-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.

1780 Chromosome 9p Duplication Promotes T-Cell Exhaustion and Enhances Stem Cell Clonogenic Potential in JAK2-Mutant Myeloproliferative Neoplasms

Program: Oral and Poster Abstracts
Session: 631. Myeloproliferative Syndromes and Chronic Myeloid Leukemia: Basic and Translational: Poster I
Hematology Disease Topics & Pathways:
Fundamental Science, Research, MPN, Chronic Myeloid Malignancies, Diseases, immune mechanism, Myeloid Malignancies, Biological Processes
Saturday, December 9, 2023, 5:30 PM-7:30 PM

Ruggiero Norfo1,2*, Chiara Carretta1*, Sandra Parenti1*, Filippo Badii1,3*, Matteo Bertesi1*, Sebastiano Rontauroli, PhD1*, Lara Tavernari1*, Elena Genovese, PhD1*, Samantha Sperduti4,5*, Elena Enzo1*, Margherita Mirabile1*, Francesca Pedrazzi1*, Chiara Pessina6*, Ilaria Colugnat6*, Barbara Mora, MD7*, Monica Maccaferri, MD8*, Elena Tenedini, PhD9,10*, Silvia Martinelli, PhD11*, Elisa Bianchi, PhD1*, Livio Casarini4,5*, Leonardo Potenza, MD, PhD10*, Mario Luppi, MD8*, Enrico Tagliafico10*, Paola Guglielmelli, MD, PhD12, Manuela Simoni4,5*, Francesco Passamonti, MD13*, Alessandro Maria Vannucchi14 and Rossella Manfredini, PhD, BSc1,2*

1Centre for Regenerative Medicine "S. Ferrari", University of Modena and Reggio Emilia, Modena, Italy
2Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
3Department of Cancer Biology, Thomas Jefferson University and Sidney Kimmel Cancer Center, Philadelphia, PA
4Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
5Center for Genome Research, University of Modena and Reggio Emilia, Modena, Italy
6SMeL Citogenetica e Genetica Medica, ASST Sette Laghi, Varese, Italy
7Division of Hematology, Ospedale ASST Sette Laghi, Varese, Italy
8Hematology Unit and Chair, Department of Medical and Surgical Sciences, Azienda Ospedaliera Universitaria di Modena, University of Modena and Reggio Emilia, Modena, Italy
9Department of Laboratory Medicine and Pathology, Diagnostic Hematology and Clinical Genomics Unit, Modena University Hospital, Modena, Italy
10Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, AUSL/AOU Policlinico, Modena, Italy
11Department of Laboratory Medicine and Pathology, Diagnostic Hematology and Clinical Genomics Unit, Azienda Ospedaliera Universitaria di Modena, Modena, Italy
12Azienda Ospedaliero-Universitaria Careggi, Florence, Italy
13Dipartimento di Oncologia ed Ematologia, Università degli Studi di Milano, Policlinico di Milano, Ospedale Maggiore, Fondazione I.R.C.C.S. Ca Granda, Milano, Italy
14CRIMM, Center for Research and Innovation of Myeloproliferative Neoplasms, Azienda Ospedaliero-Universitaria Careggi, Firenze, Italy

Myeloproliferative Neoplasms (MPNs) are a diverse group of clonal hematopoietic disorders originating from a single hematopoietic stem cell, causing an excessive production of mature blood cells. Classic forms of MPNs include polycythemia vera, essential thrombocythemia, and myelofibrosis. The most common mutation is a gain-of-function point mutation in the JAK2 gene, known as JAK2V617F. This mutation leads to constant activation of the JAK-STAT signaling pathway, resulting in the overgrowth of MPN cells. Furthermore, the variant allele frequency (VAF) of JAK2V617F has a significant impact on the severity and phenotype of the disease. Alongside many described genetic mutations, cytogenetic abnormalities are commonly observed in MPNs, particularly involving chromosome 9. As the JAK2 gene is located on the short arm of this chromosome, we hypothesized that chromosome 9 copy number abnormalities might be a disease modifier in JAK2V617F-mutant MPN patients.

To characterize the biological effects of chromosome 9 copy number abnormalities on JAK2-mutated MPN cells, we analyzed circulating CD34+ hematopoietic stem and progenitor cells (HSPCs) as well as monocytes and granulocytes from 32 MPN patients. Through Next Generation Sequencing, we categorized the patients into three main groups based on JAK2 mutation and copy number status: patients with two copies of the JAK2 gene with either heterozygous (VAF between 20% and 60%; n=10) or homozygous (VAF > 60% n=10) JAK2V617F mutation, or patients carrying mutant JAK2 and gene amplification (n=12).

In-depth analysis of JAK2-amplified patients revealed that the amplification involved the entire chromosome 9p, thus including other gene loci like CD274, which encodes programmed death-ligand 1 (PD-L1). Further investigation of the order-of-events and clonal hierarchies through droplet digital PCR on CD34+ cell-derived colonies showed that most colonies of 9p-duplicated patients had three copies of JAK2, with 2 out of 3 alleles harboring the JAK2 mutation and that point mutations are frequently the initial pathogenic event in clonal evolution, followed by amplification of the JAK2-mutated allele. Functionally, CD34+ cells from +9p patients displayed high clonogenicity and gave rise to a greater number of primitive colonies (Colony Forming Unit-Granulocyte, Erythrocyte, Monocyte, Megakaryocyte, CFU-GEMM).

As JAK2 hyperactivation had been previously reported to lead to increased PD-L1 expression, we further explored the functional significance of CD274 amplification in +9p patients. Our analysis showed increased PD-L1 messenger RNA and protein levels in +9p patient CD14+ monocytes compared to those from patients carrying only two copies of chromosome 9. Moreover, immunofluorescence analysis demonstrated significant re-localization of PD-L1 to the cytoplasmic membrane in monocytes from +9p patients, but not in JAK2V617F-homozygous patients (panel A).

Increased levels of PD-L1, an immune checkpoint known to curb T cell activation, led us to analyze the T cell compartment, which resulted enriched in CD3+/CD8+/CD57-/PD-1+ exhausted T-cells in +9p patients compared to other MPN patients and healthy donors (panel B).

In conclusion, our comprehensive characterization of the molecular interplay between JAK2V617F and chromosome 9 alterations, along with their immunological implications due to PD-L1 hyperactivation, fills a critical knowledge gap and provides valuable insights into the disease progression of 9p-MPNs. Further analysis is ongoing to explore the associations between 9p duplication and hematological parameters in 9p-MPN patients for a better understanding of the clinical implications of this genetic abnormality in MPNs.

Disclosures: Mora: Novartis: Speakers Bureau. Luppi: Novartis: Membership on an entity's Board of Directors or advisory committees; Abbvie: Membership on an entity's Board of Directors or advisory committees; Gilead Sci: Membership on an entity's Board of Directors or advisory committees, Other: Travel Grant; Sanofi: Membership on an entity's Board of Directors or advisory committees; Grifols: Membership on an entity's Board of Directors or advisory committees; Daiichi-Sankyo: Membership on an entity's Board of Directors or advisory committees; MSD: Membership on an entity's Board of Directors or advisory committees; Jazz Pharma: Membership on an entity's Board of Directors or advisory committees. Guglielmelli: GSK: Speakers Bureau; Novartis: Other: Other member of advisory board, speaker at meeting, Speakers Bureau; Abbvie: Other: Other member of advisory board, speaker at meeting, Speakers Bureau. Passamonti: Novartis, GSK, Bristol Myers Squibb, Celgene, Sierra Oncology, AbbVie, Janssen, Roche, AOP Orphan, Karyopharm, Kyowa Kirin, MEI, Sumitomo: Honoraria; BMS: Consultancy, Honoraria, Research Funding; Abbvie: Consultancy, Honoraria; Roche: Consultancy. Vannucchi: AOP: Honoraria; Roche: Honoraria; Abbvie: Honoraria; BMS: Honoraria; GSK: Honoraria; Novartis: Honoraria; Incyte: Honoraria.

*signifies non-member of ASH