Virtual Event

The Spatial Biology Week™ 2023

Discover new avenues and angles for accelerating scientific research

2-6 October 2023

FREE REGISTRATION

ONLINE

Event agenda

What will the future of spatial biology look like? From the way hypothesis are generated and multiomic data are collected to advance drug discovery and develop targeted treatment, the new spatial paradigm has opened up new avenues and angles for accelerating scientific research.

The Spatial Biology Week™ represents a unique opportunity for the scientific community to connect with and learn from world-class experts. This 5-day virtual meeting explored a broad spectrum of topics from various research areas, showcasing spatial biology’s far-reaching and transformative impact.

Watch the talks on-demand. Registration is free, check the full event agenda below.

The Spatial Biology Week™ speakers list includes:

  • Moran Amit, MD, Ph.D. – Assistant Professor, MD Anderson Cancer Center
  • Idris Boudhabhay, MD – Ph.D. Candidate, Centre de Recherche des Cordeliers, Unité INSERM UMRS1138, Department of Nephrology and Kidney Transplantation, Necker Hospital
  • Michaeline Bunting, Ph.D. – Sr. Director Product Management and Marketing, Bio-Techne
  • Jared Burks, Ph.D. – Core Facility Director, MD Anderson Cancer Center
  • Michael DeCuypere, MD, Ph.D. – Director of Translational Research and Clinical Trials in Neuro-Oncology & Assistant Professor of Neurological Surgery, Northwestern University
  • Pedro Espinosa Gonzalez, M.Sc. – Field Applications Scientist, Indica Labs
  • Sami Farhi, Ph.D. – Group Leader, Klarman Cell Observatory at the Broad Institute of MIT and Harvard, Optical Profiling Platform
  • Sammy Ferri-Borgogno, Ph.D. – Instructor, MD Anderson Cancer Center
  • Bora Gurel, MD, Ph.D. – Clinician Scientist and Histopathologist, Institute of Cancer Research
  • Mauro Gwerder, M.Sc. University of Bern
  • Amy B. Heimberger, MD, Ph.D. – Vice Chair for Research and Professor, Northwestern University
  • Kelly Hunter, M.Sc. – Chief Scientific Officer, ProPathUK
  • Joanna Kowal, Ph.D. – Senior Scientific Affairs Manager, Lunaphore Technologies
  • Catalina Lee Chang, Ph.D. – Assistant Professor at Northwestern University, The Feinberg School of Medicine
  • James R. Mansfield – SVP Research Business Development, Visiopharm
  • Jimin Min, Ph.D. – Postdoctoral Fellow, Maitra’s lab, MD Anderson Cancer Center
  • Hinda Najem, MD, Ph.D. – Postdoctoral Fellow, Heimberger’s lab, Northwestern University
  • Matthew Pugh, MD, Ph.D. – Jean Shanks Pathsoc Intermediate Fellow and Associate Clinical Professor, University of Birmingham
  • David R. Raleigh, MD, Ph.D. – Associate Professor, University of California San Francisco
  • Albert Santamaria Martinez, Ph.D. – Scientist, Oricchio’s lab, Ecole Polytechnique Fédérale de Lausanne
  • Denis Schapiro, Ph.D. – Group Leader, University Hospital Heidelberg
  • Farzad Sekhavati, Ph.D. – Project Manager Tissue Analytics, Lunaphore Technologies
  • Sheila Singh, MD, Ph.D., FRCS(C) – Professor of Surgery and Biochemistry, Chief Pediatric Neurosurgeon at McMaster Children’s Hospital, Hamilton Health Sciences, Director of McMaster Centre for Discovery in Cancer Research
  • Adam M. Sonabend Worthalter, M.D. – Associate Professor of Neurological Surgery, Northwestern University
  • Antonio Sorrentino, Ph.D. – Head of Translational Strategy, Lunaphore Technologies
  • Florian Wünnemann, Ph.D. – Postdoctoral Fellow, Denis Schapiro’s lab, Heidelberg University, Heidelberg University Hospital, Bioquant

Access the full agenda and speakers bio below.

DISCLAIMER: Participation in the event is the sole decision of the speakers. The presentations are based on their personal experience and do not imply any official endorsement by their institutions.

What is the registration cost? Is it necessary?
Registration to access the recordings of the event is required and it is free. Upon registration, you will have access to the webcast.

Have all talks been recorded?
Some talks were streamed only during the The Spatial Biology Week™ and are not available on-demand.

I need support to complete my registration/access the talks
To receive support, please email [email protected]

I have a question regarding the event
Please send your request to [email protected]

October 2

Dissecting tissue and diseases: multimodal approaches for spatial analysis

Jared K. Burks
Marco Cassano
Sammy Ferri-Borgogno
Sheila Singh
Matthew Pugh
Michaeline Bunting

Integrating the spatial omics approach with other multimodal techniques can enable a more comprehensive view of cell to cell interaction, tissue architecture, and pathological conditions. This session covers innovative research projects and their related methods that integrate multiple technologies from single cell to spatial multiomics and multimodal imaging. These approaches will enhance understanding of biological mechanisms underlying disease development, progression, and therapeutic resistance.

Watch now →

Speakers

Marco Cassano, Ph.D.

Marco Cassano, Ph.D.

Head of Scientific Affairs

Lunaphore

Jared K Burks, Ph.D.

Jared K Burks, Ph.D.

Professor, Co-Director of the North Campus Flow Cytometry & Cellular Imaging Core Facility

The University of Texas MD Anderson Cancer Center

I started my career at Texas A&M University learning about patterns in genes and proteins, allowing and facilitating subcellular protein trafficking.  As I have progressed to MD Anderson Cancer Center, I have scaled to cellular trafficking attempting to understand the spatial distribution of cells in organ systems during disease.  As in many parts of life, form equals functions.  How our cells organize speaks to how they function and respond to their local environment.  Bringing together multi-omics approaches allows for greater clarity in these imaging snapshots that are collected.

Why does spatial organization of cells matter? Can these data help us to better understand disease progression? We need to explore the niche or neighborhood in the multicellular system we want to study. Since proximity speaks to cellular activity, spatial organization of cells will certainly play a role in modulating clinical outcomes.  However, this type of complex study cannot be performed due to the shortage of tools to analyze single cell with spatial context in the multicellular system such as cancer.  A tumor cell clearly recognizes the importance of its neighborhood and restructures the neighborhood to meet its needs for its own benefit. What if we compare this interaction to a model system developed by our own behaviours? How do we decide where we want to live in a city?

With higher-plexed imaging, we can now characterize spatially resolved mRNA (spatial transcriptomics). and protein (spatial proteomics) simultaneously.  An analogy to this is that we can tell whether an individual has enough money to buy food to live in a particular neighborhood of a city. These points bring us back to another aspect, tissue heterogeneity. To avoid redundancy, we need to analyze a single tissue section using as many omics platforms as we can so that one unique and reliable city map can be generated

Sorry, this talk is not accessible on-demand.

Speaker

Sammy Ferri-Borgogno, Ph.D.

Sammy Ferri-Borgogno, Ph.D.

Instructor

The University of Texas MD Anderson Cancer Center

Dr. Sammy Ferri-Borgogno is currently an Instructor in the Department of Gynecologic Oncology and Reproductive Medicine, where she is devoted to elucidating immune landscapes and crosstalk signaling networks that interplay in the ovarian tumor microenvironment to modulate clinical outcomes. During her doctoral training at the University of Turin, Dr. Ferri-Borgogno studied how cancer biology, -omics technologies and immunology might be integrated to understand host-tumor responses and identify new diagnostic markers and therapeutic targets for solid tumors. Dr. Ferri-Borgogno started her postdoctoral training in the laboratory of Dr. Anirban Maitra in the Department of Translational Molecular Pathology at MD Anderson, with the goal to develop and expand her passionate interest in cancer biology as well as explore a more translational research approach. Her willing to explore new fields and to expand her research knowledge in cancer biology, made Dr. Ferri-Borgogno to join the laboratory of Dr. Samuel Mok. As a senior post-doctoral fellow in the lab she had primary responsibility for all research initiatives and projects related to clarify the molecular/cellular interactions existing between stromal cells and cancer immunity to identify markers in the ovarian tumor microenvironment that can simultaneously normalize aberrant angiogenesis and increase tumor immune cell activation and infiltration, thus resulting in significant treatment outcomes. As an Instructor, Dr. Ferri-Borgogno is currently applying cutting-edge technologies such as Spatial Transcriptomics (ST) and Imaging Mass Cytometry (IMC) to characterize the ovarian cancer tumor microenvironment with the final goal to identify spatial biomarkers associated with chemoresistance.

Glioblastoma (GBM) is the most common primary adult brain tumor, with standard-of-care therapy inevitably leading to therapy resistance in 96% of patients, who relapse with incurable disease. This talk will review the biological profile of treatment-refractory glioblastoma, and discuss the application of an integrative multi-omics target discovery platform for human GBM which incorporates DNA cellular barcoding, glycocapture profiling of the cell surface proteome, RNA sequencing, single-cell sequencing and genome-wide CRISPR screening of primary and recurrent GBM. We will introduce a scientific program designed to address our gap in knowledge of the spatial heterogeneity of GBM, which is built together with patients and families who chose to participate in an innovative rapid autopsy GBM donor program. 

Watch now →

Speaker

Sheila Kumari Singh, M.D., Ph.D., FRCSC

Sheila Kumari Singh, M.D., Ph.D., FRCSC

Pediatric Neurosurgeon, McMaster Children's Hospital
Professor of Surgery,  Professor of Biochemistry and Biomedical Sciences, Division Head, Neurosurgery, Hamilton Health Sciences
Director, Centre for Discovery in Cancer Research, McMaster University
Senior Canada Research Chair in Human Cancer Stem Cell Biology, University Scholar, and Chair, McMaster College of Health Inventors

McMaster Children's Hospital | Hamilton Health Services | McMaster University | McMaster College of Health Inventors

Dr. Sheila Singh is a professor of surgery and biochemistry, chief pediatric neurosurgeon at McMaster Children’s Hospital, former Division Head of Neurosurgery at Hamilton Health Sciences, and inaugural director of the new Cancer Research Centre at McMaster University. She holds a Tier 1/ Senior Canada Research Chair in Human Brain Cancer Stem Cell Biology, and is the founding Director of the McMaster Surgeon Scientist Program. Her PhD thesis described the novel identification of a population of cancer stem cells that exclusively drive the formation of brain tumours. Since 2007, Dr. Singh's lab applies a developmental neurobiology framework to the study of brain tumorigenesis. Building upon previous cell culture techniques developed for the isolation of normal neural stem cells (NSC) and applying them to brain tumours, and through development of a xenograft model to efficiently study brain tumour initiating cell (BTIC) activity, Dr. Singh's lab aims to understand the molecular mechanisms that govern BTIC self- renewal. Dr. Singh is currently studying the regulation of BTIC signaling pathways in glioblastoma, brain metastases and childhood medulloblastoma, with an ultimate goal of selectively targeting the BTIC with appropriately tailored drug and molecular therapies. Her laboratory is funded by CCSRI, CIHR, TFRI, CRS, the Stem Cell Network, McMaster Surgical Associates, Brain Canada and the Boris Family Fund. She is scientific founder and prior CEO of a startup company, Empirica Therapeutics, a brain cancer therapeutics company that is seeking new, data-driven and polytherapeutic treatment options for patients with Glioblastoma and brain metastases. Empirica was acquired by Century Therapeutics Inc (Philadelphia) in June 2020, resulting in the creation of a Canadian subsidiary, Century Canada, based in the McMaster Innovation Park in Hamilton.

Whilst the peripheral immune response to COVID-19 infection is well characterised, less is known about the tissue response in the lungs of affected patients. We conducted a multi-omic investigation in post-mortem lung tissue from fatal COVID-19 infection comprising bulk Quantseq RNA sequencing, spatial transcriptomic profiling on the Nanostring GeoMx, multiplex immunohistochemistry on the Lunaphore COMET and in-situ transcriptomcics with RNAscope. We found the SARS-CoV-2 virus was progressively cleared in more temporally advanced disease. Furthermore, collagen VI was found to be transcriptionally up-regulated by bulk sequencing and within the interstitium on spatial transcriptomics. These findings were supported by multiplex immunohistochemistry at a proteomic level which showed fibrillar deposition of collagen VI in the interstitum. To validate these findings clinically, we looked at blood markers for collagen VI synthesis and degradation by ELISA in a cohort of severe and mildly infected COVID-19 patients. Blood markers of collagen VI synthesis (PRO-C6) was significantly predictive for mortality in hospitalised (p = 0.0065) and intensive care (p = 0.028) COVID-19 patients. Overall, blood PRO-C6 levels, based on increase interstitial lung deposition, could help identify high-risk of patients enabling better treatment stratification. The study also demonstrates the power of post-mortem investigation and spatial profiling in elucidating clinically relevant insights into complex disease phenomena.

Watch now →

Speaker

Matthew Pugh, MD, PhD, FRCPath

Matthew Pugh, MD, PhD, FRCPath

Jean Shanks Pathsoc Intermediate Fellow and Associate Clinical Professor

University of Birmingham

RNAscope in situ hybridization (ISH) technology is powerful method for detecting RNA expression with single molecule sensitivity in spatial and morphological context. Backed by over 8,000 peer reviewed publications, RNAscope technology enables visualization of the widest range of diverse RNAs commercially available. 

In this presentation, Michaeline Bunting, PhD. from the Spatial Biology Division at BioTechne will discuss RNAscope™ multiomic spatial applications and how they can be used to uncover new insights into health and disease and advance therapeutic development. Learn more about how RNAscope works and future directions to automate RNAscope on the Lunaphore COMET™ system to develop the first fully automated same-slide spatial multiomics solution.  

Watch now

Speaker

Michaeline Bunting, Ph.D.

Michaeline Bunting, Ph.D.

Sr. Director, Product Management Spatial Biology Division

Advanced Cell Diagnostics, a BioTechne brand

Michaeline is the Sr. Director of Product Marketing in the Spatial Biology Division at BioTechne. She has over 20 years experience in the life science industry leading product management, marketing, and research & development in multiplex biomarker analysis. Michaeline received her PhD in Biochemistry from the University of Utah, performing research in the areas of inflammatory disease and cancer.

October 3

Navigating the brain architecture with spatial cell profiling

Amy B. Heimberger
Hinda J. Najem
Moran Amit
Adam M. Sonabend
David R. Raleigh

The brain is a complex organ that exhibits cellular and molecular heterogeneity within and between brain regions. This session features cutting edge research projects where spatial biology has been used to interrogate the brain and gather previously unknown insights. The discussions emphasize the field’s transformative impact on advancing knowledge of brain function and its diseases.

Watch now →

Speaker

Amy B Heimberger, M.D., FAANS, Ph.D.

Amy B Heimberger, M.D., FAANS, Ph.D.

Jean Malnati Miller Professor of Neurological Surgery, Vice-Chair for Research in the Department of Neurological Surgery at Northwestern University
Scientific Director of The Malnati Brain Tumor Institute, and the Interim Associate Director of Translational Research of the Robert H. Lurie Comprehensive Cancer Center

Northwestern University | Robert H. Lurie Comprehensive Cancer Center

Amy B Heimberger’s, MD, FAANS, PhD (h.c.), research program focuses on immune therapeutic strategies for patients with Central Nervous System (CNS) malignancies and she studies tumor-mediated mechanisms of immune suppression. She has been intricately involved in a wide variety of bench-to-bedside immune therapeutics, including those that were developed in her laboratory and arising from her own patents. She has expertise in the IND process and has been the PI of multiple clinical trials including the advocation for window-of-opportunity designs to evaluate the CNS tumor microenvironment. Dr. Heimberger has been awarded the United States Presidential Early Career Award for Scientists and Engineers and she holds multiple National Institute of Health and foundation grants. She has served on a wide variety of NIH study sections and was the Chair of the Clinical Neuroimmunology and Brain Tumor Study Section. Dr. Heimberger’s research interests are complemented by her surgical specialization in awake craniotomies and mapping. She has been named by the US News and World Report as a Top Doc and is a member of the American Society of Clinical Investigators. Dr. Heimberger was appointed by President Biden to serve on the National Cancer Advisory Board in 2021.

To increase the throughput of spatial profiling of the human central nervous system (CNS), a CNS-specific library of 100+ markers with an associated analysis platform was developed and validated using the Lunaphore COMET. Optimizing antibody concentrations, cycle order, and standard operating procedures, while managing subsequent image analysis with background and autofluorescence subtraction, have allowed for the visualization of molecular signaling at the subcellular level. Our approach is project-driven, from onboarding projects to interrogation sessions with investigators, with the associated panels customized to each specific scientific question. This strategy can be used to validate data such as single-cell sequencing or for novel discovery and hypothesis generation.

Watch now →

Speaker

Hinda Najem, M.D., Ph.D.

Hinda Najem, M.D., Ph.D.

Postdoctoral Research Fellow

Northwestern University

Dr. Najem is currently a postdoctoral fellow at Dr. Amy Heimberger’s Lab at Northwestern University. She obtained her medical doctor degree in July 2020 from St. Joseph University, Beirut, Lebanon. While working on her clinical medical practice, she worked for almost 3 years in pre-clinical research, when she obtained her master’s degree in Biological and Medical Sciences in December 2018 at the Physiology and Physiopathology Laboratory, St. Joseph University, Beirut, Lebanon. She joined Dr. Heimberger’s lab in November 2020 at MD Anderson Cancer Center and transferred to Northwestern University in May 2021. She is working on brain tumors’ immune profiling, immune modulation, molecular pathways responsible for immunosuppression in the tumor microenvironment, and has published 15 manuscripts in that time. She is also working on pre-clinical animal models of gliomas and on humanized mouse models, studying the effect of STING agonists and other immune therapeutic strategies.

Over the past decade, emerging technologies have enabled investigators to begin to bridge the gap between cancer and neuroscience research to unveil the interactive roles of nerves in cancer. We are now beginning to define how the nervous system contributes to cancer initiation, growth, spread, recurrence, and even resistance to oncologic therapeutic strategies. Indeed, neuromodulation with both genetic and pharmacological approaches has been shown to affect not only tumor growth but also antitumor immune response. Collectively, studies have demonstrated a significant and pressing need to recognize cancer neuroscience interplay as a hallmark of cancer to 1) establish the neoneurogenic process as a highly relevant therapeutic target for both the prevention and treatment of cancer, 2) foster interdisciplinary cross-talk among experts in cancer biology and neuroscience, which have traditionally progressed along parallel paths, and 3) integrate human variables such as biological sex, age, race, and gender as underlying factors that can impact cancer neuroscience.  

This session introduces the current viewpoint of burgeoning cancer neuroscience field and discusses the most seminal papers over the last 10 years as well as promising up-and-coming directions of the field towards its establishment as a hallmark of cancer and implement it as a therapeutic approach in clinical oncology.

Watch now →

Speaker

Moran Amit, M.D., Ph.D.

Moran Amit, M.D., Ph.D.

Assistant Professor, Head and Neck Surgery

The University of Texas MD Anderson Cancer Center

Dr. Amit is dedicated to key areas of cancer research that will develop therapeutic approaches to improve patients’ survival, treatment responses, and quality of life. During his residency training, Dr. Amit became interested in the biology of cancer. After completing his residency, he undertook further training in Head and Neck Surgical Oncology at The University of Texas MD Anderson Cancer Center, where he completed a clinical fellowship as a head and neck surgical oncologist and a postdoctoral fellowship with a focus on the neurobiology of cancer.
Dr. Amit was recruited to the Department of Head and Neck Surgical Oncology at MD Anderson in April 2019 and has an active research laboratory focusing on cancer-related neurogenesis and axonogenesis and on the role of neuro-immunology in cancer. One exciting finding of Dr. Amit’s lab involved describing a mechanism by which head and neck tumors can reprogram adrenergic signaling in neurons and the effect of targeting the peripheral nervous system on cancer initiation and progression. This will serve as a basis for combining treatments targeting the nervous system with other therapies, such as immunotherapy, in the treatment of head and neck cancers.
Dr. Amit is continuing his work by building a team to study how the neural microenvironment affects responses to chemotherapy and immunotherapy in patients with non-melanoma skin cancer and other cancers, and to develop novel strategies to combat resistance to these therapies.

Talk abstract N/A.

Watch now  →

Speaker

Adam M. Sonabend, M.D.

Adam M. Sonabend, M.D.

Associate Professor of Neurological Surgery

Northwestern University

Intratumor heterogeneity underlies cancer evolution and treatment resistance, but targetable mechanisms driving intratumor heterogeneity are poorly understood. Meningiomas are the most common primary intracranial tumors and are resistant to all current medical therapies. High-grade meningiomas cause significant neurological morbidity and mortality and are distinguished from low-grade meningiomas by increased intratumor heterogeneity arising from clonal evolution and divergence. Here we integrate spatial transcriptomic and spatial protein profiling approaches across high-grade meningiomas to identify genomic, biochemical, and cellular mechanisms linking intratumor heterogeneity to molecular, temporal, and spatial evolution. We show divergent intratumor gene and protein expression programs distinguish high-grade meningiomas that are otherwise grouped together by current clinical classification systems. Analyses of matched pairs of primary and recurrent meningiomas reveal spatial expansion of subclonal copy number variants underlies treatment resistance. Multiplexed sequential immunofluorescence (seqIF) and spatial deconvolution of meningioma single-cell RNA sequencing show decreased immune infiltration, decreased MAPK signaling, increased PI3K-AKT signaling, and increased cell proliferation drive meningioma recurrence. To translate these findings to clinical practice, we use epigenetic editing and lineage tracing approaches in meningioma organoid models to identify new molecular therapy combinations that target intratumor heterogeneity and block tumor growth. Our results establish a foundation for personalized medical therapy to treat patients with high-grade meningiomas and provide a framework for understanding therapeutic vulnerabilities driving intratumor heterogeneity and tumor evolution. 

Sorry, this talk is not accessible on-demand.

Speaker

David R. Raleigh, M.D., Ph.D.

David R. Raleigh, M.D., Ph.D.

Associate Professor
Robert and Ruth Halperin Endowed Chair in Meningioma Research
Brain Tumor Center Principal Investigator and Preclinical Therapeutics Core Director
Departments of Radiation Oncology, Neurological Surgery, and Pathology

University of California San Francisco

David R. Raleigh, MD, PhD, is an Associate Professor and Physician Scientist in the Departments of Radiation Oncology, Neurological Surgery, and Pathology at the University of California San Francisco (UCSF). His lab is focused on understanding the genomic, biochemical, and cellular drivers of intratumor heterogeneity and tumor evolution, and his research integrates human samples with diverse evolutionary and preclinical models. Dr. Raleigh is the Robert and Ruth Halperin Endowed Chair in Meningioma Research, and the Director of the Preclinical Therapeutics Core in the UCSF Brain Tumor Center. In addition to illuminating how tumors develop and respond to treatment, his research has shed light on fundamental signaling mechanisms through the Hedgehog pathway and neurofibromatosis tumor suppressors that are relevant to cancer predisposition. As a practicing physician, Dr. Raleigh treats adult and pediatric patients with nervous system tumors using radiotherapy and radiosurgery.

October 4

Spatial biomarkers for translational research

Antonio Sorrentino
Idris Boudhabhay
Kelly Hunter
Sami Farhi
Jimin Min
Catalina Lee-Chang

Spatial biology techniques have become highly valuable in biomarker development, where bulk analyses have reached a bottleneck. Researchers are pursuing novel biomarker signatures to better understand diseases and stratify patients. This session showcases real world examples of spatial biology studies to identify clinically relevant biomarkers. These case studies demonstrate how the identification and characterization of spatial biomarkers are fundamental for personalized medical and diagnostic approaches.

Sorry, this talk is not accessible on-demand.

Speaker

Antonio Sorrentino, Ph.D.

Antonio Sorrentino, Ph.D.

Head of Translational Strategy

Lunaphore Technologies

Dr. Antonio Sorrentino is the Head of Translational Strategy at Lunaphore Technologies, leading strategy inception, planning and execution as well as clinical market development.Antonio has 20 years of experience in life sciences and diagnostics, and a long-standing focus in molecular oncology, translational medicine and biomarker discovery and development. He earned a MSc in Biomolecular Sciences at “Sapienza” University of Rome and a PhD in Molecular Pathology from the Catholic University Medical School in Rome. Prior to Lunaphore, Antonio held multiple senior roles at Exiqon, QIAGEN, Covance/Labcorp Drug Development, where he was responsible for various aspects of global commercial operations including business and market development, primarily with partners from the pharmaceutical and medical device industries.

With the advent of complement therapeutics, it becomes critical to understand whether and how complement contributes to the pathophysiology of diseases affecting the kidney to provide personalized treatments. During the talk, we will illustrate how we developed an ultra-high in situ Complementomics approach using the COMET Imager. We combined the staining for complement proteins, receptors and activation fragments with tissue structure markers and markers for phenotyping of the immune infiltrateThrough this innovative approach, we successfully untangled the intricacies of complement activation within diverse kidney diseases and kidney cancers, as well as its interconnectedness with the infiltration of immune cells. Ultimately, this method sheds light on the role of complement in various disorders, opening doors to the discovery of novel biomarkers and therapeutic strategies.

Watch now →

Speaker

Idris Boudhabhay, M.D.

Idris Boudhabhay, M.D.

Senior Nephrologist, Department of Nephrology and Kidney Transplantation, Necker Hospital
PhD Student, Team “Inflammation, Complement and Cancer”, Centre de Recherche des Cordeliers, INSERM

Necker Hospital | Centre de Recherche des Cordeliers, INSERM

Idris Boudhabhay is an adult nephrologist working in Necker Hospital in Paris and he is currently doing is PhD in cellular biology under the supervision of Prof. Lubka Roumenina in the team “Inflammation, Complement and Cancer” at the Centre de Recherche des Cordeliers, INSERM, in Paris. His research focus on the impact of the complement system in kidney physiology and pathology. He is currently developing a COMPLEMENTOMICS approach using hyperplex imaging to explore the involvement of the complement system in different kidney diseases and kidney cancers in order to identify new biomarkers and therapeutic avenues.

Speaker

Kelly Hunter, MSc

Kelly Hunter, MSc

Chief Scientific Officer

ProPath UK

High dimensional morphological profiling allows the generation of unbiased fingerprints indicative of cell states, and allows identification of genes lying on common pathways or drugs acting via common mechanisms of actions. We have developed a system for pooled genetic screening of iPSC-derived neurons which combines peptide barcoding of short guide RNAs for CRISPR perturbation; a panel of antibodies for morphological and cell state characterization via highly multiplexed immunostaining; and subsequent feature extraction pipelines to reduce the data dimensionality into useful feature vectors. Our pilot experiments validate computationally predicted interactions between genes related to Huntington’s disease.

Sorry, this talk is not accessible on-demand.

Speaker

Sami Farhi, Ph.D.

Sami Farhi, Ph.D.

Director of the Optical Profiling Platform

Broad Institute of MIT and Harvard

Sami Farhi is the director of the Optical Profiling Platform. The platform’s goal is to bring next-generation imaging approaches to bear on large-scale biological problems, with a focus on profiling the spatial organization of tissues, high-content optical screens, and all-optical studies of the electrical properties of cells. He joined the Broad Institute in July 2018 after completing his Ph.D. in chemical biology at Harvard University. He also holds a B.S. in chemical biology from the University of California, Berkeley.

Intraductal papillary mucinous neoplasms (IPMN) of the pancreas are bona fide precursor lesions of pancreatic ductal adenocarcinoma (PDAC). The most common subtype of IPMNs harbors a gastric foveolar-type epithelium, and these low-grade mucinous neoplasms are harbingers of IPMNs with high-grade dysplasia and cancer. To explore the molecular underpinning of gastric differentiation in IPMNs, we conducted spatial transcriptomics on a cohort of IPMNs, followed by hyper-plex cyclic immunofluorescence staining using the COMET platform and cross-species validation studies. Our study identifies NKX6-2 as a key transcription factor driving indolent gastric differentiation in IPMN pathogenesis. 

Sorry, this talk is not accessible on-demand.

Speaker

Jimin Min, Ph.D.

Jimin Min, Ph.D.

Postdoctoral Fellow

The University of Texas MD Anderson Cancer Center

Jimin Min is a postdoctoral fellow at MD Anderson. With over 10 years of experience in gastrointestinal cancer research, Dr. Min has been at the forefront of exploring potential biomarkers for detecting cancer risk across various disease stages, from precancers to metastasis. Her current work harnesses cutting-edge techniques like spatial omics, focusing on pancreatic cancer. Dr. Min’s dedication to unraveling the complexities of tumors and their microenvironments propels her to continuously seek innovative approaches in the field of spatial biology in cancer.

Clinical studies with immune checkpoint inhibitors (ICI) have shown limited response rates, yet several case reports have shown encouraging responses and outcomes. Novel, combined modality treatments, including immunotherapies, have the potential to be effective in chordoma, a disease where conventional cytotoxic chemotherapy and radiotherapy to date have failed to provide meaningful long-term control. To optimize an immunotherapy intervention, we need a comprehensive understanding of the immune composition, the dynamic communication between cells, and the expression of immune checkpoint molecules of chordomas. Here, we comprehensively characterized the immune landscape of chordomas by employing high throughput methods such as single-cell transcriptomics (scRNA-seq), and single-Tcell/Bcell receptor sequence. Our data show that chordomas induce tumor-reactive B- and T-cell immunity inside and outside (blood) the tumor. Spatial transcriptomics and proteomics revealed that most T cells localize in the peritumoral areas, with a broad expression of PD-1 and CTLA-4, and exhausted phenotype. Investigating the cell-cell interactions driving tumor-mediated T-cell exhaustion, we identified M2 tumor-associated macrophages and a plasma cell phenotype-like subset upregulating TGFB and IL10 expression, referred to as regulatory B cells (Bregs). Thus, immunosuppression governs the chordoma microenvironment. However, the existence of tumor-reactive T cells in the circulation that are not subject to the tumor immunosuppressive suggests that a neoadjuvant ICI intervention is preferred over an adjuvant intervention. Using DNA methylome, we established a fast and easy way to define the immune composition of the tumor from biopsies. Thus, the DNA methylome of biopsies might be an effective way to stratify and select patients is a high probability to respond to neoadjuvant ICI.

Sorry, this talk is not accessible on-demand.

Speaker

Catalina Lee-Chang, Ph.D.

Catalina Lee-Chang, Ph.D.

Assistant Professor of Neurological Surgery

Feinberg School of Medicine - Northwestern University

Catalina Lee-Chang is an immunologist, with a strong background in B-cell immunology. She obtained her PhD at the University of Lille (France) where she studied the place of B-cell-mediated immune response in chronic inflammatory diseases such as multiple sclerosis (MS). Dr. Lee-Chang joined the NIH (Baltimore, MD, USA) as a postdoctoral trainee to study the role of cellular B-cell immunity in cancer escape and metastasis. The Lee-Chang is a translational neuro-oncology lab focusing on understanding the interaction between CNS tumors and the immune system. We study how tumors suppress the immune response to leverage current and novel immunotherapies. We are particularly interested in developing genetically engineered B-cell-based therapies that promote an anti-tumoral cytotoxic immune response.

October 5

From pixels to biological insights: the key role of image analysis

Denis Schapiro
Florian Wünnemann
Mauro Gwerder
Farzad Sekhavati
Pedro Espinosa Gonzalez
James R. Mansfield

As the number of biomarkers detected by spatial biology techniques increases, the complexity of images escalates, and their interpretation becomes more challenging. Quantitative data must be extracted from images to infer cellular interactions and dependencies. Speakers showcased novel workflows to transform hyperplex images into biologically relevant datasets that can address basic and translation research questions.

Sorry, this talk is not accessible on-demand.

Speaker

Denis Schapiro, Ph.D.

Denis Schapiro, Ph.D.

Group Leader, Institute for Computational Biomedicine and Institute of Pathology

University Hospital Heidelberg

Dr. Denis Schapiro is a Group Leader at the Heidelberg University Hospital focusing on spatial omics technologies and analysis. Before moving to Heidelberg, he was an Independent Fellow at the Laboratory of Systems Pharmacology at Harvard Medical School and the Klarman Cell Observatory at the Broad Institute where he was a Damon Runyon Quantitative Biology Fellow mentored by Prof. Peter Sorger and Prof. Aviv Regev. Previously, he was supported by the SNFS Mobility Fellowship. Denis obtained his PhD from the University of Zurich and ETH Zurich in the laboratory of Prof. Bernd Bodenmiller. Denis is the lead developer of the histology topography cytometry analysis toolbox (histoCAT) and the multiple-choice microscopy pipeline (MCMICRO).

Highly-multiplexed imaging technologies enable the acquisition of dozens of markers generating large amounts of data in the process. The streamlined processing of such complex data through steps like image preprocessing, single cell segmentation, quantification of marker intensities and cell phenotyping poses a major challenge. In this talk, I will highlight how we utilized MCMICRO to process terabytes of Lunaphore COMET data with a focus on myocardial infarction.

Sorry, this talk is not accessible on-demand.

Speaker

Florian Wünnemann, Ph.D.

Florian Wünnemann, Ph.D.

Postdoctoral Researcher, Institute for Computational Biomedicine and Institute of Pathology

University Hospital Heidelberg, Schapiro Group

Florian Wünnemann is a postdoctoral researcher at the Institute for Computational Biomedicine, University Hospital Heidelberg, under the guidance of Dr. Denis Schapiro. Utilizing single-cell as well as spatial omics technologies, Florian aims to unravel the molecular basis of human diseases, particularly in the realm of cardiovascular health. During his Postdoc, in the Schapiro group, his focus is on developing bioinformatic pipelines for spatial omics analysis to better understand immune cell infiltration during acute myocardial infarction.

Tumor buds are clusters of up to 4 tumor cells that detached themselves from the main tumor. They are a powerful prognostic factor in colorectal cancer among others. However, they are also notoriously cumbersome to annotate and investigate, as there currently exist no phenotypic marker that differentiates them from other tumor cells.

 
We’ve built a pipeline to automatically detect tumor buds on COMET images as well as assess their expression patterns. I will discuss problems and lessons learned when working with sequential immunofluorescence images, and how it is important to take the biological context of the research question into account. 

Sorry, this talk is not accessible on-demand.

Speaker

Mauro Gwerder

Mauro Gwerder

PhD Candidate

Institute of Tissue Medicine and Pathology, University of Bern

I completed my BSc degree at the University of Bern in Cell biology. I continued my studies with an MSc degree in bioinformatics and computational biology, completing a project in the ODYSSEY research group, led by Inti Zlobec. Using a weakly supervised deep learning approach, I worked on the detection of lymph node metastases in lung cancer. I continued my work within the group and started as a Ph.D. student in 2022. I am particularly interested in spatial analysis, image analysis, AI, deep learning, and biostatistics. Odyssey research group: ODYSSEY (digitalpathologybern.com)

Spatial biology enables the interrogation of tissue architecture on a single-cell level. Transforming the rich image output into meaningful quantitative data is the next significant hurdle on the path to fully comprehend and interpret the intricate imaging data. HORIZON™ is an image analysis software tailored to COMET™ hyperplex immunofluorescence images. It seamlessly extends the COMET™ workflow, smoothly handling the large data sets. With its straightforward and user-friendly approach, this analysis tool supports cell-based analysis and provides researchers with a toolset for early and high-level assessment of data in a hyperplex immunofluorescent image. We will walk through image analysis workflow on HORIZON™ from a COMET™ image to biological insights.

Watch now →

Speaker

Farzad Sekhavati, Ph.D.

Farzad Sekhavati, Ph.D.

Project Manager, Image Data Scientist

Lunaphore Technologies

A trained biophysicist, with many years in the field of image data science. I have experienced biology as a physicist, pathology as a data scientist, and project management as software developer. I have always fascinated by the exchange of expertise across fields to grow together.
- Years of experience as image data scientist
- PhD in biophysics
- Masters in micro-fluidics and nanotechnology

The tumor microenvironment (TME) is the seat of multiple cell interactions, including tumor cells, immune cells, stromal cells, and others. The identification of the markers expressed, and their spatial distribution can help not only to establish a prognosis of the disease, but also to direct therapeutic selection.

In this presentation, Mr. Pedro Espinosa Gonzalez presents how the HALO® and HALO AI platform can be used to analyze images of immuno-oncology panels encompassing a highly multiplexed biomarker panel from the COMET™ platform.

Watch now →

Speaker

Pedro Espinosa

Pedro Espinosa

Field Applications Scientist

Indica Labs

Understanding the immune system in solid tissues requires highplex staining, imaging, and analysis for multi-marker cellular phenotypes. We present a comprehensive workflow for highplex images, covering tissue and cell segmentation and cellular phenotyping in a single software package. Visualization templates for marker subsets and pre-trained nuclear segmentation algorithms are reusable and paint-to-train tissue segmentation is easy to perform. Spatial biodistribution metrics, heatmaps and interactive partitioned t-SNE plots can be generated for each tissue type with a minimum of work. This workflow for highplex image analysis enables biologists and immunologists by circumventing the need for expert programming for each specific application. 

Watch now →

Speaker

James R. Mansfield

James R. Mansfield

Senior Vice President

Visiopharm

James R. Mansfield is a scientist with over 30 years of experience, the last 15 of which have been in multiplex pathology and the in situ phenotyping of immune cells. He is currently the Senior Vice President of Research Business Development at Visiopharm A/S, where he is working on their strategy for multiplex pathology, highplex image analysis, and spatial transcriptomics. He is an associate editor of the American Journal of Nuclear Medicine and Molecular Imaging, holds 7 patents, has over 60 publications, and has served as an invited speaker, session chair, and organizer at a variety of international conferences.

October 6

Success stories from technology adoption to data analysis in spatial biology

Joanna Kowal
Bora Gurel
Michael DeCuypere
Shashwat Tripathi
Albert Santamaria-Martínez

Spatial biology is the new frontier in life sciences. However, despite the great potential of spatial techniques, their adoption remains challenging for many laboratories In this session, attendees will discover how spatial biology enables success. From basic research laboratory to translational research group, success stories stemming from Lunaphore’s Access Lab projects featured their journey in quickly adopting spatial biology and integrating a customized workflow in the field of immunology, immuno oncology, and beyond.

Sorry, this talk is not accessible on-demand.

Speaker

Joanna Kowal, Ph.D.

Joanna Kowal, Ph.D.

Senior Scientific Affairs Manager

Lunaphore Technologies

Joanna got fascinated by cell biology during her master’s degree studies at Jagiellonian University, Krakow, Poland. Afterwards she joined Institut Curie, Paris, France, where she developed a proteomic cartography of extracellular vesicles in the frame of her Ph.D. research project. She obtained a Ph.D. degree from Université Paris Descartes in 2016. During her postdoctoral training at the University of Lausanne, Joanna interrogated the interaction within the tumor microenvironment with a focus on immune and malignant cells in the context of primary brain tumors. She joined Lunaphore in 2020 and since then works on bringing spatial biology to every laboratory.

Cancer (PCa) is a predominant male cancer, with rising incidence and progression rates emphasizing the need for effective treatments. Chronic inflammation plays a pivotal role in PCa progression, influencing genetic changes and the tumor environment. This is exacerbated by reactive oxygen species leading to DNA damage, increasing mutation risks, and altering the tumor environment to support growth. Contrary to traditional beliefs, our study of 825 samples revealed significant inflammation in prostate cancer. Using Hyperplex IF techniques, we discerned distinct CD3+ cell patterns in castration-resistant prostate cancer (CRPC), correlating with patient survival. Notably, nodular inflammation patterns hinted at tertiary lymphoid structures (TLSs) fostering immunosuppression. Further analyses differentiated between “hot” and “cold” tumors, and highlighted associations with mature and immature TLSs, potentially indicative of different immune responses. Our findings provide a nuanced understanding of the CRPC immune context, presenting avenues for enhanced immunotherapy and improved patient prognosis in prostate cancer treatment. 

Watch now  →

Speaker

Bora Gurel, M.D., Ph.D.

Bora Gurel, M.D., Ph.D.

Clinician Scientist and Pathologist

The Institute of Cancer Research London

Dr. Bora Gurel completed his medical degree at Ankara University and specialized in pathology at Hacettepe University School of Medicine in Turkey. Driven by an interest in prostate cancer research, he undertook a molecular pathology fellowship at the De Marzo lab at Johns Hopkins Medical School in Baltimore. There, his investigations honed in on the molecular underpinnings of prostate cancer and the role of inflammation in its early stages.
Currently at the Institute of Cancer Research in London, Dr. Gurel's work pivots towards understanding the inflammatory mechanisms associated with castration-resistant prostate cancers (CRPC). His research has led to contributions in several notable publications in the field of prostate cancer.

The immune microenvironment of pediatric gliomas is relatively understudied despite being a leading cause of childhood mortality. Pilocytic astrocytomas (PA) are the most common pediatric glioma and surgical resection is often curative. However, PAs are not always amenable to surgery and can have significant long-term morbidity and mortality. To identify PA patients that may be responsive to immune checkpoint inhibitors and to guide prioritization of available immunotherapeutic treatments, orthogonal immune profiling strategies were used to identify patients that may benefit from the next generation of immune therapeutics. 

Sorry, this talk is not accessible on-demand.

Speakers

Michael DeCuypere, M.D., Ph.D.

Michael DeCuypere, M.D., Ph.D.

Director of Translational Research and Clinical Trials in Neurosurgery, Ann and Robert H. Lurie Children’s Hospital of Chicago
Assistant Professor of Neurological Surgery, Feinberg School of Medicine - Northwestern University

Ann and Robert H. Lurie Children’s Hospital of Chicago | Feinberg School of Medicine - Northwestern University

Dr. Michael DeCuypere received his medical degree and doctorate in neurobiology from the University of Tennessee Health Science Center combined MD, PhD program. He then completed neurosurgical residency training with the Semmes-Murphey Clinic and the University of Tennessee Department of Neurosurgery. He has completed fellowships in Minimally Invasive Neurosurgical Oncology and Complex Endoscopy with Dr. Charlie Teo in Sydney, Australia and Pediatric Neurosurgery with Dr. Rick Boop at LeBonheur Children’s Hospital / St Jude Children’s Research Hospital. His current research interests focus on understanding the complex immunological microenvironment of pediatric brain tumors, including the identification of therapeutic targets for clinical translation.
Shashwat Tripathi, M.D.

Shashwat Tripathi, M.D.

PhD Candidate

Northwestern University

Non-Hodgkin lymphomas are a very heterogenous group of tumors arising from lymphocytes at different stages of differentiation. In order to reliably anticipate responses to treatment, lymphoma models must preserve the spatial organization and the functional interdependency between malignant and non-malignant cells. However, modeling lymphoma ex vivo is challenging due its highly heterogeneous nature and its intrinsic cellular composition. To address these challenges, we have established a tissue explant culture system for murine and human lymphoma that can potentially be used to assess therapy response in lymphoma patients (personalized medicine) and can help uncover novel aspects of lymphoma biology. 

Sorry, this talk is not accessible on-demand.

Speaker

Albert Santamaria-Martínez, Ph.D.

Albert Santamaria-Martínez, Ph.D.

Scientist

Cancer Genomics lab - EPFL

Albert Santamaria-Martínez received his PhD by the University of Barcelona in 2009 and then moved to Switzerland to continue his postdoctoral training at EPFL. His work on translational cancer research is mainly focused on the tumor microenvironment, metastasis, and cancer stem cells. His achievements were recognized with the Pfizer prize in oncology in 2013. He was awarded an SNF Ambizione grant in 2015 and led his own team from 2015 to 2020. He serves as external review for more than 20 international peer-reviewed journals. Currently, he is a scientist in the Cancer Genomics lab (EPFL), and he leads the development of a line of research aiming at establishing a tumor tissue explant platform for personalized medicine and spatial biology research using fresh human tumor samples.