ѻý

Jump to Main Content

ASH Annual Meeting and Exposition

Scientific Program

Scientific Program sessions are developed by ASH’s 18 Scientific Committees and highlight important basic and translational research topics across the field of hematology. The sessions will take place in person and stream simultaneously on the virtual platform (the recording will be available on demand). Sessions will consist of didactic presentations followed by panel discussions and a question-and-answer period with all speakers. Session descriptions for this program will be available in the Annual Meeting App.

JOINT SESSION: Molecular Characterization of Lymphomas in Children, Adolescents, and Young Adults

Scientific Committee on Blood Disorders in Childhood + Scientific Committee on Lymphoid Neoplasia

Dynamic advances in the molecular characterization of lymphomas continue to shape how these malignancies are classified and inform therapeutic targeting. This joint session will provide a state-of-the-science update on next-generation technologies used to explore that special group of lymphoma subtypes that are most prevalent in pediatric, adolescents, and young adults. The speakers represent experts in these methods and will highlight exciting applications in elucidating lymphoma biology (especially age-related differences in pathogenesis), including enhancing clinical management.

Dr. Lisa Roth will discuss ongoing efforts to delineate the genomic landscape of classic Hodgkin lymphoma (cHL). Historically, genetic evaluation of cHL has been limited by the scarcity of Hodgkin and Reed-Sternberg (HRS) cells in cHL tumors. This has been overcome in recent years using fluorescence-activated cell sorting to isolate HRS cells from cHL tissues and evaluation of circulating tumor DNA, which have revealed tumors with high mutational burden, complex structural variants, and genomic subtypes with distinct clinical and molecular characteristics. The applicability of these findings to develop prognostic biomarkers and novel therapeutics will be discussed.

Dr. Tomohiro Aoki will discuss emerging single-cell technologies that have enabled better understanding of the comprehensive interactions within the cHL tumor microenvironment at an unprecedented resolution. Considering current pathogenesis models where HRS cells “recruit and re-educate” their microenvironment to create an immuno-suppressive milieu, cHL has emerged as one of the most exciting fields for application of these cutting-edge technologies. Dr. Aoki will summarize new insights gained using CyTOF, single-cell RNA sequencing and multiplexed imaging techniques, and discuss potential implications for clinical decisions.

Dr. Birgit Burkhardt will provide a comprehensive update of recurrent genetic variants reported in Burkitt lymphoma (BL) in several recent studies. Covering both endemic and sporadic BL, Dr. Burkhardt will highlight the impact of patient age at diagnosis on molecular characteristics and its implications for pathogenesis. Incorporation of specific variants as potential biomarkers for risk group stratification in upcoming BL clinical trials will also be discussed.

Dr. Björn Chapuy will discuss novel (epi)genomic analyses employed to define molecular heterogeneity in B-cell lymphoma subtypes and identify clinically actionable alterations, with a focus on primary mediastinal large B-cell lymphomas (PBMLs). With biometric features that often overlap with cHL, PMBLs are distinguished by their typical manifestation in young female patients and sensitivity to PD-1 blockade. Dr. Chapuy will present evolving approaches to comprehensive genetic analysis used to inform and test response to therapies that target such mechanisms of immune escape.

Co-Chairs:

DZܳԱ,Ѷ,ѳ
Weill Cornell Medicine
New York, NY

Jean L. Koff, MD,MSc
Winship Cancer Institute of Emory University
ٱԳٲ,Ҵ

Speakers:

Lisa Giulino Roth, MD
Weill Cornell Medicine
New York, NY
Genomic Landscape in Hodgkin Lymphoma

Tomohiro Aoki, MD, PhD
Princess Margaret Cancer Center
մǰDzԳٴ,,䲹Բ岹
Sequencing Technologies in Hodgkin Lymphoma

ܰ󲹰,Ѷ,ʳ
University Hospital Münster
ѳܱԲٱ,ұԲ
Age-Related Molecular Changes in Burkitt Lymphoma

ö󲹱ܲ
Charité - University Medical Center Berlin, Campus Benjamin Franklin
,ұԲ
Functional Genomics of Non-Hodgkin Lymphoma in Children, Adolescents, and Young Adults

back to top

JOINT SESSION: Therapeutic Gene Editing of Stem Cells in Classical and Malignant Hematology

Scientific Committee on Bone Marrow Failure + Scientific Committee on Transplantation Biology and Cellular Therapies

Gene editing is a novel technology to change specific nucleotides or knockouts in cells, and these are starting to be used clinically. This joint-scientific committee session will include 4 talks that span from basic science to clinical use of gene-edited stem cell products and CAR-T products in malignant and classical hematology.

Dr. Justin Eyquem will discuss his latest work using novel gene editing technologies to improve CAR T-cell function and facilitate their manufacturing. He will present new gene edits associated with improved CAR T-cell functional persistence and novel types of vectors for the generation of CAR T cell in vivo.

Dr. Paulo Rio will present the latest results in optimizing novel genome editing tools, including prime editing, to correct hematopoietic stem cells (HSCs) from Fanconi Anemia (FA) patients. She will discuss the latest advances from her lab in optimizing prime editing technology to efficiently target long-term repopulating HSCs and correct the most common FA mutations in both cell lines and primary HSCs from FA patients.

Dr. Pietro Genovese will present the innovative “epitope editing” strategy for overcoming on-target/off-tumor toxicities in acute myeloid leukemia (AML) immunotherapy. Epitope mapping and library screenings have identified specific amino acid changes that have avoided antibody binding. These have now been introduced into HSPCs via base-editing techniques. Epitope-edited HSPC’s were resistant to CAR-T cell therapy, and enabled specific eradication of patient-derived AML xenografts. Dr. Genovese will discuss the potential of precision immunotherapy for relapsed/refractory AML, non-genotoxic conditioning approaches, and advances in multiplex engineering of HSPCs.

Dr. Julia Skokowa will review recent efforts to establish clinical genome editing to correct severe congenital neutropenia-associated mutations as well as gene editing efforts to inhibit mutated genes in patients' HSCs that may be applicable to other bone marrow failure syndromes. She will also discuss key considerations that are particularly important for gene therapy of pre-leukemic bone marrow syndromes, where gene editing of HSCs should be extremely safe so as not to potentiate the leukemogenic transformation of hematopoiesis. Finally, Dr. Skokowa will present her thoughts on the ethics of gene editing for pre-leukemic bone marrow failure.

Co-Chairs:

Marcela V Maus, MD, PhD
Harvard University/Massachusetts General Hospital
DzٴDz,Ѵ

ᲹԲԲղ,Ѷ
Tel Aviv University, Israel
Ramat Gan, Israel

Speakers:

ܲپܱ,ʳ
University of California San Francisco
San Fransisco, CA
New Gene Editing Technologies

ʲܱ龱,ʳ
IIS-FJD, UAM
Ѳ,貹
Gene Editing for Fanconi Anemia Bone Marrow Failure

ʾٰұԴDZ,ʳ
Boston Children's Hospital
DzٴDz,Ѵ
Stem Cell Gene Editing

Julia Skokowa, MD, PhD
University Hospital Tübingen
üԲ,ұԲ
Gene Editing for Severe Congenital Neutropenia – Current Advances and Critical Considerations

back to top

JOINT SESSION: Emerging Therapeutic Strategies Targeting Epigenetic, Transcriptomic, and Metabolic Mechanisms

Scientific Committee on Epigenetics and Genomics + Scientific Committee on Myeloid Neoplasia

Recent developments in therapeutic targeting of various mechanisms of epigenetic, transcriptomic, and metabolic vulnerabilities in cancer utilize novel technologies and may provide future breakthroughs. Given the extensive amount of knowledge derived from high throughput methodologies in the last decade, rational targeting is now possible, with sophisticated mechanistic studies to study the effects of targeting. This session will focus on novel research aimed at identifying and targeting these molecules, including novel targeting approaches such as heterobifunctional molecules and others.

Co-Chairs:

Dinesh S. Rao, MD, PhD
University of California Los Angeles
Los Angeles, CA

ٲԾٲԴǷɲ쾱,ʳ
Cincinnati Children's Hospital
侱ԳԲԲپ,

Speakers:

Hannah J Uckelmann, PhD
Frankfurt Cancer Institute
԰ڳܰ,ұԲ
Novel Inhibitors of Transcriptional Mechanisms

Gerald R. Crabtree, MD
Stanford University
ٲԴڴǰ,䴡
Heterobifunctional Molecules Targeting Epigenetic Mechanisms

Ჹٳ,ʳ
The Scripps Research Institute
San Diego, CA
Inhibitors of RNAs and RNA Binding Proteins.

dzܰٲԱDzԱ,ʳ
Cincinnati Children's Hospital
侱ԳԲԲپ,
Targeting Metabolic Vulnerabilities

back to top

Metabolic Deregulation in Hematopoietic Neoplasia: Tumor, The Environment, and Therapies.

Scientific Committee on Hematopathology and Clinical Laboratory Hematology

Cellular metabolism and its role in the pathogenesis of hematologic malignancies is an emerging area of research. Recent studies implicate various cellular mechanisms by which oncogenic activation of metabolic pathways contribute to lymphoid and myeloid malignancies as well as their respective tumor environment. In this session, the speakers will discuss the role of deregulated cellular metabolism that contributes to lymphoid and myeloid malignancies and how they may provide novel insights with therapeutic relevance. The metabolic control of T cell differentiation and their function in immunity will also be discussed. This session highlights the opportunities for gaining novel mechanistic insights of hematologic malignancies through global metabolomic studies that has potential to impact the field of T-cell immunology, immunotherapies and facilitate translation to clinical practice.

Chair:

Megan S. S Lim, MD,PhD
Memorial Sloan Kettering Cancer Center
New York City, NY

Speakers:

Jürgen Maximilian Ruland
Technical University of Munich
ѳܲԾ,ұԲ
Novel Insights in Metabolic Deregulation in Lymphoid Malignancies

ʲDZҲDZ,Ѷ
Queen Mary University of London
London, United Kingdom
Targeting Metabolic Deregulation in Acute Myeloid Leukemia

ԲԻ岹ҴDZٳ,ʳ
Allen Institute
𲹳ٳٱ,´
Metabolic Control of T Cell Differentiation and Function in Tumor Immunity

back to top

JOINT SESSION: The Fuel & Physiology of Regeneration

Scientific Committee on Hematopoiesis + Scientific Committee on Stem Cells and Regenerative Medicine

The regulation of hematopoietic stem cell (HSC) regeneration involves a sophisticated interplay between signals from the niche and the nuclear regulatory machinery. Extensive work has been done to identify many key players in both areas. However, conveying the signal to the nucleus is not sufficient without the “doers,” the machineries that execute the cellular processes critical for regeneration. Recent studies have identified several vulnerable processes in HSC physiology, such as proteostasis and energy metabolism, which become imbalanced during HSC exhaustion or dysfunction, i.e., processes involved in HSC culture expansion or aging. This session will closely examine the critical physiological processes and their interplay with other regulatory cues, including nutrient-derived signals (minerals and vitamins) crucial for maintaining healthy regeneration without malignant transformation.

Dr. Robert Signer, will discuss how protein homeostasis has emerged as fundamentally and preferentially important in HSCs. He will review how the protein homeostasis network is uniquely configured to promote hematopoietic stem cell self-renewal. Dr. Signer will further discuss how mechanisms of translational control, protein folding, and protein degradation are rewired throughout life to preserve stem cell fitness and will evaluate consequences of protein homeostasis disruption.

Dr. Marie-Dominique Filippi, will discuss the role of metabolism in HSC functions and how HSCs are highly responsive to changes in metabolite availability. New mechanisms will be presented on how HSC metabolic needs are remodeled during regenerative conditions. Additionally, Dr. Filippi will discuss the usage of branch chain amino acids as a new cell-autonomous metabolic checkpoint that influences HSC replicative lifespan.

Dr. Nina Cabezas-Wallscheid, will discuss how mouse and human HSCs and multipotent progenitors are metabolically regulated from intrinsic to dietary-derived metabolites. Through her work using integrated low-input multi-layer omics data, she will highlight research demonstrating distinct metabolic and epigenetic hubs that are essential in HSCs (and not for their downstream progenitors)

Dr. Britta Will, will discuss integrating nutrients into HSC regulation and iron homeostasis in HSCs. Her lab has uncovered a key role of the readily accessible intracellular labile iron pool in instructing HSC self-renewal. More recently, she has focused on identifying the molecular mechanisms of action, particularly focusing on metabolic and non-enzymatic pathways that rely on iron.

Co-Chairs:

Hanna Mikkola, MD PhD
University of California, Los Angeles
Los Angeles, CA

Keisuke Ito, MD, PhD
Albert Einstein College of Medicine
DzԳ,

Speakers:

DzԱ,ʳ
University of California San Diego
La Jolla, CA
Proteomic Health in Hematopoietic Stem Cells

Ѳ-ٴdzԾܱ󾱱辱,ʳ
Cincinnati Children's Hospital Research Foundation
侱ԳԲԲپ,
Energy Management in Hematopoietic Stem Cells

Բ䲹-²𾱻,ʳ
ETH Zürich
ü,ɾٳԻ
Integrating Nutrients to Hematopoietic Stem Cell Regulation

ٳٲ¾,ʳ
Albert Einstein College of Medicine
DzԳ,
Iron Homeostasis in Hematopoietic Stem Cells.

back to top

Paradigm Shifts in Hemostasis: From Mechanisms to Therapies and Back

Scientific Committee on Hemostasis

Coagulation is not a linear process that ends at clot formation. Rather, coagulation is a network of ever-growing complexity with many mechanisms and pathways for regulation and feedback. The consequences of coagulation extend well beyond clot formation and into processes related to immunity, inflammation, vascular biology and regeneration. This session will highlight three examples for how bi-directional interactions from bench to bedside and back have paved the way to paradigm shifts in mechanistic insights, diagnosis, and treatment options for bleeding disorders and thrombotic complications.

Dr. Annette von Drygalski will discuss the most recent therapeutic paradigms in hemophilia. The continued unraveling of genetic and biochemical mechanisms underlying impaired blood clotting remains critical to inform the development of groundbreaking therapeutic strategies for Hemophilia A and B. Such strategies include extended half-life clotting factor preparations, non-factor-based molecules modifying hemostasis and, lastly, gene therapy. The new therapies have great potential to improve joint bleed prevention and reduce the sequelae of hemophilic arthropathy, which remains one of the most prominent aspects of clinical management in daily practice. Dr. von Drygalski will discuss pathophysiological mechanisms contributing to hemophilic arthropathy, new imaging modalities and joint outcome measures, paving the way for more targeted and individualized treatment strategies.

Dr. Daniël Verhoef will discuss the discovery and (pre)clinical development of a modified [PR1] recombinant form of coagulation factor X, called VMX-C001, which can bypass factor Xa direct oral anticoagulants (DOACs) in patients requiring immediate reversal of anticoagulation. It carries a unique 16 amino acid insertion that was derived from a snake venom factor X that makes VMX-C001 insensitive to inhibition by FXa DOACs. A single intravenous administration of VMX-C001 was shown to restore coagulation assays in animals and human volunteers on FXa-DOACs. VMX-C001 also prevented rivaroxaban induced bleeding in a primate liver injury model. VMX-C001 holds promise as an effective FXa DOAC reversal agent given its favorable half-life, short infusion time, FXa DOAC independent dosing and lack of a prothrombotic effect.

Dr. Cristina Puy will present a comprehensive exploration of coagulation factor XI dynamic interaction with diverse ligands, substrates, and endothelial cell receptors. Delving into the intricate mechanisms, Dr. Puy will elucidate factor XI pivotal role in modulating endothelial cell permeability and barrier function, shedding light on its implications in thrombotic and inflammatory conditions. This presentation will deepen our understanding of factor XI multifaceted involvement in health and disease.

Chair:

Laurent O. Mosnier, PhD
The Scripps Research Institute
La Jolla, CA

Speakers:

Annette von Drygalski, MD, PharmD, RMSK
University of California San Diego (UCSD)
SAN DIEGO, CA
Paradigm Shifts in Hemophilia.

Daniël Verhoef, MSc, PhD
VarmX B.V.
𾱻,ٳԻ
Paradigm Shifts in the Reversal of DOACs

Cristina Puy Garcia, PhD
Oregon Health and Science University
ʴǰٱԻ,
New Mechanisms That May Fuel the Paradigm Shifts of the Future

back to top

Host Factors Driving Anti-Leukemia Immunotherapy Outcomes

Scientific Committee on Immunology and Host Defense

This session will cover how the immune microenvironment of the host interacts with leukemia cells and how the donor immune system can exert graft-versus-leukemia (GVL) effects. The speakers will focus on the characterization of immune effector cells and leukemia cells using high resolution RNA and protein analysis, the characterization of metabolites that impact the GVL effect and kinase inhibition strategies to overcome immune escape mechanisms of leukemia cells.

Dr. Catherine Wu will discuss the components critical to devising effective immunotherapy for the treatment of myeloid leukemia, namely the identification of myeloid malignancy antigen targets, and the detection of critical immune cell effectors. The analysis of GvL responses following allogeneic stem cell transplant for AML/MDS, through single cell transcriptome and spatial analysis of marrow specimens, is a particularly instructive setting from which to detect key antigens and cell subpopulations implicated in anti-leukemia responses. The insights gained from the study of such GVL settings promise to provide a roadmap for devising new immunotherapeutic approaches for myeloid malignancies.

Dr. Takanori Teshima will discuss mechanisms of leukemia relapse after allogeneic hematopoietic stem cell transplantation and novel strategies to prevent relapse by inhibiting immune escape of leukemic cells, restoring immune cell dysfunction, and turning “COLD” into “HOT” immune microenvironment. In addition, novel strategies to separate GVL from GVHD by controlling migration and activation of donor T cells in the vicinity of leukemic cells.

Chair:

Dzܱ𾱲,Ѷ
Universitätsklinikum Freiburg
𾱲ܰ,ұԲ

Speakers:

Catherine J. Wu, MD
Harvard Medical School
DzٴDz,Ѵ
Immune Microenvironment and GVL Effects

ղ첹Դǰձ󾱳,Ѷ,ʳ
Hokkaido University
ǰ,ǰ첹,貹
Firing Up COLD Tumor Microenvironment to Facilitate GVL

Գ첹,Ѷ,ʳ
German Cancer Research Center (DKFZ)
𾱻,ұԲ
Immune Metabolism in T Cells Acting Against Leukemia Cells

back to top

Crosstalk Between Iron Homeostasis and Metabolism

Scientific Committee on Iron and Heme

Maintaining iron balance is fundamental to preserve metabolic functions and energy requirements. This session reflects the current understanding of the reciprocal relationship existing between iron homeostasis and energetic metabolism. It aims to highlight the regulatory role of heme and iron on glucose and lipid metabolism as well as the impact of metabolism on the modulation of the iron status. Novel findings will be presented about the physiologic and pathologic implications of iron-mediated regulation of metabolism, and metabolite-driven control of iron and heme homeostasis. The session will focus on b-thalassemia, sickle cell disease and obesity, and will have broad relevance to all hematologic and non-hematological diseases.

Dr. Antonella Nai will discuss the role of Transferrin Receptor2 (TFR2) in the reciprocal regulation between iron homeostasis, erythropoiesis, and glucose metabolism. TFR2 is an iron sensor acting as a brake of erythropoietin signaling in erythroid cells. Its genetic inactivation in the hematopoietic compartment enhances erythropoiesis and improves anemia in ß-thalassemia. She will present unpublished data demonstrating that Tfr2 deficiency increases the metabolic activity of erythroid cells, thus promoting glucose consumption and reducing blood glycemia. This is particularly relevant in ß-thalassemia, whereby glucose intolerance and diabetes are common and invalidating complications.

Dr. Wei Ying will focus on the connection between iron homeostasis and lipid metabolism. He will present findings related to the role of iron homeostasis in regulating hepatocyte lipogenesis and neighboring cell responses, including hepatic stellate cells, and describe how alterations of these mechanisms drive non-alcoholic fatty liver disease, steatohepatitis and obesity.

Dr. Leitinger will share how heme triggers a unique bioenergetic switch in macrophages, characterized by a metabolic shift from oxidative phosphorylation towards glucose consumption, and its critical role in the effective clearance and detoxification of heme in sickle cell disease. Based on the previous identification of the phosphofructokinase PFKFB3 as key mediator of the metabolic switching in heme-detoxifying macrophages, he will present data about a potential therapeutic strategy involving the inhibition of PFKFB3 in a mouse model of sickle cell disease. Furthermore, the general clinical relevance of these processes for patients with hemolytic disorders will be discussed.

Chair:

ԳվԳ,ʳ
New York Blood Center
New York, NY

Speakers:

ԳٴDzԱ,ʳ
IRCCS San Raffaele Scientific Institute
Ѿ,ٲ
Iron, Erythropoiesis, and Glucose Metabolism

±۾Բ,ʳ
University of California San Diego
La Jolla, CA
Iron and Lipid Metabolism

ǰ𾱳پԲ
University of Virginia
󲹰dzٳٱ,մ
Heme and Macrophage Metabolism.

back to top

JOINT SESSION: Newly Described Functions in the Vascular Space

Scientific Committee on Megakaryocytes and Platelets + Scientific Committee on Thrombosis and Vascular Biology

Dysregulated interactions between platelets, immune cells, endothelial cells, and their plasma environment underlie numerous age-related diseases. This joint session will provide an integrated view of the vascular space and will highlight the cellular and molecular mechanisms underlying diseases that primarily afflict the elderly. Specifically, the session will also on novel discoveries addressing how platelet and endothelial aging may affect neurodegenerative disorders and thrombotic disease, and immune thrombocytopenia (ITP), and will shed light into the role of fibrinolytic factors in shaping the pro-atherogenic plasma lipid profile.

Dr. Claus Nerlov, will discuss how aging affects hematopoietic stem cells (HSCs). He will describe how increased HSC platelet bias due to how age-related increases in TGFbeta signaling is an evolutionarily conserved feature of murine and human hematopoiesis. The role of HSC platelet bias in suppressing lymphoid cell production during aging and how TGFbeta receptor inhibition can increase the ability of aged HSCs to support antiviral immunity will also be discussed.  

Dr. Nichola Cooper, will discuss the current understanding of ITP pathogenesis and how the results from novel treatments in ITP may depict different disease types in ITP. ITP is a common bleeding disorder with an increase in incidence after age 60. Understanding the disease phenotype and evolution is needed to improve patient outcomes. However, the diagnosis remains presumptive, and treatment is largely empiric, with many patients rotating through several treatments before having a platelet response.

Dr. Lisa Lesniewski will discuss the phenotype of the aging vasculature and the hematopathology underlying mechanisms and potential therapeutic strategies being explored to reverse arterial aging to reduce cardiovascular (CVD) risk in older adults. This is important since several factors contributing to dysfunction of the endothelium (i.e., oxidative stress and inflammation, deregulated nutrient sensing, and cellular senescence) increase with aging and could impact CVD risk.

Dr. Ze Zheng will discuss research demonstrating that hepatocytes are an unappreciated source of tissue-type plasminogen activator (tPA), sense metabolic stresses, and impact the production of tPA and its inhibitor PAI1, thereby influencing the extent of fibrinolysis in obesity. She will introduce findings that tPA binds directly to hepatocyte apoB, preventing it from being loaded with lipids and assembled into lipoproteins. Conversely, tPA's inhibitor, PAI1, has the opposite effect, sequestering free tPA and preventing its interaction with apoB to promote VLDL assembly and increase apoB-lipoprotein cholesterol levels.

Co-Chairs:

Lijun Xia, MD, PhD
Oklahoma Medical Research Foundation
Oklahoma City, OK

Ჹٳܳ±𾱱,ʳ
Versiti Blood Research Institute
Ѿɲܰ,±

Speakers:

ܲDZ,ʳ
University of Oxford
Oxford, United Kingdom
Platelets and Aging

DZǴDZ,Ѷ
Imperial College
London, ENG, United Kingdom
New Insights in MK/PLT Functions and Diseases.

Ծɲ쾱,ʳ
University of Utah Health
Salt Lake City, UT
Age-Related Endothelial Function

ܱܳԲ,ѵ,ʳ
Medical College of Wisconsin; Versiti Blood Research Institute
Ѿɲܰ,±
Novel Functions of Fibrinolytic Factors in Shaping the Pro-Atherogenic Lipid Profile in Plasma.

back to top

Leveraging Single Cell Multi-Omics to Understand Mechanisms of Myeloid Lineage Expansion in Aging and Disease.

Scientific Committee on Myeloid Biology

Single cell multi-omic studies of normal and malignant hematopoiesis highlight how hematology remains an ideal testing ground for technologic innovation. Continued advancements in single cell sequencing and the analysis of these complex datasets have informed functional experiments that have enabled the field to improve its understanding of hematopoietic stem cell behavior and myeloid differentiation in response to aging, inflammation, and malignant transformation. This session will showcase innovative transcriptomic and functional genomic studies of the pathways that govern myeloid differentiation in response to aging, inflammation, and malignancy. The talks will focus on how advances in single cell lineage tracing have improved the fields’ understanding of hematopoietic stem cell commitment to the myeloid lineage early in life; explore how aging and inflammatory conditions impact myelopoiesis; and address how the tumor microenvironment can impact the function of mature myeloid cells.

Chair:

Esther A. Obeng, MD, PhD
St. Jude Children's Research Hospital
ѱ𳾱󾱲,ձ

Speakers:

ܰԳپ,ʳ
University of Cambridge
Cambridge, ENG, United Kingdom
Hematopoietic Stem Cell Commitment to the Myeloid Lineage

John E. Dick, PhD
Princess Margaret Cancer Centre, University Health Network
մǰDzԳٴ,,䴡
Hematopoietic Stem Cell and Myeloid Progenitor Cell Response to Inflammation

Allon Moshe Klein, PhD
Harvard Medical School
DzٴDz,Ѵ
Myeloid Progenitor and Mature Neutrophil Responses in Cancer.

back to top

Proteo-Genomics to Better Study Multiple Myeloma Biology and Evolution

Scientific Committee on Plasma Cell Neoplasia

Cancer proteogenomics provides new biological and diagnostic knowledge that can improve our understanding of malignant transformation and therapeutic outcomes. This area is new in plasma cell neoplasia. In this session the speakers will discuss how proteogenomics can lead to a better study of multiple myeloma (MM) biology and support the development of novel therapeutic targets in MM. 

Dr. Jan Krönke will discuss the impact of proteogenomic studies on our understanding of MM biology. He will outline how proteomics improve risk stratification, and reveal deregulated proteins and pathways that can be exploited for new therapies in MM. 

Dr. Francesco Maura will present the genomic landscape of MM and its precursor conditions, with a particular focus on the temporal acquisition of various combinations of genomic drivers. He will highlight the relevance of these genomic patterns in predicting MM progression and identifying the most effective immunotherapy for MM patients. 

Dr. Fabiana Perna will describe a strategy for probing the MM surface proteome (surfaceome) with mass-spectrometry and transcriptomic analyses to identify novel immunotherapeutic targets. She will discuss the contribution that the surfaceome makes to MM biology as well as the pre-clinical development of novel chimeric antigen receptor platforms for MM.

Chair:

ʲDZ,Ѷ,ʳ
Arnie Charbonneau Cancer Institute, University of Calgary
䲹,,䲹Բ岹

Speakers:

Dz԰,Ѷ
Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt Universität zu Berlin, and Berlin Institute of Health
,ұԲ
Defining the Proteomic Landscape of Multiple Myeloma

ԳѲܰ,Ѷ
University of Miami
Palmetto Bay, FL
Genomic Drivers Involved in Myeloma Progression and Development of Resistance

Fabiana Perna, MD, PhD
H. Lee Moffitt Cancer Center and Research Institute
ղ貹,
Proteo-Genomics to Better Study Cell Surfaceome in Multiple Myeloma

back to top

Ineffective Erythropoiesis: Insights Into Molecular Mechanisms and Disease Pathophysiology

Scientific Committee on Red Cell Biology

Ineffective erythropoiesis is the leading cause of red cell-related diseases, including anemia, which stands as the most prevalent hematologic disease, affecting millions of individuals worldwide. Understanding the underlying mechanisms of ineffective erythropoiesis is pivotal for advancing diagnostic and therapeutic approaches in hereditary anemias and adult-onset conditions. This session will explore recent breakthroughs in research related to ineffective erythropoiesis, focusing on areas of hemoglobinopathies, malaria infection, and myeloid neoplasms. By bringing together experts at the forefront of these investigations, the session will not only enhance understanding of the pathogenesis of ineffective erythropoiesis but also catalyze the development of innovative approaches to treat related diseases.

Chair:

ʱԲ,Ѷ,ʳ
Northwestern University Medical School
󾱳,

Speakers:

Wassim El Nemer, PhD
French Blood Establishment (EFS)
Ѳ𾱱,Գ
Post-Transcriptional and Signaling Mechanisms in Ineffective Erythropoiesis in Hemoglobinopathies

Elizabeth S. Egan, MD, PhD
Stanford University School of Medicine
ٲԴڴǰ,䴡
Plasmodium Falciparum Influences Erythropoiesis

ٴdzܱٴDZ,ʳ
Division of Hematology, Department of Medicine
𲹳ٳٱ,´
Ineffective Erythropoiesis in Myeloid Neoplasms

back to top

Understanding the Impact of Donor and Recipient Metabolic Variability on Blood Transfusion Outcomes

Scientific Committee on Transfusion Medicine

Blood transfusion requires the donation of blood products from a diverse array of individuals with unique genetics and environmental inputs. This results in the quality and overall behavior of the blood products being less standardized and predictable, with direct consequences on transfused recipients. This session will focus on how recently recognized unique genetic and environmental backgrounds of both the blood donor and the transfusion recipient influence the outcomes of patients. Newly applied metabolic tools reveal metabolic variability in donors and recipients, shedding light on fundamental aspects of these differential responses of transfusion in patients, which could guide future strategies for more personalized and effective blood transfusions.

Chair:

Cassandra Dorothy Josephson, MD
Johns Hopkins All Children’s Hospital
St. Petersburg, FL

Speakers:

Բ'Ի,ʳ
University of Colorado Anschutz Medical Campus
ܰǰ,䰿
Blood Donor Genetics and Biology Impact Red Blood Cell Transfusion Outcomes

ѴǰٳٴDZ,Ѷ
Bloodworks and University of Washington, Pathology and Laboratory Medicine
𲹳ٳٱ,´
Unveiling Platelet Metabolism: Implications for Storage and Transfusion

Dz,ʳ
Laboratory of Blood-Borne Parasites, Lindsley F. Kimball Research Institute, New York Blood Center
New York, NY
Impact of RBC Metabolism on the Disease Outcomes of Transfusion Dependent Patients