pathogenesis of diseases.
Collapse
Affiliation(s)
| | - Ao Chen
- BGI Research, Shenzhen 518083, China
| | | | - Jan Mulder
- Department of Neuroscience, Karolinska Institute, Stockholm, Sweden
| | - Holger Heyn
- Centro Nacional de Análisis Genómico (CNAG), Barcelona, Spain
| | - Xun Xu
- BGI Research, Hangzhou 310030, China; BGI Research, Shenzhen 518083, China.
| |
Collapse
|
19
|
Bell ATF, Mitchell JT, Kiemen AL, Lyman M, Fujikura K, Lee JW, Coyne E, Shin SM, Nagaraj S, Deshpande A, Wu PH, Sidiropoulos DN, Erbe R, Stern J, Chan R, Williams S, Chell JM, Ciotti L, Zimmerman JW, Wirtz D, Ho WJ, Zaidi N, Thompson E, Jaffee EM, Wood LD, Fertig EJ, Kagohara LT. PanIN and CAF transitions in pancreatic carcinogenesis revealed with spatial data integration. Cell Syst 2024; 15:753-769.e5. [PMID: 39116880 PMCID: PMC11409191 DOI: 10.1016/j.cels.2024.07.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 02/06/2024] [Accepted: 07/08/2024] [Indexed: 08/10/2024]
Abstract
This study introduces a new imaging, spatial transcriptomics (ST), and single-cell RNA-sequencing integration pipeline to characterize neoplastic cell state transitions during tumorigenesis. We applied a semi-supervised analysis pipeline to examine premalignant pancreatic intraepithelial neoplasias (PanINs) that can develop into pancreatic ductal adenocarcinoma (PDAC). Their strict diagnosis on formalin-fixed and paraffin-embedded (FFPE) samples limited the single-cell characterization of human PanINs within their microenvironment. We leverage whole transcriptome FFPE ST to enable the study of a rare cohort of matched low-grade (LG) and high-grade (HG) PanIN lesions to track progression and map cellular phenotypes relative to single-cell PDAC datasets. We demonstrate that cancer-associated fibroblasts (CAFs), including antigen-presenting CAFs, are located close to PanINs. We further observed a transition from CAF-related inflammatory signaling to cellular proliferation during PanIN progression. We validate these findings with single-cell high-dimensional imaging proteomics and transcriptomics technologies. Altogether, our semi-supervised learning framework for spatial multi-omics has broad applicability across cancer types to decipher the spatiotemporal dynamics of carcinogenesis.
Collapse
Affiliation(s)
- Alexander T F Bell
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Convergence Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jacob T Mitchell
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Convergence Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Bloomberg Kimmel Immunology Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Genetic Medicine, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Ashley L Kiemen
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD, USA; Department of Pathology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Melissa Lyman
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Convergence Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Kohei Fujikura
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Jae W Lee
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Convergence Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Bloomberg Kimmel Immunology Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Pathology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Erin Coyne
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Convergence Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Bloomberg Kimmel Immunology Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sarah M Shin
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Convergence Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Bloomberg Kimmel Immunology Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sushma Nagaraj
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Convergence Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Atul Deshpande
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Convergence Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Bloomberg Kimmel Immunology Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Pei-Hsun Wu
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD, USA
| | - Dimitrios N Sidiropoulos
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Convergence Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Bloomberg Kimmel Immunology Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Rossin Erbe
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Convergence Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Bloomberg Kimmel Immunology Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Genetic Medicine, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | | | | | | | | | - Lauren Ciotti
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Convergence Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jacquelyn W Zimmerman
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Convergence Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Bloomberg Kimmel Immunology Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA; The Skip Viragh Center for Clinical and Translational Research, Baltimore, MD, USA
| | - Denis Wirtz
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD, USA; Department of Materials Science and Engineering, The Johns Hopkins University, Baltimore, MD, USA; Johns Hopkins Physical Sciences - Oncology Center, The Johns Hopkins University, Baltimore, MD, USA
| | - Won Jin Ho
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Convergence Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Bloomberg Kimmel Immunology Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA; The Skip Viragh Center for Clinical and Translational Research, Baltimore, MD, USA
| | - Neeha Zaidi
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Convergence Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Bloomberg Kimmel Immunology Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA; The Skip Viragh Center for Clinical and Translational Research, Baltimore, MD, USA
| | - Elizabeth Thompson
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Pathology, Johns Hopkins School of Medicine, Baltimore, MD, USA; The Skip Viragh Center for Clinical and Translational Research, Baltimore, MD, USA
| | - Elizabeth M Jaffee
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Convergence Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Bloomberg Kimmel Immunology Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA; The Skip Viragh Center for Clinical and Translational Research, Baltimore, MD, USA
| | - Laura D Wood
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Pathology, Johns Hopkins School of Medicine, Baltimore, MD, USA; The Skip Viragh Center for Clinical and Translational Research, Baltimore, MD, USA
| | - Elana J Fertig
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Convergence Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Bloomberg Kimmel Immunology Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Applied Mathematics and Statistics, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD, USA; The Skip Viragh Center for Clinical and Translational Research, Baltimore, MD, USA.
| | - Luciane T Kagohara
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Convergence Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Bloomberg Kimmel Immunology Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA; The Skip Viragh Center for Clinical and Translational Research, Baltimore, MD, USA.
| |
Collapse
|
20
|
Kiemen AL, Almagro-Pérez C, Matos V, Forjaz A, Braxton AM, Dequiedt L, Parksong J, Cannon CD, Yuan X, Shin SM, Babu JM, Thompson ED, Cornish TC, Ho WJ, Wood LD, Wu PH, Barrutia AM, Hruban RH, Wirtz D. 3D histology reveals that immune response to pancreatic precancers is heterogeneous and depends on global pancreas structure. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.03.606493. [PMID: 39149369 PMCID: PMC11326156 DOI: 10.1101/2024.08.03.606493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly lethal cancer for which few effective therapies exist. Immunotherapies specifically are ineffective in pancreatic cancer, in part due to its unique stromal and immune microenvironment. Pancreatic intraepithelial neoplasia, or PanIN, is the main precursor lesion to PDAC. Recently it was discovered that PanINs are remarkably abundant in the grossly normal pancreas, suggesting that the vast majority will never progress to cancer. Here, through construction of 48 samples of cm3-sized human pancreas tissue, we profiled the immune microenvironment of 1,476 PanINs in 3D and at single-cell resolution to better understand the early evolution of the pancreatic tumor microenvironment and to determine how inflammation may play a role in cancer progression. We found that bulk pancreatic inflammation strongly correlates to PanIN cell fraction. We found that the immune response around PanINs is highly heterogeneous, with distinct immune hotspots and cold spots that appear and disappear in a span of tens of microns. Immune hotspots generally mark locations of higher grade of dysplasia or locations near acinar atrophy. The immune composition at these hotspots is dominated by naïve, cytotoxic, and regulatory T cells, cancer associated fibroblasts, and tumor associated macrophages, with little similarity to the immune composition around less-inflamed PanINs. By mapping FOXP3+ cells in 3D, we found that regulatory T cells are present at higher density in larger PanIN lesions compared to smaller PanINs, suggesting that the early initiation of PanINs may not exhibit an immunosuppressive response. This analysis demonstrates that while PanINs are common in the pancreases of most individuals, inflammation may play a pivotal role, both at the bulk and the microscopic scale, in demarcating regions of significance in cancer progression.
Collapse
Affiliation(s)
- Ashley L. Kiemen
- Department of Pathology, Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins School of Medicine, Baltimore, MD
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD
- Institute for NanoBioTechnology, Johns Hopkins University
- Department of Functional Anatomy & Evolution, Johns Hopkins School of Medicine, Baltimore, MD
| | - Cristina Almagro-Pérez
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD
- Bioengineering and Aerospace Engineering Department, Universidad Carlos III de Madrid, Leganés, Spain
| | - Valentina Matos
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD
- Bioengineering and Aerospace Engineering Department, Universidad Carlos III de Madrid, Leganés, Spain
| | - Andre Forjaz
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD
| | - Alicia M. Braxton
- Department of Pathology, Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins School of Medicine, Baltimore, MD
| | - Lucie Dequiedt
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD
| | - Jeeun Parksong
- Department of Pathology, Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins School of Medicine, Baltimore, MD
| | - Courtney D. Cannon
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD
| | - Xuan Yuan
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD
| | - Sarah M. Shin
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD
| | - Jaanvi Mahesh Babu
- Department of Pathology, Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins School of Medicine, Baltimore, MD
| | - Elizabeth D. Thompson
- Department of Pathology, Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins School of Medicine, Baltimore, MD
| | - Toby C. Cornish
- Department of Pathology and Data Science Institute, Medical College of Wisconsin, Milwaukee, WI
| | - Won Jin Ho
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD
| | - Laura D. Wood
- Department of Pathology, Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins School of Medicine, Baltimore, MD
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD
| | - Pei-Hsun Wu
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD
- Institute for NanoBioTechnology, Johns Hopkins University
| | - Arrate Muñoz Barrutia
- Bioengineering and Aerospace Engineering Department, Universidad Carlos III de Madrid, Leganés, Spain
- Bioengineering Division, Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| | - Ralph H. Hruban
- Department of Pathology, Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins School of Medicine, Baltimore, MD
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD
| | - Denis Wirtz
- Department of Pathology, Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins School of Medicine, Baltimore, MD
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD
- Institute for NanoBioTechnology, Johns Hopkins University
| |
Collapse
|
21
|
Makohon-Moore AP. Emerging and extensive clonal evolution in the pancreas. Trends Cancer 2024; 10:669-670. [PMID: 38977383 PMCID: PMC11415008 DOI: 10.1016/j.trecan.2024.06.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Accepted: 06/25/2024] [Indexed: 07/10/2024]
Abstract
Pancreatic cancer is one of the most lethal malignancies, yet much remains to be learned regarding how its precursors develop. In a recent Nature publication, Braxton and Kiemen et al. found that the normal, adult pancreas harbors hundreds to thousands of pancreatic cancer precursors evolving by a variety of routes.
Collapse
Affiliation(s)
- Alvin P Makohon-Moore
- Hackensack Meridian Health Center for Discovery and Innovation, Nutley, NJ, USA; Hackensack Meridian School of Medicine, Nutley, NJ, USA; Georgetown University Lombardi Comprehensive Cancer Center, Washington, DC, USA.
| |
Collapse
|
22
|
Kiemen AL, Wu PH, Braxton AM, Cornish TC, Hruban RH, Wood LD, Wirtz D, Zwicker D. Power-law growth models explain incidences and sizes of pancreatic cancer precursor lesions and confirm spatial genomic findings. SCIENCE ADVANCES 2024; 10:eado5103. [PMID: 39058773 PMCID: PMC11277401 DOI: 10.1126/sciadv.ado5103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 06/25/2024] [Indexed: 07/28/2024]
Abstract
Pancreatic ductal adenocarcinoma is a rare but lethal cancer. Recent evidence suggests that pancreatic intraepithelial neoplasia (PanIN), a microscopic precursor lesion that gives rise to pancreatic cancer, is larger and more prevalent than previously believed. Better understanding of the growth-law dynamics of PanINs may improve our ability to understand how a miniscule fraction makes the transition to invasive cancer. Here, using three-dimensional tissue mapping, we analyzed >1000 PanINs and found that lesion size is distributed according to a power law. Our data suggest that in bulk, PanIN size can be predicted by general growth behavior without consideration for the heterogeneity of the pancreatic microenvironment or an individual's age, history, or lifestyle. Our models suggest that intraductal spread and fusing of lesions drive our observed size distribution. This analysis lays the groundwork for future mathematical modeling efforts integrating PanIN incidence, morphology, and molecular features to understand tumorigenesis and demonstrates the utility of combining experimental measurement with dynamic modeling in understanding tumorigenesis.
Collapse
Affiliation(s)
- Ashley L. Kiemen
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Oncology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Pei-Hsun Wu
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Alicia M. Braxton
- Department of Comparative Medicine, Medical University of South Carolina, Charleston, SC, USA
| | - Toby C. Cornish
- Department of Pathology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Ralph H. Hruban
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Oncology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Laura D. Wood
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Oncology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Denis Wirtz
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Oncology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - David Zwicker
- Max Planck Institute for Dynamics and Self-Organization, Göttingen, Germany
| |
Collapse
|
23
|
Mi H, Sivagnanam S, Ho WJ, Zhang S, Bergman D, Deshpande A, Baras AS, Jaffee EM, Coussens LM, Fertig EJ, Popel AS. Computational methods and biomarker discovery strategies for spatial proteomics: a review in immuno-oncology. Brief Bioinform 2024; 25:bbae421. [PMID: 39179248 PMCID: PMC11343572 DOI: 10.1093/bib/bbae421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 07/11/2024] [Accepted: 08/09/2024] [Indexed: 08/26/2024] Open
Abstract
Advancements in imaging technologies have revolutionized our ability to deeply profile pathological tissue architectures, generating large volumes of imaging data with unparalleled spatial resolution. This type of data collection, namely, spatial proteomics, offers invaluable insights into various human diseases. Simultaneously, computational algorithms have evolved to manage the increasing dimensionality of spatial proteomics inherent in this progress. Numerous imaging-based computational frameworks, such as computational pathology, have been proposed for research and clinical applications. However, the development of these fields demands diverse domain expertise, creating barriers to their integration and further application. This review seeks to bridge this divide by presenting a comprehensive guideline. We consolidate prevailing computational methods and outline a roadmap from image processing to data-driven, statistics-informed biomarker discovery. Additionally, we explore future perspectives as the field moves toward interfacing with other quantitative domains, holding significant promise for precision care in immuno-oncology.
Collapse
Affiliation(s)
- Haoyang Mi
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, United States
| | - Shamilene Sivagnanam
- The Knight Cancer Institute, Oregon Health and Science University, Portland, OR 97201, United States
- Department of Cell, Development and Cancer Biology, Oregon Health and Science University, Portland, OR 97201, United States
| | - Won Jin Ho
- Department of Oncology, Johns Hopkins University School of Medicine, MD 21205, United States
- Convergence Institute, Johns Hopkins University, Baltimore, MD 21205, United States
| | - Shuming Zhang
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, United States
| | - Daniel Bergman
- Department of Oncology, Johns Hopkins University School of Medicine, MD 21205, United States
- Convergence Institute, Johns Hopkins University, Baltimore, MD 21205, United States
| | - Atul Deshpande
- Department of Oncology, Johns Hopkins University School of Medicine, MD 21205, United States
- Convergence Institute, Johns Hopkins University, Baltimore, MD 21205, United States
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD 21205, United States
| | - Alexander S Baras
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD 21205, United States
- Department of Pathology, Johns Hopkins University School of Medicine, MD 21205, United States
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21205, United States
| | - Elizabeth M Jaffee
- Department of Oncology, Johns Hopkins University School of Medicine, MD 21205, United States
- Convergence Institute, Johns Hopkins University, Baltimore, MD 21205, United States
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD 21205, United States
| | - Lisa M Coussens
- The Knight Cancer Institute, Oregon Health and Science University, Portland, OR 97201, United States
- Department of Cell, Development and Cancer Biology, Oregon Health and Science University, Portland, OR 97201, United States
- Brenden-Colson Center for Pancreatic Care, Oregon Health and Science University, Portland, OR 97201, United States
| | - Elana J Fertig
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, United States
- Department of Oncology, Johns Hopkins University School of Medicine, MD 21205, United States
- Convergence Institute, Johns Hopkins University, Baltimore, MD 21205, United States
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD 21205, United States
- Department of Applied Mathematics and Statistics, Johns Hopkins University Whiting School of Engineering, Baltimore, MD 21218, United States
| | - Aleksander S Popel
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, United States
- Department of Oncology, Johns Hopkins University School of Medicine, MD 21205, United States
| |
Collapse
|
24
|
Randriamanantsoa SJ, Raich MK, Saur D, Reichert M, Bausch AR. Coexisting mechanisms of luminogenesis in pancreatic cancer-derived organoids. iScience 2024; 27:110299. [PMID: 39055943 PMCID: PMC11269295 DOI: 10.1016/j.isci.2024.110299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 05/02/2024] [Accepted: 06/14/2024] [Indexed: 07/28/2024] Open
Abstract
Lumens are crucial features of the tissue architecture in both the healthy exocrine pancreas, where ducts shuttle enzymes from the acini to the intestine, and in the precancerous lesions of the highly lethal pancreatic ductal adenocarcinoma (PDAC), similarly displaying lumens that can further develop into cyst-like structures. Branched pancreatic-cancer derived organoids capture key architectural features of both the healthy and diseased pancreas, including lumens. However, their transition from a solid mass of cells to a hollow tissue remains insufficiently explored. Here, we show that organoids display two orthogonal but complementary lumen formation mechanisms: one relying on fluid intake for multiple microlumen nucleation, swelling and fusion, and the other involving the death of a central cell population, thereby hollowing out cavities. These results shed further light on the processes of luminogenesis, deepening our understanding of the early formation of PDAC precancerous lesions, including cystic neoplasia.
Collapse
Affiliation(s)
- Samuel J. Randriamanantsoa
- Technical University of Munich, TUM School of Natural Sciences, Department of Bioscience, Chair for Cellular Biophysics E27, 85748 Garching, Germany
- Technical University of Munich, Center for Functional Protein Assemblies (CPA), 85748 Garching, Germany
- Technical University of Munich, Center for Organoid Systems and Tissue Engineering (COS), 85748 Garching, Germany
| | - Marion K. Raich
- Technical University of Munich, TUM School of Natural Sciences, Department of Bioscience, Chair for Cellular Biophysics E27, 85748 Garching, Germany
- Technical University of Munich, Center for Functional Protein Assemblies (CPA), 85748 Garching, Germany
- Technical University of Munich, Center for Organoid Systems and Tissue Engineering (COS), 85748 Garching, Germany
| | - Dieter Saur
- Technical University of Munich, School of Medicine, Klinikum rechts der Isar, Medical Clinic and Polyclinic II, 81675 Munich, Germany
- German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), Partner site Munich, 69120 Heidelberg, Germany
| | - Maximilian Reichert
- Technical University of Munich, Center for Functional Protein Assemblies (CPA), 85748 Garching, Germany
- Technical University of Munich, Center for Organoid Systems and Tissue Engineering (COS), 85748 Garching, Germany
- Technical University of Munich, School of Medicine, Klinikum rechts der Isar, Medical Clinic and Polyclinic II, 81675 Munich, Germany
- German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), Partner site Munich, 69120 Heidelberg, Germany
- Technical University of Munich, Klinikum rechts der Isar, Medical Clinic and Polyclinic II, Translational Pancreatic Cancer Research Center, 81675 Munich, Germany
| | - Andreas R. Bausch
- Technical University of Munich, TUM School of Natural Sciences, Department of Bioscience, Chair for Cellular Biophysics E27, 85748 Garching, Germany
- Technical University of Munich, Center for Functional Protein Assemblies (CPA), 85748 Garching, Germany
- Technical University of Munich, Center for Organoid Systems and Tissue Engineering (COS), 85748 Garching, Germany
| |
Collapse
|
25
|
Du W, Zhou B, Forjaz A, Shin SM, Wu F, Crawford AJ, Nair PR, Johnston AC, West-Foyle H, Tang A, Kim D, Fan R, Kiemen AL, Wu PH, Phillip JM, Ho WJ, Sanin DE, Wirtz D. High-motility pro-tumorigenic monocytes drive macrophage enrichment in the tumor microenvironment. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.16.603739. [PMID: 39071324 PMCID: PMC11275814 DOI: 10.1101/2024.07.16.603739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
Enrichment of tumor-associated macrophages (TAMΦs) in the tumor microenvironment correlates with worse clinical outcomes in triple-negative breast cancer (TNBC) patients, prompting the development of therapies to inhibit TAMΦ infiltration. However, the lackluster efficacy of CCL2-based chemotaxis blockade in clinical trials suggests that a new understanding of monocyte/macrophage infiltration may be necessary. Here we demonstrate that random migration, and not only chemotaxis, drives macrophage tumor infiltration. We identified tumor- associated monocytes (TAMos) that display a dramatically enhanced migration capability, induced rapidly by the tumor microenvironment, that drives effective tumor infiltration, in contrast to low-motility differentiated macrophages. TAMo, not TAMΦ, promotes cancer cell proliferation through activation of the MAPK pathway. IL-6 secreted both by cancer cells and TAMo themselves enhances TAMo migration by increasing dendritic protrusion dynamics and myosin- based contractility via the JAK2/STAT3 signaling pathway. Independent from CCL2 mediated chemotaxis, IL-6 driven enhanced migration and pro-proliferative effect of TAMo were validated in a syngeneic TNBC mouse model. Depletion of IL-6 in cancer cells significantly attenuated monocyte infiltration and reversed TAMo-induced cancer cell proliferation. This work reveals the critical role random migration plays in monocyte driven TAMΦ enrichment in a tumor and pinpoints IL-6 as a potential therapeutic target in combination with CCL2 to ameliorate current strategies against TAMΦ infiltration.
Collapse
|
26
|
Gou F, Liu J, Xiao C, Wu J. Research on Artificial-Intelligence-Assisted Medicine: A Survey on Medical Artificial Intelligence. Diagnostics (Basel) 2024; 14:1472. [PMID: 39061610 PMCID: PMC11275417 DOI: 10.3390/diagnostics14141472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 07/04/2024] [Accepted: 07/05/2024] [Indexed: 07/28/2024] Open
Abstract
With the improvement of economic conditions and the increase in living standards, people's attention in regard to health is also continuously increasing. They are beginning to place their hopes on machines, expecting artificial intelligence (AI) to provide a more humanized medical environment and personalized services, thus greatly expanding the supply and bridging the gap between resource supply and demand. With the development of IoT technology, the arrival of the 5G and 6G communication era, and the enhancement of computing capabilities in particular, the development and application of AI-assisted healthcare have been further promoted. Currently, research on and the application of artificial intelligence in the field of medical assistance are continuously deepening and expanding. AI holds immense economic value and has many potential applications in regard to medical institutions, patients, and healthcare professionals. It has the ability to enhance medical efficiency, reduce healthcare costs, improve the quality of healthcare services, and provide a more intelligent and humanized service experience for healthcare professionals and patients. This study elaborates on AI development history and development timelines in the medical field, types of AI technologies in healthcare informatics, the application of AI in the medical field, and opportunities and challenges of AI in the field of medicine. The combination of healthcare and artificial intelligence has a profound impact on human life, improving human health levels and quality of life and changing human lifestyles.
Collapse
Affiliation(s)
- Fangfang Gou
- State Key Laboratory of Public Big Data, College of Computer Science and Technology, Guizhou University, Guiyang 550025, China
| | - Jun Liu
- The Second People's Hospital of Huaihua, Huaihua 418000, China
| | - Chunwen Xiao
- The Second People's Hospital of Huaihua, Huaihua 418000, China
| | - Jia Wu
- State Key Laboratory of Public Big Data, College of Computer Science and Technology, Guizhou University, Guiyang 550025, China
- Research Center for Artificial Intelligence, Monash University, Melbourne, Clayton, VIC 3800, Australia
| |
Collapse
|
27
|
Kiemen AL, Dequiedt L, Shen Y, Zhu Y, Matos-Romero V, Forjaz A, Campbell K, Dhana W, Cornish T, Braxton AM, Wu P, Fishman EK, Wood LD, Wirtz D, Hruban RH. PanIN or IPMN? Redefining Lesion Size in 3 Dimensions. Am J Surg Pathol 2024; 48:839-845. [PMID: 38764379 PMCID: PMC11189722 DOI: 10.1097/pas.0000000000002245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/21/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) develops from 2 known precursor lesions: a majority (∼85%) develops from pancreatic intraepithelial neoplasia (PanIN), and a minority develops from intraductal papillary mucinous neoplasms (IPMNs). Clinical classification of PanIN and IPMN relies on a combination of low-resolution, 3-dimensional (D) imaging (computed tomography, CT), and high-resolution, 2D imaging (histology). The definitions of PanIN and IPMN currently rely heavily on size. IPMNs are defined as macroscopic: generally >1.0 cm and visible in CT, and PanINs are defined as microscopic: generally <0.5 cm and not identifiable in CT. As 2D evaluation fails to take into account 3D structures, we hypothesized that this classification would fail in evaluation of high-resolution, 3D images. To characterize the size and prevalence of PanINs in 3D, 47 thick slabs of pancreas were harvested from grossly normal areas of pancreatic resections, excluding samples from individuals with a diagnosis of an IPMN. All patients but one underwent preoperative CT scans. Through construction of cellular resolution 3D maps, we identified >1400 ductal precursor lesions that met the 2D histologic size criteria of PanINs. We show that, when 3D space is considered, 25 of these lesions can be digitally sectioned to meet the 2D histologic size criterion of IPMN. Re-evaluation of the preoperative CT images of individuals found to possess these large precursor lesions showed that nearly half are visible on imaging. These findings demonstrate that the clinical classification of PanIN and IPMN fails in evaluation of high-resolution, 3D images, emphasizing the need for re-evaluation of classification guidelines that place significant weight on 2D assessment of 3D structures.
Collapse
Affiliation(s)
- Ashley L. Kiemen
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins School of Medicine, Baltimore, MD
- Department of Oncology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins School of Medicine, Baltimore, MD
| | - Lucie Dequiedt
- Department of Chemical & Biomolecular Engineering, Johns Hopkins University, Baltimore, MD
| | - Yu Shen
- Department of Chemical & Biomolecular Engineering, Johns Hopkins University, Baltimore, MD
| | - Yutong Zhu
- Department of Chemical & Biomolecular Engineering, Johns Hopkins University, Baltimore, MD
| | - Valentina Matos-Romero
- Department of Chemical & Biomolecular Engineering, Johns Hopkins University, Baltimore, MD
| | - André Forjaz
- Department of Chemical & Biomolecular Engineering, Johns Hopkins University, Baltimore, MD
| | - Kurtis Campbell
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins School of Medicine, Baltimore, MD
| | - Will Dhana
- Department of Chemical & Biomolecular Engineering, Johns Hopkins University, Baltimore, MD
| | - Toby Cornish
- Department of Pathology, University of Colorado School of Medicine, Aurora, CO
| | - Alicia M. Braxton
- Department of Comparative Medicine, Medical University of South Carolina, Charleston, SC
| | - PeiHsun Wu
- Department of Chemical & Biomolecular Engineering, Johns Hopkins University, Baltimore, MD
| | - Elliot K. Fishman
- Department of Radiology and Radiological Science, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins School of Medicine, Baltimore, MD
| | - Laura D. Wood
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins School of Medicine, Baltimore, MD
- Department of Oncology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins School of Medicine, Baltimore, MD
| | - Denis Wirtz
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins School of Medicine, Baltimore, MD
- Department of Oncology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins School of Medicine, Baltimore, MD
- Department of Chemical & Biomolecular Engineering, Johns Hopkins University, Baltimore, MD
| | - Ralph H. Hruban
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins School of Medicine, Baltimore, MD
- Department of Oncology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins School of Medicine, Baltimore, MD
| |
Collapse
|
28
|
Liu Y, Uttam S. Perspective on quantitative phase imaging to improve precision cancer medicine. JOURNAL OF BIOMEDICAL OPTICS 2024; 29:S22705. [PMID: 38584967 PMCID: PMC10996848 DOI: 10.1117/1.jbo.29.s2.s22705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 03/03/2024] [Accepted: 03/15/2024] [Indexed: 04/09/2024]
Abstract
Significance Quantitative phase imaging (QPI) offers a label-free approach to non-invasively characterize cellular processes by exploiting their refractive index based intrinsic contrast. QPI captures this contrast by translating refractive index associated phase shifts into intensity-based quantifiable data with nanoscale sensitivity. It holds significant potential for advancing precision cancer medicine by providing quantitative characterization of the biophysical properties of cells and tissue in their natural states. Aim This perspective aims to discuss the potential of QPI to increase our understanding of cancer development and its response to therapeutics. It also explores new developments in QPI methods towards advancing personalized cancer therapy and early detection. Approach We begin by detailing the technical advancements of QPI, examining its implementations across transmission and reflection geometries and phase retrieval methods, both interferometric and non-interferometric. The focus then shifts to QPI's applications in cancer research, including dynamic cell mass imaging for drug response assessment, cancer risk stratification, and in-vivo tissue imaging. Results QPI has emerged as a crucial tool in precision cancer medicine, offering insights into tumor biology and treatment efficacy. Its sensitivity to detecting nanoscale changes holds promise for enhancing cancer diagnostics, risk assessment, and prognostication. The future of QPI is envisioned in its integration with artificial intelligence, morpho-dynamics, and spatial biology, broadening its impact in cancer research. Conclusions QPI presents significant potential in advancing precision cancer medicine and redefining our approach to cancer diagnosis, monitoring, and treatment. Future directions include harnessing high-throughput dynamic imaging, 3D QPI for realistic tumor models, and combining artificial intelligence with multi-omics data to extend QPI's capabilities. As a result, QPI stands at the forefront of cancer research and clinical application in cancer care.
Collapse
Affiliation(s)
- Yang Liu
- University of Illinois Urbana-Champaign, Beckman Institute for Advanced Science and Technology, Cancer Center at Illinois, Department of Bioengineering, Department of Electrical and Computer Engineering, Urbana, Illinois, United States
- University of Pittsburgh, Departments of Medicine and Bioengineering, Pittsburgh, Pennsylvania, United States
| | - Shikhar Uttam
- University of Pittsburgh, Department of Computational and Systems Biology, Pittsburgh, Pennsylvania, United States
| |
Collapse
|
29
|
Song AH, Williams M, Williamson DFK, Chow SSL, Jaume G, Gao G, Zhang A, Chen B, Baras AS, Serafin R, Colling R, Downes MR, Farré X, Humphrey P, Verrill C, True LD, Parwani AV, Liu JTC, Mahmood F. Analysis of 3D pathology samples using weakly supervised AI. Cell 2024; 187:2502-2520.e17. [PMID: 38729110 PMCID: PMC11168832 DOI: 10.1016/j.cell.2024.03.035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 01/15/2024] [Accepted: 03/25/2024] [Indexed: 05/12/2024]
Abstract
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.
Collapse
Affiliation(s)
- Andrew H Song
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; Cancer Program, Broad Institute of Harvard and MIT, Cambridge, MA, USA; Data Science Program, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Mane Williams
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Cancer Program, Broad Institute of Harvard and MIT, Cambridge, MA, USA; Data Science Program, Dana-Farber Cancer Institute, Boston, MA, USA; Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| | - Drew F K Williamson
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; Cancer Program, Broad Institute of Harvard and MIT, Cambridge, MA, USA; Data Science Program, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Sarah S L Chow
- Department of Mechanical Engineering, Bioengineering, and Laboratory Medicine & Pathology, University of Washington, Seattle, WA, USA
| | - Guillaume Jaume
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; Cancer Program, Broad Institute of Harvard and MIT, Cambridge, MA, USA; Data Science Program, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Gan Gao
- Department of Mechanical Engineering, Bioengineering, and Laboratory Medicine & Pathology, University of Washington, Seattle, WA, USA
| | - Andrew Zhang
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Cancer Program, Broad Institute of Harvard and MIT, Cambridge, MA, USA; Data Science Program, Dana-Farber Cancer Institute, Boston, MA, USA; Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Bowen Chen
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; Cancer Program, Broad Institute of Harvard and MIT, Cambridge, MA, USA; Data Science Program, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Alexander S Baras
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Robert Serafin
- Department of Mechanical Engineering, Bioengineering, and Laboratory Medicine & Pathology, University of Washington, Seattle, WA, USA
| | - Richard Colling
- Nuffield Department of Surgical Sciences, University of Oxford, UK; Department of Cellular Pathology, Oxford University Hospitals NHS Foundations Trust, John Radcliffe Hospital, Oxford, UK
| | - Michelle R Downes
- Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON, Canada
| | - Xavier Farré
- Public Health Agency of Catalonia, Lleida, Spain
| | - Peter Humphrey
- Department of Pathology, Yale School of Medicine, New Haven, CT, USA
| | - Clare Verrill
- Nuffield Department of Surgical Sciences, University of Oxford, UK; Department of Cellular Pathology, Oxford University Hospitals NHS Foundations Trust, John Radcliffe Hospital, Oxford, UK; NIHR Oxford Biomedical Research Centre, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Lawrence D True
- Department of Laboratory Medicine & Pathology, University of Washington School of Medicine, Seattle, WA, USA
| | - Anil V Parwani
- Department of Pathology, The Ohio State University, Columbus, OH, USA
| | - Jonathan T C Liu
- Department of Mechanical Engineering, Bioengineering, and Laboratory Medicine & Pathology, University of Washington, Seattle, WA, USA.
| | - Faisal Mahmood
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; Cancer Program, Broad Institute of Harvard and MIT, Cambridge, MA, USA; Data Science Program, Dana-Farber Cancer Institute, Boston, MA, USA.
| |
Collapse
|
30
|
Braxton AM, Kiemen AL, Grahn MP, Forjaz A, Parksong J, Mahesh Babu J, Lai J, Zheng L, Niknafs N, Jiang L, Cheng H, Song Q, Reichel R, Graham S, Damanakis AI, Fischer CG, Mou S, Metz C, Granger J, Liu XD, Bachmann N, Zhu Y, Liu Y, Almagro-Pérez C, Jiang AC, Yoo J, Kim B, Du S, Foster E, Hsu JY, Rivera PA, Chu LC, Liu F, Fishman EK, Yuille A, Roberts NJ, Thompson ED, Scharpf RB, Cornish TC, Jiao Y, Karchin R, Hruban RH, Wu PH, Wirtz D, Wood LD. 3D genomic mapping reveals multifocality of human pancreatic precancers. Nature 2024; 629:679-687. [PMID: 38693266 DOI: 10.1038/s41586-024-07359-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 03/26/2024] [Indexed: 05/03/2024]
Abstract
Pancreatic intraepithelial neoplasias (PanINs) are the most common precursors of pancreatic cancer, but their small size and inaccessibility in humans make them challenging to study1. Critically, the number, dimensions and connectivity of human PanINs remain largely unknown, precluding important insights into early cancer development. Here, we provide a microanatomical survey of human PanINs by analysing 46 large samples of grossly normal human pancreas with a machine-learning pipeline for quantitative 3D histological reconstruction at single-cell resolution. To elucidate genetic relationships between and within PanINs, we developed a workflow in which 3D modelling guides multi-region microdissection and targeted and whole-exome sequencing. From these samples, we calculated a mean burden of 13 PanINs per cm3 and extrapolated that the normal intact adult pancreas harbours hundreds of PanINs, almost all with oncogenic KRAS hotspot mutations. We found that most PanINs originate as independent clones with distinct somatic mutation profiles. Some spatially continuous PanINs were found to contain multiple KRAS mutations; computational and in situ analyses demonstrated that different KRAS mutations localize to distinct cell subpopulations within these neoplasms, indicating their polyclonal origins. The extensive multifocality and genetic heterogeneity of PanINs raises important questions about mechanisms that drive precancer initiation and confer differential progression risk in the human pancreas. This detailed 3D genomic mapping of molecular alterations in human PanINs provides an empirical foundation for early detection and rational interception of pancreatic cancer.
Collapse
Affiliation(s)
- Alicia M Braxton
- Department of Pathology, Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Comparative Medicine, Medical University of South Carolina, Charleston, SC, USA
| | - Ashley L Kiemen
- Department of Pathology, Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Mia P Grahn
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - André Forjaz
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Jeeun Parksong
- Department of Pathology, Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jaanvi Mahesh Babu
- Department of Pathology, Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jiaying Lai
- Institute for Computational Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Lily Zheng
- Institute for Computational Medicine, Johns Hopkins University, Baltimore, MD, USA
- McKusick-Nathans Institute for Genetic Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Noushin Niknafs
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Liping Jiang
- State Key Laboratory of Molecular Oncology, National Cancer Center, National Clinical Research Center for Cancer and Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Haixia Cheng
- State Key Laboratory of Molecular Oncology, National Cancer Center, National Clinical Research Center for Cancer and Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Qianqian Song
- State Key Laboratory of Molecular Oncology, National Cancer Center, National Clinical Research Center for Cancer and Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Rebecca Reichel
- Department of Pathology, Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sarah Graham
- Department of Pathology, Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Alexander I Damanakis
- Department of Pathology, Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Catherine G Fischer
- Department of Pathology, Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Stephanie Mou
- Department of Pathology, Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Cameron Metz
- Department of Pathology, Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Julie Granger
- Department of Pathology, Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Xiao-Ding Liu
- Department of Pathology, Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Pathology, Molecular Pathology Research Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Niklas Bachmann
- Department of Pathology, Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Yutong Zhu
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - YunZhou Liu
- Institute for Computational Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Cristina Almagro-Pérez
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Ann Chenyu Jiang
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Jeonghyun Yoo
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Bridgette Kim
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Scott Du
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Eli Foster
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Jocelyn Y Hsu
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Paula Andreu Rivera
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Linda C Chu
- Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Fengze Liu
- Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Elliot K Fishman
- Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Alan Yuille
- Department of Computer Science, Johns Hopkins University, Baltimore, MD, USA
| | - Nicholas J Roberts
- Department of Pathology, Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Elizabeth D Thompson
- Department of Pathology, Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Robert B Scharpf
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Toby C Cornish
- Department of Pathology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Yuchen Jiao
- State Key Laboratory of Molecular Oncology, National Cancer Center, National Clinical Research Center for Cancer and Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
- Institute of Cancer Research, Henan Academy of Innovations in Medical Science, Zhengzhou, China.
| | - Rachel Karchin
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- McKusick-Nathans Institute for Genetic Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Ralph H Hruban
- Department of Pathology, Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Pei-Hsun Wu
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Denis Wirtz
- Department of Pathology, Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA.
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Laura D Wood
- Department of Pathology, Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Convergence Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
31
|
Daly AC, Cambuli F, Äijö T, Lötstedt B, Marjanovic N, Kuksenko O, Smith-Erb M, Fernandez S, Domovic D, Van Wittenberghe N, Drokhlyansky E, Griffin GK, Phatnani H, Bonneau R, Regev A, Vickovic S. Tissue and cellular spatiotemporal dynamics in colon aging. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.22.590125. [PMID: 38712088 PMCID: PMC11071407 DOI: 10.1101/2024.04.22.590125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
Tissue structure and molecular circuitry in the colon can be profoundly impacted by systemic age-related effects, but many of the underlying molecular cues remain unclear. Here, we built a cellular and spatial atlas of the colon across three anatomical regions and 11 age groups, encompassing ~1,500 mouse gut tissues profiled by spatial transcriptomics and ~400,000 single nucleus RNA-seq profiles. We developed a new computational framework, cSplotch, which learns a hierarchical Bayesian model of spatially resolved cellular expression associated with age, tissue region, and sex, by leveraging histological features to share information across tissue samples and data modalities. Using this model, we identified cellular and molecular gradients along the adult colonic tract and across the main crypt axis, and multicellular programs associated with aging in the large intestine. Our multi-modal framework for the investigation of cell and tissue organization can aid in the understanding of cellular roles in tissue-level pathology.
Collapse
Affiliation(s)
- Aidan C. Daly
- New York Genome Center, New York, NY, USA
- Center for Computational Biology, Flatiron Institute, New York, NY, USA
| | | | - Tarmo Äijö
- Center for Computational Biology, Flatiron Institute, New York, NY, USA
| | - Britta Lötstedt
- New York Genome Center, New York, NY, USA
- Klarman Cell Observatory Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Science for Life Laboratory, Department of Gene Technology, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Nemanja Marjanovic
- Klarman Cell Observatory Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Olena Kuksenko
- Klarman Cell Observatory Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
| | | | | | | | | | - Eugene Drokhlyansky
- Klarman Cell Observatory Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Gabriel K Griffin
- Klarman Cell Observatory Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Pathology, Brigham and Women’s Hospital, Boston, MA, USA
| | - Hemali Phatnani
- New York Genome Center, New York, NY, USA
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
| | - Richard Bonneau
- Center for Computational Biology, Flatiron Institute, New York, NY, USA
- Center for Data Science, New York University, New York, NY, USA
- Current address: Genentech, 1 DNA Way, South San Francisco, CA, USA
| | - Aviv Regev
- Klarman Cell Observatory Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
- Current address: Genentech, 1 DNA Way, South San Francisco, CA, USA
| | - Sanja Vickovic
- New York Genome Center, New York, NY, USA
- Klarman Cell Observatory Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Biomedical Engineering and Herbert Irving Institute for Cancer Dynamics, Columbia University, New York, NY, USA
- Science for Life Laboratory, Department of Immunology, Genetics and Pathology, Beijer Laboratory for Gene and Neuro Research, Uppsala University, Uppsala, Sweden
| |
Collapse
|
32
|
Wang L, Li M, Hwang TH. The 3D Revolution in Cancer Discovery. Cancer Discov 2024; 14:625-629. [PMID: 38571426 DOI: 10.1158/2159-8290.cd-23-1499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2024]
Abstract
SUMMARY The transition from 2D to 3D spatial profiling marks a revolutionary era in cancer research, offering unprecedented potential to enhance cancer diagnosis and treatment. This commentary outlines the experimental and computational advancements and challenges in 3D spatial molecular profiling, underscoring the innovation needed in imaging tools, software, artificial intelligence, and machine learning to overcome implementation hurdles and harness the full potential of 3D analysis in the field.
Collapse
Affiliation(s)
- Linghua Wang
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
- UTHealth Houston Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Mingyao Li
- Statistical Center for Single-Cell and Spatial Genomics, Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Tae Hyun Hwang
- Department of Artificial Intelligence and Informatics, Mayo Clinic, Jacksonville, Florida
| |
Collapse
|
33
|
Dequiedt L, Forjaz A, Lo JO, McCarty O, Wu PH, Rosenberg A, Wirtz D, Kiemen A. Three-dimensional reconstruction of fetal rhesus macaque kidneys at single-cell resolution reveals complex inter-relation of structures. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.12.07.570622. [PMID: 38106004 PMCID: PMC10723390 DOI: 10.1101/2023.12.07.570622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
Abstract
Kidneys are among the most structurally complex organs in the body. Their architecture is critical to ensure proper function and is often impacted by diseases such as diabetes and hypertension. Understanding the spatial interplay between the different structures of the nephron and renal vasculature is crucial. Recent efforts have demonstrated the value of three-dimensional (3D) imaging in revealing new insights into the various components of the kidney; however, these studies used antibodies or autofluorescence to detect structures and so were limited in their ability to compare the many subtle structures of the kidney at once. Here, through 3D reconstruction of fetal rhesus macaque kidneys at cellular resolution, we demonstrate the power of deep learning in exhaustively labelling seventeen microstructures of the kidney. Using these tissue maps, we interrogate the spatial distribution and spatial correlation of the glomeruli, renal arteries, and the nephron. This work demonstrates the power of deep learning applied to 3D tissue images to improve our ability to compare many microanatomical structures at once, paving the way for further works investigating renal pathologies.
Collapse
Affiliation(s)
- Lucie Dequiedt
- Department of Chemical & Biomolecular Engineering, Johns Hopkins University
| | - André Forjaz
- Department of Chemical & Biomolecular Engineering, Johns Hopkins University
| | - Jamie O Lo
- Department of Obstetrics and Gynecology, Oregon Health and Sciences University
- Division of Reproductive and Developmental Sciences, Oregon National Primate Research Center
| | - Owen McCarty
- Department of Biomedical Engineering, Oregon Health and Sciences University
| | - Pei-Hsun Wu
- Department of Chemical & Biomolecular Engineering, Johns Hopkins University
- Institute for NanoBioTechnology, Johns Hopkins University
| | - Avi Rosenberg
- Department of Pathology, Johns Hopkins School of Medicine
| | - Denis Wirtz
- Department of Chemical & Biomolecular Engineering, Johns Hopkins University
- Institute for NanoBioTechnology, Johns Hopkins University
- Department of Pathology, Johns Hopkins School of Medicine
- Department of Oncology, Johns Hopkins School of Medicine
| | - Ashley Kiemen
- Institute for NanoBioTechnology, Johns Hopkins University
- Department of Pathology, Johns Hopkins School of Medicine
- Department of Oncology, Johns Hopkins School of Medicine
| |
Collapse
|
34
|
Ekvall M, Bergenstråhle L, Andersson A, Czarnewski P, Olegård J, Käll L, Lundeberg J. Spatial landmark detection and tissue registration with deep learning. Nat Methods 2024; 21:673-679. [PMID: 38438615 PMCID: PMC11009106 DOI: 10.1038/s41592-024-02199-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 01/30/2024] [Indexed: 03/06/2024]
Abstract
Spatial landmarks are crucial in describing histological features between samples or sites, tracking regions of interest in microscopy, and registering tissue samples within a common coordinate framework. Although other studies have explored unsupervised landmark detection, existing methods are not well-suited for histological image data as they often require a large number of images to converge, are unable to handle nonlinear deformations between tissue sections and are ineffective for z-stack alignment, other modalities beyond image data or multimodal data. We address these challenges by introducing effortless landmark detection, a new unsupervised landmark detection and registration method using neural-network-guided thin-plate splines. Our proposed method is evaluated on a diverse range of datasets including histology and spatially resolved transcriptomics, demonstrating superior performance in both accuracy and stability compared to existing approaches.
Collapse
Affiliation(s)
- Markus Ekvall
- Science for Life Laboratory, School of Engineering Sciences in Chemistry, Biotechnology and Health, Royal Institute of Technology - KTH, Solna, Sweden.
| | - Ludvig Bergenstråhle
- Science for Life Laboratory, School of Engineering Sciences in Chemistry, Biotechnology and Health, Royal Institute of Technology - KTH, Solna, Sweden
| | - Alma Andersson
- Science for Life Laboratory, School of Engineering Sciences in Chemistry, Biotechnology and Health, Royal Institute of Technology - KTH, Solna, Sweden
| | - Paulo Czarnewski
- Science for Life Laboratory, School of Engineering Sciences in Chemistry, Biotechnology and Health, Royal Institute of Technology - KTH, Solna, Sweden
| | - Johannes Olegård
- Department of Computer and Systems Sciences, Stockholm University, Kista, Sweden
| | - Lukas Käll
- Science for Life Laboratory, School of Engineering Sciences in Chemistry, Biotechnology and Health, Royal Institute of Technology - KTH, Solna, Sweden
| | - Joakim Lundeberg
- Science for Life Laboratory, School of Engineering Sciences in Chemistry, Biotechnology and Health, Royal Institute of Technology - KTH, Solna, Sweden.
| |
Collapse
|
35
|
Yoshikawa AL, Omura T, Takahashi-Kanemitsu A, Susaki EA. Blueprints from plane to space: outlook of next-generation three-dimensional histopathology. Cancer Sci 2024; 115:1029-1038. [PMID: 38316137 PMCID: PMC11006986 DOI: 10.1111/cas.16095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Revised: 01/02/2024] [Accepted: 01/16/2024] [Indexed: 02/07/2024] Open
Abstract
Here, we summarize the literature relevant to recent advances in three-dimensional (3D) histopathology in relation to clinical oncology, highlighting serial sectioning, tissue clearing, light-sheet microscopy, and digital image analysis with artificial intelligence. We look forward to a future where 3D histopathology expands our understanding of human pathophysiology and improves patient care through cross-disciplinary collaboration and innovation.
Collapse
Affiliation(s)
- Akira Leon Yoshikawa
- Department of Biochemistry and Systems Biomedicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
- Department of Pathology Informatics, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
- Department of Pathology, Kameda Medical Center, Chiba, Japan
| | - Takaki Omura
- Department of Biochemistry and Systems Biomedicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
- Department of Neurosurgery and Neuro-Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Atsushi Takahashi-Kanemitsu
- Department of Biochemistry and Systems Biomedicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Etsuo A Susaki
- Department of Biochemistry and Systems Biomedicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| |
Collapse
|
36
|
Swanton C, Bernard E, Abbosh C, André F, Auwerx J, Balmain A, Bar-Sagi D, Bernards R, Bullman S, DeGregori J, Elliott C, Erez A, Evan G, Febbraio MA, Hidalgo A, Jamal-Hanjani M, Joyce JA, Kaiser M, Lamia K, Locasale JW, Loi S, Malanchi I, Merad M, Musgrave K, Patel KJ, Quezada S, Wargo JA, Weeraratna A, White E, Winkler F, Wood JN, Vousden KH, Hanahan D. Embracing cancer complexity: Hallmarks of systemic disease. Cell 2024; 187:1589-1616. [PMID: 38552609 DOI: 10.1016/j.cell.2024.02.009] [Citation(s) in RCA: 73] [Impact Index Per Article: 73.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 01/25/2024] [Accepted: 02/08/2024] [Indexed: 04/02/2024]
Abstract
The last 50 years have witnessed extraordinary developments in understanding mechanisms of carcinogenesis, synthesized as the hallmarks of cancer. Despite this logical framework, our understanding of the molecular basis of systemic manifestations and the underlying causes of cancer-related death remains incomplete. Looking forward, elucidating how tumors interact with distant organs and how multifaceted environmental and physiological parameters impinge on tumors and their hosts will be crucial for advances in preventing and more effectively treating human cancers. In this perspective, we discuss complexities of cancer as a systemic disease, including tumor initiation and promotion, tumor micro- and immune macro-environments, aging, metabolism and obesity, cancer cachexia, circadian rhythms, nervous system interactions, tumor-related thrombosis, and the microbiome. Model systems incorporating human genetic variation will be essential to decipher the mechanistic basis of these phenomena and unravel gene-environment interactions, providing a modern synthesis of molecular oncology that is primed to prevent cancers and improve patient quality of life and cancer outcomes.
Collapse
Affiliation(s)
- Charles Swanton
- The Francis Crick Institute, London, UK; Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK.
| | - Elsa Bernard
- The Francis Crick Institute, London, UK; INSERM U981, Gustave Roussy, Villejuif, France
| | | | - Fabrice André
- INSERM U981, Gustave Roussy, Villejuif, France; Department of Medical Oncology, Gustave Roussy, Villejuif, France; Paris Saclay University, Kremlin-Bicetre, France
| | - Johan Auwerx
- Laboratory of Integrative Systems Physiology, Ecole Polytechnique Federale de Lausanne, Lausanne, Switzerland
| | - Allan Balmain
- UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, CA, USA
| | | | - René Bernards
- Division of Molecular Carcinogenesis, Oncode Institute, the Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Susan Bullman
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - James DeGregori
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | | | - Ayelet Erez
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Gerard Evan
- The Francis Crick Institute, London, UK; Kings College London, London, UK
| | - Mark A Febbraio
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | - Andrés Hidalgo
- Department of Immunobiology, Yale University, New Haven, CT 06519, USA; Area of Cardiovascular Regeneration, Centro Nacional de Investigaciones Cardiovasculares, 28029 Madrid, Spain
| | - Mariam Jamal-Hanjani
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | - Johanna A Joyce
- Department of Oncology, Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland
| | | | - Katja Lamia
- Department of Molecular Medicine, Scripps Research Institute, La Jolla, CA, USA
| | - Jason W Locasale
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC, USA; Department of Molecular and Structural Biochemistry, North Carolina State University, Raleigh, NC, USA
| | - Sherene Loi
- Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia; The Sir Department of Medical Oncology, The University of Melbourne, Parkville, VIC, Australia
| | | | - Miriam Merad
- Department of immunology and immunotherapy, Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kathryn Musgrave
- Translational and Clinical Research Institute, Newcastle University, Newcastle, UK; Department of Haematology, The Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Ketan J Patel
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - Sergio Quezada
- Cancer Immunology Unit, Research Department of Haematology, University College London Cancer Institute, London, UK
| | - Jennifer A Wargo
- Department of Surgical Oncology, Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ashani Weeraratna
- Sidney Kimmel Cancer Center, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Eileen White
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA; Ludwig Princeton Branch, Ludwig Institute for Cancer Research, Princeton, NJ, USA
| | - Frank Winkler
- Neurology Clinic and National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany; Clinical Cooperation Unit Neuro-oncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - John N Wood
- Molecular Nociception Group, WIBR, University College London, London, UK
| | | | - Douglas Hanahan
- Lausanne Branch, Ludwig Institute for Cancer Research, Lausanne, Switzerland; Swiss institute for Experimental Cancer Research (ISREC), EPFL, Lausanne, Switzerland; Agora Translational Cancer Research Center, Lausanne, Switzerland.
| |
Collapse
|
37
|
Joshi S, Forjaz A, Han KS, Shen Y, Queiroga V, Xenes D, Matelsk J, Wester B, Barrutia AM, Kiemen AL, Wu PH, Wirtz D. Generative interpolation and restoration of images using deep learning for improved 3D tissue mapping. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.07.583909. [PMID: 38496512 PMCID: PMC10942457 DOI: 10.1101/2024.03.07.583909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
The development of novel imaging platforms has improved our ability to collect and analyze large three-dimensional (3D) biological imaging datasets. Advances in computing have led to an ability to extract complex spatial information from these data, such as the composition, morphology, and interactions of multi-cellular structures, rare events, and integration of multi-modal features combining anatomical, molecular, and transcriptomic (among other) information. Yet, the accuracy of these quantitative results is intrinsically limited by the quality of the input images, which can contain missing or damaged regions, or can be of poor resolution due to mechanical, temporal, or financial constraints. In applications ranging from intact imaging (e.g. light-sheet microscopy and magnetic resonance imaging) to sectioning based platforms (e.g. serial histology and serial section transmission electron microscopy), the quality and resolution of imaging data has become paramount. Here, we address these challenges by leveraging frame interpolation for large image motion (FILM), a generative AI model originally developed for temporal interpolation, for spatial interpolation of a range of 3D image types. Comparative analysis demonstrates the superiority of FILM over traditional linear interpolation to produce functional synthetic images, due to its ability to better preserve biological information including microanatomical features and cell counts, as well as image quality, such as contrast, variance, and luminance. FILM repairs tissue damages in images and reduces stitching artifacts. We show that FILM can decrease imaging time by synthesizing skipped images. We demonstrate the versatility of our method with a wide range of imaging modalities (histology, tissue-clearing/light-sheet microscopy, magnetic resonance imaging, serial section transmission electron microscopy), species (human, mouse), healthy and diseased tissues (pancreas, lung, brain), staining techniques (IHC, H&E), and pixel resolutions (8 nm, 2 μm, 1mm). Overall, we demonstrate the potential of generative AI in improving the resolution, throughput, and quality of biological image datasets, enabling improved 3D imaging.
Collapse
Affiliation(s)
- Saurabh Joshi
- Department of Chemical & Biomolecular Engineering, Johns Hopkins University, Baltimore MD
- The Johns Hopkins Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD
| | - André Forjaz
- Department of Chemical & Biomolecular Engineering, Johns Hopkins University, Baltimore MD
- The Johns Hopkins Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD
| | - Kyu Sang Han
- Department of Chemical & Biomolecular Engineering, Johns Hopkins University, Baltimore MD
- The Johns Hopkins Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD
| | - Yu Shen
- Department of Chemical & Biomolecular Engineering, Johns Hopkins University, Baltimore MD
- The Johns Hopkins Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD
- Departments of Pathology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins School of Medicine, Baltimore, MD
| | - Vasco Queiroga
- Department of Chemical & Biomolecular Engineering, Johns Hopkins University, Baltimore MD
- The Johns Hopkins Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD
| | - Daniel Xenes
- Research and Exploratory Development Department, Johns Hopkins Applied Physics Laboratory, Laurel, MD
| | - Jordan Matelsk
- Research and Exploratory Development Department, Johns Hopkins Applied Physics Laboratory, Laurel, MD
| | - Brock Wester
- Research and Exploratory Development Department, Johns Hopkins Applied Physics Laboratory, Laurel, MD
| | - Arrate Munoz Barrutia
- Bioengineering Department, Universidad Carlos III de Madrid and Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| | - Ashley L. Kiemen
- The Johns Hopkins Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD
- Departments of Pathology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins School of Medicine, Baltimore, MD
| | - Pei-Hsun Wu
- Department of Chemical & Biomolecular Engineering, Johns Hopkins University, Baltimore MD
- The Johns Hopkins Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD
| | - Denis Wirtz
- Department of Chemical & Biomolecular Engineering, Johns Hopkins University, Baltimore MD
- The Johns Hopkins Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD
- Departments of Pathology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins School of Medicine, Baltimore, MD
- Department of Oncology, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, MD
| |
Collapse
|
38
|
Vanea C, Džigurski J, Rukins V, Dodi O, Siigur S, Salumäe L, Meir K, Parks WT, Hochner-Celnikier D, Fraser A, Hochner H, Laisk T, Ernst LM, Lindgren CM, Nellåker C. Mapping cell-to-tissue graphs across human placenta histology whole slide images using deep learning with HAPPY. Nat Commun 2024; 15:2710. [PMID: 38548713 PMCID: PMC10978962 DOI: 10.1038/s41467-024-46986-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 03/15/2024] [Indexed: 04/01/2024] Open
Abstract
Accurate placenta pathology assessment is essential for managing maternal and newborn health, but the placenta's heterogeneity and temporal variability pose challenges for histology analysis. To address this issue, we developed the 'Histology Analysis Pipeline.PY' (HAPPY), a deep learning hierarchical method for quantifying the variability of cells and micro-anatomical tissue structures across placenta histology whole slide images. HAPPY differs from patch-based features or segmentation approaches by following an interpretable biological hierarchy, representing cells and cellular communities within tissues at a single-cell resolution across whole slide images. We present a set of quantitative metrics from healthy term placentas as a baseline for future assessments of placenta health and we show how these metrics deviate in placentas with clinically significant placental infarction. HAPPY's cell and tissue predictions closely replicate those from independent clinical experts and placental biology literature.
Collapse
Affiliation(s)
- Claudia Vanea
- Nuffield Department of Women's & Reproductive Health, University of Oxford, Oxford, UK.
- Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Oxford, UK.
| | | | | | - Omri Dodi
- Faculty of Medicine, Hadassah Hebrew University Medical Center, Jerusalem, Israel
| | - Siim Siigur
- Department of Pathology, Tartu University Hospital, Tartu, Estonia
| | - Liis Salumäe
- Department of Pathology, Tartu University Hospital, Tartu, Estonia
| | - Karen Meir
- Department of Pathology, Hadassah Hebrew University Medical Center, Jerusalem, Israel
| | - W Tony Parks
- Department of Laboratory Medicine & Pathobiology, University of Toronto, Toronto, Canada
| | | | - Abigail Fraser
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
- MRC Integrative Epidemiology Unit at the University of Bristol, Bristol, UK
| | - Hagit Hochner
- Braun School of Public Health, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Triin Laisk
- Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Linda M Ernst
- Department of Pathology and Laboratory Medicine, NorthShore University HealthSystem, Chicago, USA
- Department of Pathology, University of Chicago Pritzker School of Medicine, Chicago, USA
| | - Cecilia M Lindgren
- Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Oxford, UK
- Centre for Human Genetics, Nuffield Department, University of Oxford, Oxford, UK
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Nuffield Department of Population Health Health, University of Oxford, Oxford, UK
| | - Christoffer Nellåker
- Nuffield Department of Women's & Reproductive Health, University of Oxford, Oxford, UK.
- Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Oxford, UK.
| |
Collapse
|
39
|
Forjaz A, Vaz E, Romero VM, Joshi S, Braxton AM, Jiang AC, Fujikura K, Cornish T, Hong SM, Hruban RH, Wu PH, Wood LD, Kiemen AL, Wirtz D. Three-dimensional assessments are necessary to determine the true, spatially-resolved composition of tissues. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.12.04.569986. [PMID: 38106231 PMCID: PMC10723352 DOI: 10.1101/2023.12.04.569986] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
Abstract
Methods for spatially resolved cellular profiling using thinly cut sections have enabled in-depth quantitative tissue mapping to study inter-sample and intra-sample differences in normal human anatomy and disease onset and progression. These methods often profile extremely limited regions, which may impact the evaluation of heterogeneity due to tissue sub-sampling. Here, we applied CODA, a deep learning-based tissue mapping platform, to reconstruct the three-dimensional (3D) microanatomy of grossly normal and cancer-containing human pancreas biospecimens obtained from individuals who underwent pancreatic resection. To compare inter- and intra-sample heterogeneity, we assessed bulk and spatially resolved tissue composition in a cohort of two-dimensional (2D) whole slide images (WSIs) and a cohort of thick slabs of pancreas tissue that were digitally reconstructed in 3D from serial sections. To demonstrate the marked under sampling of 2D assessments, we simulated the number of WSIs and tissue microarrays (TMAs) necessary to represent the compositional heterogeneity of 3D data within 10% error to reveal that tens of WSIs and hundreds of TMA cores are sometimes needed. We show that spatial correlation of different pancreatic structures decay significantly within a span of microns, demonstrating that 2D histological sections may not be representative of their neighboring tissues. In sum, we demonstrate that 3D assessments are necessary to accurately assess tissue composition in normal and abnormal specimens and in order to accurately determine neoplastic content. These results emphasize the importance of intra-sample heterogeneity in tissue mapping efforts.
Collapse
Affiliation(s)
- André Forjaz
- Department of Chemical & Biomolecular Engineering, Johns Hopkins University, Baltimore, MD
| | - Eduarda Vaz
- Department of Chemical & Biomolecular Engineering, Johns Hopkins University, Baltimore, MD
| | - Valentina Matos Romero
- Department of Chemical & Biomolecular Engineering, Johns Hopkins University, Baltimore, MD
| | - Saurabh Joshi
- Department of Chemical & Biomolecular Engineering, Johns Hopkins University, Baltimore, MD
| | - Alicia M. Braxton
- Department of Comparative Medicine, Medical University of South Carolina, Charleston, SC
| | - Ann C. Jiang
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD
| | - Kohei Fujikura
- Department of Medical Genetics, Life Sciences Institute, University of British Columbia, Vancouver, BC, Canada
| | - Toby Cornish
- Department of Pathology, University of Colorado School of Medicine, Aurora, CO
| | - Seung-Mo Hong
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Ralph H. Hruban
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins School of Medicine, Baltimore, MD
- Department of Oncology, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, MD
| | - Pei-Hsun Wu
- Department of Chemical & Biomolecular Engineering, Johns Hopkins University, Baltimore, MD
- The Johns Hopkins Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD
| | - Laura D. Wood
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins School of Medicine, Baltimore, MD
- Department of Oncology, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, MD
| | - Ashley L. Kiemen
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins School of Medicine, Baltimore, MD
- Department of Oncology, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, MD
- The Johns Hopkins Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD
| | - Denis Wirtz
- Department of Chemical & Biomolecular Engineering, Johns Hopkins University, Baltimore, MD
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins School of Medicine, Baltimore, MD
- Department of Oncology, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, MD
- The Johns Hopkins Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD
| |
Collapse
|
40
|
Liu P, Zhang T, Huang Y. Three-dimensional model of normal human dermal tissue using serial tissue sections. Front Bioeng Biotechnol 2024; 12:1347159. [PMID: 38511132 PMCID: PMC10953291 DOI: 10.3389/fbioe.2024.1347159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 02/15/2024] [Indexed: 03/22/2024] Open
Abstract
Background: This study aims to construct a three-dimensional model of skin dermis utilizing continuous tissue sections, with the primary objective of obtaining anatomical structure data for normal human dermal tissues. Methods: Normal skin tissue specimens were acquired, paraffin-embedded, and subjected to HE staining. Panoramic images of skin sections were captured using a microscope. Tissue section images were aligned using the SIFT and StackReg image alignment methods, with analysis conducted using the OpenCV module. Mimics17 software facilitated the reconstruction of the skin dermal 3D model, enabling the calculation of dermal porosity and the void diameter. Results: Panoramic skin slices exhibited high-resolution differentiation of dermal fibers and cellular structures. Both SIFT and StackReg image alignment methods yielded similar results, although the SIFT method demonstrated greater robustness. Successful reconstruction of the three-dimensional dermal structure was achieved. Quantitative analysis revealed a dermal porosity of 18.96 ± 4.41% and an average pore diameter of 219.29 ± 34.27 μm. Interestingly, the porosity of the dermis exhibited a gradual increase from the papillary layer to the fourth layer, followed by a transient decrease and then a gradual increase. The distribution of the mean pore diameter mirrored the pattern observed in porosity distribution. Conclusion: Utilizing the continuous skin tissue slice reconstruction technique, this study successfully reconstructed a high-precision three-dimensional tissue structure of the skin. The quantitative analysis of dermal tissue porosity and average pore diameter provides a standardized dataset for the development of biomimetic tissue-engineered skin.
Collapse
Affiliation(s)
- Peng Liu
- Department of Burn and Plastic, Guangzhou Red Cross Hospital, Medical College, Jinan University, Guangzhou, China
| | - Tao Zhang
- Department of Burn and Plastic, Guangzhou Red Cross Hospital, Medical College, Jinan University, Guangzhou, China
| | - Yihui Huang
- Department of Pediatric Medicine, Guangzhou Red Cross Hospital, Medical College, Jinan University, Guangzhou, China
| |
Collapse
|
41
|
Kathiriya IS, Dominguez MH, Rao KS, Muncie-Vasic JM, Devine WP, Hu KM, Hota SK, Garay BI, Quintero D, Goyal P, Matthews MN, Thomas R, Sukonnik T, Miguel-Perez D, Winchester S, Brower EF, Forjaz A, Wu PH, Wirtz D, Kiemen AL, Bruneau BG. A disrupted compartment boundary underlies abnormal cardiac patterning and congenital heart defects. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.05.578995. [PMID: 38370632 PMCID: PMC10871243 DOI: 10.1101/2024.02.05.578995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/20/2024]
Abstract
Failure of septation of the interventricular septum (IVS) is the most common congenital heart defect (CHD), but mechanisms for patterning the IVS are largely unknown. We show that a Tbx5+/Mef2cAHF+ progenitor lineage forms a compartment boundary bisecting the IVS. This coordinated population originates at a first- and second heart field interface, subsequently forming a morphogenetic nexus. Ablation of Tbx5+/Mef2cAHF+ progenitors cause IVS disorganization, right ventricular hypoplasia and mixing of IVS lineages. Reduced dosage of the CHD transcription factor TBX5 disrupts boundary position and integrity, resulting in ventricular septation defects (VSDs) and patterning defects, including Slit2 and Ntn1 misexpression. Reducing NTN1 dosage partly rescues cardiac defects in Tbx5 mutant embryos. Loss of Slit2 or Ntn1 causes VSDs and perturbed septal lineage distributions. Thus, we identify essential cues that direct progenitors to pattern a compartment boundary for proper cardiac septation, revealing new mechanisms for cardiac birth defects.
Collapse
Affiliation(s)
- Irfan S Kathiriya
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, San Francisco, CA
| | - Martin H Dominguez
- Gladstone Institutes, San Francisco, CA
- Department of Medicine, University of California, San Francisco, San Francisco, CA
- Current address: Department of Medicine (Cardiovascular Medicine), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Kavitha S Rao
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, San Francisco, CA
- Gladstone Institutes, San Francisco, CA
| | | | - W Patrick Devine
- Gladstone Institutes, San Francisco, CA
- Current address: Department of Pathology, University of California, San Francisco, San Francisco, CA
| | - Kevin M Hu
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, San Francisco, CA
- Gladstone Institutes, San Francisco, CA
- Current address: Creighton University School of Medicine, Omaha, NE
| | - Swetansu K Hota
- Gladstone Institutes, San Francisco, CA
- Current address: Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN
| | - Bayardo I Garay
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, San Francisco, CA
- Current address: University of Minnesota Medical Scientist Training Program, Minneapolis, MN
| | - Diego Quintero
- Gladstone Institutes, San Francisco, CA
- Current address: Department of Human Genetics, Emory University School of Medicine, Atlanta, GA
| | - Piyush Goyal
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, San Francisco, CA
- Gladstone Institutes, San Francisco, CA
- Current address: Touro University California, Vallejo, CA
| | - Megan N Matthews
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, San Francisco, CA
| | | | | | | | | | | | - André Forjaz
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD
| | - Pei-Hsun Wu
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD
| | - Denis Wirtz
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD
| | - Ashley L Kiemen
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD
| | - Benoit G Bruneau
- Gladstone Institutes, San Francisco, CA
- Roddenberry Center for Stem Cell Biology and Medicine, Gladstone Institutes, San Francisco, CA
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA
| |
Collapse
|
42
|
Kiemen AL, Dbouk M, Diwan EA, Forjaz A, Dequiedt L, Baghdadi A, Madani SP, Grahn MP, Jones C, Vedula S, Wu P, Wirtz D, Kern S, Goggins M, Hruban RH, Kamel IR, Canto MI. Magnetic Resonance Imaging-Based Assessment of Pancreatic Fat Strongly Correlates With Histology-Based Assessment of Pancreas Composition. Pancreas 2024; 53:e180-e186. [PMID: 38194643 PMCID: PMC10872776 DOI: 10.1097/mpa.0000000000002288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2024]
Abstract
OBJECTIVE The aim of the study is to assess the relationship between magnetic resonance imaging (MRI)-based estimation of pancreatic fat and histology-based measurement of pancreatic composition. MATERIALS AND METHODS In this retrospective study, MRI was used to noninvasively estimate pancreatic fat content in preoperative images from high-risk individuals and disease controls having normal pancreata. A deep learning algorithm was used to label 11 tissue components at micron resolution in subsequent pancreatectomy histology. A linear model was used to determine correlation between histologic tissue composition and MRI fat estimation. RESULTS Twenty-seven patients (mean age 64.0 ± 12.0 years [standard deviation], 15 women) were evaluated. The fat content measured by MRI ranged from 0% to 36.9%. Intrapancreatic histologic tissue fat content ranged from 0.8% to 38.3%. MRI pancreatic fat estimation positively correlated with microanatomical composition of fat (r = 0.90, 0.83 to 0.95], P < 0.001); as well as with pancreatic cancer precursor ( r = 0.65, P < 0.001); and collagen ( r = 0.46, P < 0.001) content, and negatively correlated with pancreatic acinar ( r = -0.85, P < 0.001) content. CONCLUSIONS Pancreatic fat content, measurable by MRI, correlates to acinar content, stromal content (fibrosis), and presence of neoplastic precursors of cancer.
Collapse
Affiliation(s)
- Ashley L. Kiemen
- Departments of Pathology, The Johns Hopkins University School of Medicine; 600 North Wolfe Street, Baltimore, Maryland 21287, USA
- Departments of Chemical and Biomolecular Engineering, The Johns Hopkins University; 3400 N Charles St, Baltimore, Maryland 21218, USA
- Oncology, The Johns Hopkins University School of Medicine; 600 North Wolfe Street, Baltimore, Maryland 21287, USA
| | - Mohamad Dbouk
- Departments of Pathology, The Johns Hopkins University School of Medicine; 600 North Wolfe Street, Baltimore, Maryland 21287, USA
- Department of Medicine, Washington University St. Louis, St. Louis, USA; 1 Brookings Dr, St. Louis, MO 63130
| | - Elizabeth Abou Diwan
- Department of Medicine, Washington University St. Louis, St. Louis, USA; 1 Brookings Dr, St. Louis, MO 63130
| | - André Forjaz
- Departments of Chemical and Biomolecular Engineering, The Johns Hopkins University; 3400 N Charles St, Baltimore, Maryland 21218, USA
| | - Lucie Dequiedt
- Departments of Chemical and Biomolecular Engineering, The Johns Hopkins University; 3400 N Charles St, Baltimore, Maryland 21218, USA
| | - Azarakhsh Baghdadi
- Radiology and Radiological Science, The Johns Hopkins University School of Medicine; 600 North Wolfe Street, Baltimore, Maryland 21287, USA
| | - Seyedeh Panid Madani
- Radiology and Radiological Science, The Johns Hopkins University School of Medicine; 600 North Wolfe Street, Baltimore, Maryland 21287, USA
| | - Mia P. Grahn
- Departments of Chemical and Biomolecular Engineering, The Johns Hopkins University; 3400 N Charles St, Baltimore, Maryland 21218, USA
| | - Craig Jones
- Computer Science, The Johns Hopkins University; 3400 N Charles St, Baltimore, Maryland 21218, USA
- Malone Center for Engineering in Healthcare, The Johns Hopkins University; 3400 N Charles St, Baltimore, Maryland 21218, USA
| | - Swaroop Vedula
- Malone Center for Engineering in Healthcare, The Johns Hopkins University; 3400 N Charles St, Baltimore, Maryland 21218, USA
| | - PeiHsun Wu
- Departments of Chemical and Biomolecular Engineering, The Johns Hopkins University; 3400 N Charles St, Baltimore, Maryland 21218, USA
| | - Denis Wirtz
- Departments of Pathology, The Johns Hopkins University School of Medicine; 600 North Wolfe Street, Baltimore, Maryland 21287, USA
- Departments of Chemical and Biomolecular Engineering, The Johns Hopkins University; 3400 N Charles St, Baltimore, Maryland 21218, USA
- Oncology, The Johns Hopkins University School of Medicine; 600 North Wolfe Street, Baltimore, Maryland 21287, USA
- Materials Science and Engineering, The Johns Hopkins University; 3400 N Charles St, Baltimore, Maryland 21218, USA
| | - Scott Kern
- Departments of Pathology, The Johns Hopkins University School of Medicine; 600 North Wolfe Street, Baltimore, Maryland 21287, USA
- Oncology, The Johns Hopkins University School of Medicine; 600 North Wolfe Street, Baltimore, Maryland 21287, USA
- Division of Gastroenterology and Hepatology, The Johns Hopkins University School of Medicine; 600 North Wolfe Street, Baltimore, Maryland 21287, USA
| | - Michael Goggins
- Departments of Pathology, The Johns Hopkins University School of Medicine; 600 North Wolfe Street, Baltimore, Maryland 21287, USA
- Oncology, The Johns Hopkins University School of Medicine; 600 North Wolfe Street, Baltimore, Maryland 21287, USA
| | - Ralph H. Hruban
- Departments of Pathology, The Johns Hopkins University School of Medicine; 600 North Wolfe Street, Baltimore, Maryland 21287, USA
- Oncology, The Johns Hopkins University School of Medicine; 600 North Wolfe Street, Baltimore, Maryland 21287, USA
| | - Ihab R. Kamel
- Radiology and Radiological Science, The Johns Hopkins University School of Medicine; 600 North Wolfe Street, Baltimore, Maryland 21287, USA
| | - Marcia Irene Canto
- Oncology, The Johns Hopkins University School of Medicine; 600 North Wolfe Street, Baltimore, Maryland 21287, USA
- Division of Gastroenterology and Hepatology, The Johns Hopkins University School of Medicine; 600 North Wolfe Street, Baltimore, Maryland 21287, USA
| |
Collapse
|
43
|
Wang B, Zou L, Chen J, Cao Y, Cai Z, Qiu Y, Mao L, Wang Z, Chen J, Gui L, Yang X. A Weakly Supervised Segmentation Network Embedding Cross-Scale Attention Guidance and Noise-Sensitive Constraint for Detecting Tertiary Lymphoid Structures of Pancreatic Tumors. IEEE J Biomed Health Inform 2024; 28:988-999. [PMID: 38064334 DOI: 10.1109/jbhi.2023.3340686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2024]
Abstract
The presence of tertiary lymphoid structures (TLSs) on pancreatic pathological images is an important prognostic indicator of pancreatic tumors. Therefore, TLSs detection on pancreatic pathological images plays a crucial role in diagnosis and treatment for patients with pancreatic tumors. However, fully supervised detection algorithms based on deep learning usually require a large number of manual annotations, which is time-consuming and labor-intensive. In this paper, we aim to detect the TLSs in a manner of few-shot learning by proposing a weakly supervised segmentation network. We firstly obtain the lymphocyte density maps by combining a pretrained model for nuclei segmentation and a domain adversarial network for lymphocyte nuclei recognition. Then, we establish a cross-scale attention guidance mechanism by jointly learning the coarse-scale features from the original histopathology images and fine-scale features from our designed lymphocyte density attention. A noise-sensitive constraint is introduced by an embedding signed distance function loss in the training procedure to reduce tiny prediction errors. Experimental results on two collected datasets demonstrate that our proposed method significantly outperforms the state-of-the-art segmentation-based algorithms in terms of TLSs detection accuracy. Additionally, we apply our method to study the congruent relationship between the density of TLSs and peripancreatic vascular invasion and obtain some clinically statistical results.
Collapse
|
44
|
Karageorgos GM, Cho S, McDonough E, Chadwick C, Ghose S, Owens J, Jung KJ, Machiraju R, West R, Brooks JD, Mallick P, Ginty F. Deep learning-based automated pipeline for blood vessel detection and distribution analysis in multiplexed prostate cancer images. FRONTIERS IN BIOINFORMATICS 2024; 3:1296667. [PMID: 38323039 PMCID: PMC10844485 DOI: 10.3389/fbinf.2023.1296667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 12/18/2023] [Indexed: 02/08/2024] Open
Abstract
Introduction: Prostate cancer is a highly heterogeneous disease, presenting varying levels of aggressiveness and response to treatment. Angiogenesis is one of the hallmarks of cancer, providing oxygen and nutrient supply to tumors. Micro vessel density has previously been correlated with higher Gleason score and poor prognosis. Manual segmentation of blood vessels (BVs) In microscopy images is challenging, time consuming and may be prone to inter-rater variabilities. In this study, an automated pipeline is presented for BV detection and distribution analysis in multiplexed prostate cancer images. Methods: A deep learning model was trained to segment BVs by combining CD31, CD34 and collagen IV images. In addition, the trained model was used to analyze the size and distribution patterns of BVs in relation to disease progression in a cohort of prostate cancer patients (N = 215). Results: The model was capable of accurately detecting and segmenting BVs, as compared to ground truth annotations provided by two reviewers. The precision (P), recall (R) and dice similarity coefficient (DSC) were equal to 0.93 (SD 0.04), 0.97 (SD 0.02) and 0.71 (SD 0.07) with respect to reviewer 1, and 0.95 (SD 0.05), 0.94 (SD 0.07) and 0.70 (SD 0.08) with respect to reviewer 2, respectively. BV count was significantly associated with 5-year recurrence (adjusted p = 0.0042), while both count and area of blood vessel were significantly associated with Gleason grade (adjusted p = 0.032 and 0.003 respectively). Discussion: The proposed methodology is anticipated to streamline and standardize BV analysis, offering additional insights into the biology of prostate cancer, with broad applicability to other cancers.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Kyeong Joo Jung
- Department of Computer Science and Engineering, The Ohio State University, Columbus, OH, United States
| | - Raghu Machiraju
- Department of Computer Science and Engineering, The Ohio State University, Columbus, OH, United States
| | - Robert West
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, United States
| | - James D. Brooks
- Department of Urology, Stanford University School of Medicine, Stanford, CA, United States
| | - Parag Mallick
- Canary Center for Cancer Early Detection, Department of Radiology, Stanford University School of Medicine, Stanford, CA, United States
| | | |
Collapse
|
45
|
Mathur S, Chen S, Rejniak KA. Exploring chronic and transient tumor hypoxia for predicting the efficacy of hypoxia-activated pro-drugs. NPJ Syst Biol Appl 2024; 10:1. [PMID: 38182612 PMCID: PMC10770176 DOI: 10.1038/s41540-023-00327-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 12/12/2023] [Indexed: 01/07/2024] Open
Abstract
Hypoxia, a low level of oxygen in the tissue, arises due to an imbalance between the vascular oxygen supply and oxygen demand by the surrounding cells. Typically, hypoxia is viewed as a negative marker of patients' survival, because of its implication in the development of aggressive tumors and tumor resistance. Several drugs that specifically target the hypoxic cells have been developed, providing an opportunity for exploiting hypoxia to improve cancer treatment. Here, we consider combinations of hypoxia-activated pro-drugs (HAPs) and two compounds that transiently increase intratumoral hypoxia: a vasodilator and a metabolic sensitizer. To effectively design treatment protocols with multiple compounds we used mathematical micro-pharmacology modeling and determined treatment schedules that take advantage of heterogeneous and dynamically changing oxygenation in tumor tissue. Our model was based on data from murine pancreatic cancers treated with evofosfamide (as a HAP) and either hydralazine (as a vasodilator), or pyruvate (as a metabolic sensitizer). Subsequently, this model was used to identify optimal schedules for different treatment combinations. Our simulations showed that schedules of HAPs with the vasodilator had a bimodal distribution, while HAPs with the sensitizer showed an elongated plateau. All schedules were more successful than HAP monotherapy. The three-compound combination had three local optima, depending on the HAPs clearance from the tissue interstitium, each two-fold more effective than baseline HAP treatment. Our study indicates that the three-compound therapy administered in the defined order will improve cancer response and that designing complex schedules could benefit from the use of mathematical modeling.
Collapse
Affiliation(s)
- Shreya Mathur
- H. Lee Moffitt Cancer Center and Research Institute, IMO High School Internship Program, Tampa, FL, USA
- University of Florida, Undergraduate Studies, Gainesville, FL, USA
| | - Shannon Chen
- H. Lee Moffitt Cancer Center and Research Institute, IMO High School Internship Program, Tampa, FL, USA
- University of Florida, Undergraduate Studies, Gainesville, FL, USA
| | - Katarzyna A Rejniak
- H. Lee Moffitt Cancer Center and Research Institute, Integrated Mathematical Oncology Department, Tampa, FL, USA.
- University of South Florida, Morsani College of Medicine, Department of Oncologic Sciences, Tampa, FL, USA.
| |
Collapse
|
46
|
Levy JJ, Davis MJ, Chacko RS, Davis MJ, Fu LJ, Goel T, Pamal A, Nafi I, Angirekula A, Suvarna A, Vempati R, Christensen BC, Hayden MS, Vaickus LJ, LeBoeuf MR. Intraoperative margin assessment for basal cell carcinoma with deep learning and histologic tumor mapping to surgical site. NPJ Precis Oncol 2024; 8:2. [PMID: 38172524 PMCID: PMC10764333 DOI: 10.1038/s41698-023-00477-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 11/14/2023] [Indexed: 01/05/2024] Open
Abstract
Successful treatment of solid cancers relies on complete surgical excision of the tumor either for definitive treatment or before adjuvant therapy. Intraoperative and postoperative radial sectioning, the most common form of margin assessment, can lead to incomplete excision and increase the risk of recurrence and repeat procedures. Mohs Micrographic Surgery is associated with complete removal of basal cell and squamous cell carcinoma through real-time margin assessment of 100% of the peripheral and deep margins. Real-time assessment in many tumor types is constrained by tissue size, complexity, and specimen processing / assessment time during general anesthesia. We developed an artificial intelligence platform to reduce the tissue preprocessing and histological assessment time through automated grossing recommendations, mapping and orientation of tumor to the surgical specimen. Using basal cell carcinoma as a model system, results demonstrate that this approach can address surgical laboratory efficiency bottlenecks for rapid and complete intraoperative margin assessment.
Collapse
Affiliation(s)
- Joshua J Levy
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA.
- Department of Computational Biomedicine, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA.
- Department of Dermatology, Geisel School of Medicine at Dartmouth, Hanover, NH, 03756, USA.
- Emerging Diagnostic and Investigative Technologies, Clinical Genomics and Advanced Technologies, Department of Pathology and Laboratory Medicine, Dartmouth Hitchcock Medical Center, Lebanon, NH, 03756, USA.
- Department of Epidemiology, Geisel School of Medicine at Dartmouth, Hanover, NH, 03756, USA.
- Program in Quantitative Biomedical Sciences, Geisel School of Medicine at Dartmouth, Hanover, NH, 03756, USA.
| | - Matthew J Davis
- Department of Dermatology, Geisel School of Medicine at Dartmouth, Hanover, NH, 03756, USA
| | | | - Michael J Davis
- Department of Dermatology, Geisel School of Medicine at Dartmouth, Hanover, NH, 03756, USA
| | - Lucy J Fu
- Geisel School of Medicine at Dartmouth, Hanover, NH, 03755, USA
| | - Tarushii Goel
- Thomas Jefferson High School for Science and Technology, Alexandria, VA, 22312, USA
- Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Akash Pamal
- Thomas Jefferson High School for Science and Technology, Alexandria, VA, 22312, USA
- University of Virginia, Charlottesville, VA, 22903, USA
| | - Irfan Nafi
- Thomas Jefferson High School for Science and Technology, Alexandria, VA, 22312, USA
- Stanford University, Palo Alto, CA, 94305, USA
| | - Abhinav Angirekula
- Thomas Jefferson High School for Science and Technology, Alexandria, VA, 22312, USA
- University of Illinois Urbana-Champaign, Champaign, IL, 61820, USA
| | - Anish Suvarna
- Thomas Jefferson High School for Science and Technology, Alexandria, VA, 22312, USA
| | - Ram Vempati
- Thomas Jefferson High School for Science and Technology, Alexandria, VA, 22312, USA
| | - Brock C Christensen
- Department of Dermatology, Geisel School of Medicine at Dartmouth, Hanover, NH, 03756, USA
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, NH, 03756, USA
- Department of Community and Family Medicine, Geisel School of Medicine at Dartmouth, Hanover, NH, 03756, USA
| | - Matthew S Hayden
- Department of Dermatology, Geisel School of Medicine at Dartmouth, Hanover, NH, 03756, USA
| | - Louis J Vaickus
- Emerging Diagnostic and Investigative Technologies, Clinical Genomics and Advanced Technologies, Department of Pathology and Laboratory Medicine, Dartmouth Hitchcock Medical Center, Lebanon, NH, 03756, USA
| | - Matthew R LeBoeuf
- Department of Dermatology, Geisel School of Medicine at Dartmouth, Hanover, NH, 03756, USA
| |
Collapse
|
47
|
Liu JTC, Chow SSL, Colling R, Downes MR, Farré X, Humphrey P, Janowczyk A, Mirtti T, Verrill C, Zlobec I, True LD. Engineering the future of 3D pathology. J Pathol Clin Res 2024; 10:e347. [PMID: 37919231 PMCID: PMC10807588 DOI: 10.1002/cjp2.347] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 10/06/2023] [Accepted: 10/15/2023] [Indexed: 11/04/2023]
Abstract
In recent years, technological advances in tissue preparation, high-throughput volumetric microscopy, and computational infrastructure have enabled rapid developments in nondestructive 3D pathology, in which high-resolution histologic datasets are obtained from thick tissue specimens, such as whole biopsies, without the need for physical sectioning onto glass slides. While 3D pathology generates massive datasets that are attractive for automated computational analysis, there is also a desire to use 3D pathology to improve the visual assessment of tissue histology. In this perspective, we discuss and provide examples of potential advantages of 3D pathology for the visual assessment of clinical specimens and the challenges of dealing with large 3D datasets (of individual or multiple specimens) that pathologists have not been trained to interpret. We discuss the need for artificial intelligence triaging algorithms and explainable analysis methods to assist pathologists or other domain experts in the interpretation of these novel, often complex, large datasets.
Collapse
Affiliation(s)
- Jonathan TC Liu
- Department of Mechanical EngineeringUniversity of WashingtonSeattleWAUSA
- Department of Laboratory Medicine & PathologyUniversity of Washington School of MedicineSeattleUSA
- Department of BioengineeringUniversity of WashingtonSeattleUSA
| | - Sarah SL Chow
- Department of Mechanical EngineeringUniversity of WashingtonSeattleWAUSA
| | | | | | | | - Peter Humphrey
- Department of UrologyYale School of MedicineNew HavenCTUSA
| | - Andrew Janowczyk
- Wallace H Coulter Department of Biomedical EngineeringEmory University and Georgia Institute of TechnologyAtlantaGAUSA
- Geneva University HospitalsGenevaSwitzerland
| | - Tuomas Mirtti
- Helsinki University Hospital and University of HelsinkiHelsinkiFinland
- Emory University School of MedicineAtlantaGAUSA
| | - Clare Verrill
- John Radcliffe HospitalUniversity of OxfordOxfordUK
- NIHR Oxford Biomedical Research CentreOxford University Hospitals NHS Foundation TrustOxfordUK
| | - Inti Zlobec
- Institute for Tissue Medicine and PathologyUniversity of BernBernSwitzerland
| | - Lawrence D True
- Department of Laboratory Medicine & PathologyUniversity of Washington School of MedicineSeattleUSA
| |
Collapse
|
48
|
Clifton K, Anant M, Aihara G, Atta L, Aimiuwu OK, Kebschull JM, Miller MI, Tward D, Fan J. STalign: Alignment of spatial transcriptomics data using diffeomorphic metric mapping. Nat Commun 2023; 14:8123. [PMID: 38065970 PMCID: PMC10709594 DOI: 10.1038/s41467-023-43915-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 11/23/2023] [Indexed: 12/18/2023] Open
Abstract
Spatial transcriptomics (ST) technologies enable high throughput gene expression characterization within thin tissue sections. However, comparing spatial observations across sections, samples, and technologies remains challenging. To address this challenge, we develop STalign to align ST datasets in a manner that accounts for partially matched tissue sections and other local non-linear distortions using diffeomorphic metric mapping. We apply STalign to align ST datasets within and across technologies as well as to align ST datasets to a 3D common coordinate framework. We show that STalign achieves high gene expression and cell-type correspondence across matched spatial locations that is significantly improved over landmark-based affine alignments. Applying STalign to align ST datasets of the mouse brain to the 3D common coordinate framework from the Allen Brain Atlas, we highlight how STalign can be used to lift over brain region annotations and enable the interrogation of compositional heterogeneity across anatomical structures. STalign is available as an open-source Python toolkit at https://github.com/JEFworks-Lab/STalign and as Supplementary Software with additional documentation and tutorials available at https://jef.works/STalign .
Collapse
Affiliation(s)
- Kalen Clifton
- Center for Computational Biology, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD, USA
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Manjari Anant
- Center for Computational Biology, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD, USA
- Department of Neuroscience, Johns Hopkins University, Baltimore, MD, USA
| | - Gohta Aihara
- Center for Computational Biology, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD, USA
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Lyla Atta
- Center for Computational Biology, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD, USA
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Osagie K Aimiuwu
- University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Justus M Kebschull
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
- Kavli Neuroscience Discovery Institute, The Johns Hopkins University, Baltimore, MD, USA
| | - Michael I Miller
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
- Kavli Neuroscience Discovery Institute, The Johns Hopkins University, Baltimore, MD, USA
| | - Daniel Tward
- Department of Computational Medicine, University of California Los Angeles, Los Angeles, CA, USA.
- Department of Neurology, University of California Los Angeles, Los Angeles, CA, USA.
| | - Jean Fan
- Center for Computational Biology, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD, USA.
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA.
- Kavli Neuroscience Discovery Institute, The Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
49
|
Kiemen AL, Wu PH, Braxton AM, Cornish TC, Hruban RH, Wood L, Wirtz D, Zwicker D. Power-law growth models explain incidences and sizes of pancreatic cancer precursor lesions and confirm spatial genomic findings. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.01.569633. [PMID: 38105957 PMCID: PMC10723372 DOI: 10.1101/2023.12.01.569633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
Abstract
Pancreatic ductal adenocarcinoma is a rare but lethal cancer. Recent evidence reveals that pancreatic intraepithelial neoplasms (PanINs), the microscopic precursor lesions in the pancreatic ducts that can give rise to invasive pancreatic cancer, are significantly larger and more prevalent than previously believed. Better understanding of the growth law dynamics of PanINs may improve our ability to understand how a miniscule fraction of these lesions makes the transition to invasive cancer. Here, using artificial intelligence (AI)-based three-dimensional (3D) tissue mapping method, we measured the volumes of >1,000 PanIN and found that lesion size is distributed according to a power law with a fitted exponent of -1.7 over > 3 orders of magnitude. Our data also suggest that PanIN growth is not very sensitive to the pancreatic microenvironment or an individual's age, family history, and lifestyle, and is rather shaped by general growth behavior. We analyze several models of PanIN growth and fit the predicted size distributions to the observed data. The best fitting models suggest that both intraductal spread of PanIN lesions and fusing of multiple lesions into large, highly branched structures drive PanIN growth patterns. This work lays the groundwork for future mathematical modeling efforts integrating PanIN incidence, morphology, genomic, and transcriptomic features to understand pancreas tumorigenesis, and demonstrates the utility of combining experimental measurement of human tissues with dynamic modeling for understanding cancer tumorigenesis.
Collapse
Affiliation(s)
- Ashley L. Kiemen
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Oncology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Pei-Hsun Wu
- Department of Chemical & Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Alicia M. Braxton
- Department of Comparative Medicine, Medical University of South Carolina, Charleston, SC
| | - Toby C. Cornish
- Department of Pathology, University of Colorado School of Medicine, Aurora, CO
| | - Ralph H. Hruban
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Oncology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Laura Wood
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Oncology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Denis Wirtz
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Oncology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Chemical & Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - David Zwicker
- Max Planck Institute for Dynamics and Self-Organization, Göttingen, Germany
| |
Collapse
|
50
|
Walsh LA, Quail DF. Decoding the tumor microenvironment with spatial technologies. Nat Immunol 2023; 24:1982-1993. [PMID: 38012408 DOI: 10.1038/s41590-023-01678-9] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Accepted: 10/10/2023] [Indexed: 11/29/2023]
Abstract
Visualization of the cellular heterogeneity and spatial architecture of the tumor microenvironment (TME) is becoming increasingly important to understand mechanisms of disease progression and therapeutic response. This is particularly relevant in the era of cancer immunotherapy, in which the contexture of immune cell positioning within the tumor landscape has been proven to affect efficacy. Although single-cell technologies have mostly replaced conventional approaches to analyze specific cellular subsets within tumors, those that integrate a spatial dimension are now on the rise. In this Review, we assess the strengths and limitations of emerging spatial technologies with a focus on their applications in tumor immunology, as well as forthcoming opportunities for artificial intelligence (AI) and the value of integrating multiomics datasets to achieve a holistic picture of the TME.
Collapse
Affiliation(s)
- Logan A Walsh
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, Quebec, Canada.
- Department of Human Genetics, Faculty of Medicine, McGill University, Montreal, Quebec, Canada.
| | - Daniela F Quail
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, Quebec, Canada.
- Department of Physiology, Faculty of Medicine, McGill University, Montreal, Quebec, Canada.
- Department of Medicine, Division of Experimental Medicine, McGill University, Montreal, Quebec, Canada.
| |
Collapse
|