-Author name in bold denotes the presenting author
-Asterisk * with author name denotes a Non-ASH member
Clinically Relevant Abstract denotes an abstract that is clinically relevant.

PhD Trainee denotes that this is a recommended PHD Trainee Session.

Ticketed Session denotes that this is a ticketed session.

2692 Unraveling Pathophysiology and Hematopoiesis of Vexas Syndrome By Multi-Omics Analyses and Targeted Gene Editing

Program: Oral and Poster Abstracts
Session: 503. Clonal Hematopoiesis, Aging and Inflammation: Poster II
Hematology Disease Topics & Pathways:
Genetic Disorders, hematopoiesis, Diseases, immune mechanism, Biological Processes, Technology and Procedures, gene editing, pathogenesis, omics technologies
Sunday, December 10, 2023, 6:00 PM-8:00 PM

Raffaella Molteni, PhD1,2,3*, Martina Fiumara, MSc3,4,5*, Corrado Campochiaro, MD1*, Alessandro Tomelleri, MD1*, Elisa Diral, MD6*, Angelica Varesi, MSc7*, Alessandra Weber, MD1,5*, Davide Stefanoni, PhD2*, Roberta Alfieri, PhD8*, Luisa Albano, MSc7*, Maddalena Panigada, PhD2*, Eleonora Cantoni, MSc2*, Daniele Canarutto, MD, PhD9,10*, Luca Basso-Ricci, MSc5*, Pamela Quaranta, PhD5*, Angelo D'Alessandro, PhD11, Marco Matucci-Cerinic, MD12*, Raffaella Di Micco5*, Serena Scala, PhD5*, Alessandro Aiuti, MD, PhD1,5,10*, Fabio Ciceri1,13*, Ivan Merelli, PhD5,8*, Lorenzo Dagna, MD1*, Simone Cenci, MD1*, Giulio Cavalli, MD, PhD1,2,12,14*, Luigi Naldini, MD, PhD1,14 and Samuele Ferrari, PhD5,14

1Vita-Salute San Raffaele University, Milan, Italy
2Division of Genetics and Cell Biology, IRCCS San Raffaele Scientific Institute, Milan, Italy
3co-first author, Milan, Italy
4Vita-Salute San Raffaele University, Milan, ITA
5San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
6Unit of Hematology and Stem Cell Transplantation, IRCCS San Raffaele Scientific Institute, Milan, Italy
7San Raffaele Telethon Institute For Gene Therapy, IRCCS San Raffaele Scientific, Milan, Italy
8Institute for Biomedical Technologies, National Research Council, Segrate, Italy
9San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Italiy, Italy
10Pediatric Immunohematology Unit and BMT Program, IRCCS San Raffaele Scientific Institute, Milan, Italy
11Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO
12Unit of Immunology, Rheumatology, Allergy and Rare diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
13Unit of Hematology and Stem Cell Transplantation, Ospedale San Raffaele, University Vita-Salute San Raffaele, Milan, Italy
14co-last author, Milan, Italy

Background and Rationale

VEXAS (vacuoles, E1 enzyme, X-linked, autoinflammatory, somatic) syndrome is a newly described, treatment-refractory, severe, highly prevalent disease (~1:4,000 males aged >50 years) caused by somatic mutations in the UBA1 (ubiquitin-activating enzyme 1) gene of hematopoietic stem/progenitor cells (HSPCs). VEXAS is hallmarked by vacuoles in myeloid/erythroid precursors and presents with systemic inflammation. About 50% patients develop hematologic malignancies, including myelodysplasia, contributing to the poor prognosis. The pathophysiology and impact of VEXAS syndrome on human hematopoiesis remain elusive. Moreover, the lack of suitable disease models hampers investigation of disease development and preclinical drug testing. To fill these gaps, here we: i) integrated multiparametric flow cytometry and multi-omics analyses on VEXAS patients to achieve comprehensive molecular and phenotypic characterization of hematopoiesis; and ii) successfully developed in vitro and in vivo models of VEXAS syndrome by cutting-edge targeted base editing strategies.

Methods

Multiparametric immunophenotypic analyses and single-cell transcriptomics were performed on peripheral blood and bone marrow (BM) cells from six VEXAS patients (p.Met41>Thr; p.Met41>Val; p.Met41>Leu; c.118-1 G>C) recruited by our Unit. Circulating monocytes were analyzed by whole RNA-sequencing (RNA-seq) and metabolomics. UBA1 mutations were introduced by gene editing technologies in myeloid cell lines, primary T cells and healthy human HSPCs. Engineered HSPCs were used to reconstitute the hematopoietic compartment of immunodeficient mice to generate a humanized VEXAS model. Variant allele frequency of UBA1 mutant cells was quantified by targeted sequencing in isolated hematopoietic lineages and HSPCs.

Results

i) Multiparametric immunophenotypic analyses of BM cells showed unbalanced composition of the HSPC compartment in VEXAS patients compared to age-matched healthy individuals, with lower abundance of stem cells, multipotent and lymphoid progenitors and expanded myeloid progenitors. This was paralleled by a significant increase of circulating myeloid-biased HSPCs and immature myeloid cells. Transcriptomic analyses of patient-derived peripheral monocytes showed sustained VEXAS-associated inflammatory signatures and heightened glycolysis, which were confirmed by metabolomic profiling. Single-cell transcriptomics on the BM hematopoietic compartment of VEXAS patients harboring different UBA1 mutations displayed genotype-dependent gene expression patterns. Single-cell RNA-seq of peripheral blood mononuclear cells from VEXAS patients is ongoing.

ii) In vitro and in vivo models of VEXAS, generated by UBA1 gene editing, faithfully recapitulated patient hematopoiesis and pathophysiology. Editing efficiency approached 90% in wild-type myeloid cell lines, primary human T cells and HSPCs. While UBA1-mutant T cells were counterselected over time in culture, myeloid ones were enriched. Colony forming unit assays of UBA1-edited HSPCs revealed an exclusive myeloid output in vitro and a significant loss of the erythroid HSPC differentiation potential compared to mock-edited controls. Transplantation of edited HSPCs in immunodeficient mice resulted in a 100-fold reduction in circulating B cells, while NK and myeloid compartments were preserved. Human BM HSPCs were 5-fold lower than in control mice, largely myeloid-biased, and displayed abnormal vacuolar morphology. Concordantly, while myeloid cells and HSPCs were mainly (>80%) UBA1 mutated, the lymphoid compartment showed predominance of wild-type alleles. Strikingly, the distribution of UBA1-mutant cells across lineages in humanized mice mirrored the composition of our VEXAS patient cohort, where UBA1-mutant cells accounted for 90% of total myeloid cells and HSPCs and for less than 5% of lymphoid lineages.

Conclusions

Our findings provide evidence that mutations in UBA1 drive accumulation of myeloid-biased HSPCs and immature myeloid precursors. Mutant lymphoid cells are negatively selected and their peripheral myeloid counterpart displays inflammatory activation. Gene editing-based in vitro and in vivo models phenocopy most hematopoietic features of the disease and hold promise to enable preclinical testing and validation of novel therapeutics to treat VEXAS syndrome.

Disclosures: D'Alessandro: Macopharma: Consultancy; Hemanext Inc: Consultancy; Omix Technologies Inc: Current equity holder in private company. Ciceri: ExCellThera: Other: Scientific Advisory Board . Cavalli: Novartis: Current Employment. Naldini: Chroma Medicine: Consultancy, Current equity holder in private company; Genespire: Consultancy, Current equity holder in private company; Genenta: Consultancy, Current equity holder in publicly-traded company, Research Funding.

*signifies non-member of ASH