-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.

4076 Spatial Transcriptomics Unveils Novel Potential Mechanisms of Disease in a MIcγ1 Multiple Myeloma in vivo Model

Program: Oral and Poster Abstracts
Session: 506. Bone Marrow Microenvironment: Poster III
Hematology Disease Topics & Pathways:
Research, Translational Research, Plasma Cell Disorders, Diseases, Lymphoid Malignancies, Biological Processes, molecular biology, Technology and Procedures, Human, pathogenesis, Animal model, omics technologies, Pathology
Monday, December 11, 2023, 6:00 PM-8:00 PM

Laura Sudupe1*, Emma Muiños-Lopez, PhD2*, Isabel A Calvo, PhD2*, Ana Rosa Lopez-Perez, PhD3*, Amaia Vilas-Zornoza4*, Sarai Sarvide5*, Purificacion Ripalda-Cemborain2*, Azari Bantan1*, Jin Ye1*, Vincenzo Lagani1*, Paula Aguirre-Ruiz2*, Patxi San-Martin2*, Itziar Cenzano, PhD4*, Marta Larrayoz, PhD2*, Laura Alvarez-Gigli, MD6*, Marta Abengozar-Muela, MD6*, Borja Saez-Ochoa, PhD2*, Phillip T Newton7*, Jesper Tegner, PhD1*, Bruno Paiva2,8*, Jose A. A Martinez-Climent, MD2,5, David Gomez-Cabrero1,9* and Felipe Prosper, MD, PhD4,8,10

1Bioscience Program, Biological and Environmental Sciences and Engineering Division (BESE), King Abdullah University of Science and Technology KAUST, Thuwal, Saudi Arabia
2Hematology and Oncology Program, Centre for Applied Medical Research (CIMA), Instituto de Investigaciones Sanitarias de Navarra (IdiSNA), Cancer Center Clinica Universidad de Navarra (CCUN), Pamplona, Spain, Pamplona, Spain
3Translational Bioinformatics Unit, Navarrabiomed, Universidad Pública de Navarra (UPNA), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Navarra, Spain., Pamplona, Spain
4Hematology and Oncology Program, Centre for Applied Medical Research (CIMA), Instituto de Investigaciones Sanitarias de Navarra (IdiSNA), Cancer Center Clinica Universidad de Navarra (CCUN), Pamplona, Spain
5Cancer Center Clinica Universidad de Navarra, Centro de Investigación Médica Aplicada (CIMA), IDISNA, CIBER-ONC number CB16/12/00369 and CB16/12/00489, Pamplona, Spain, Pamplona, Spain
6Department of Pathology, Clinica Universidad de Navarra (CUN), Pamplona, Spain
7Department of Women’s and Children’s Health, Karolinska Institutet, Sweden; Astrid Lindgren Children´s hospital, Stockholm, Sweden, Stockholm, Sweden
8Cancer Center Clinica Universidad de Navarra, Centro de Investigación Médica Aplicada (CIMA), IDISNA, CIBER-ONC number CB16/12/00369 and CB16/12/00489, Pamplona, Spain
9Translational Bioinformatics Unit, Navarrabiomed, Universidad Pública de Navarra (UPNA), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
10Hematology and Cell Therapy Department, Clinica Universidad de Navarra, Instituto de Investigaciones Sanitarias de Navarra (IdiSNA), Cancer Center Clinica Universidad de Navarra (CCUN), Pamplona, Spain

Multiple Myeloma (MM) is a malignant plasma cell (PC) disorder characterized by a heterogeneous distribution of PCs in the bone marrow (BM) where the interactions between the PCs and the BM microenvironment guide the pathogenesis of the disease. The development of single cell technologies has contributed to understand the transcriptional heterogeneity of the disease both from the tumor and the microenvironment point of view, yet the spatial organization of cellular states and niche-specific regulatory programs remain to be investigated.

To address the role of spatially resolved interactions in MM, we performed spatial transcriptomic analysis in BM from the MIcγ1mice strain (Larrayoz et al, Nat Med. 2023) a recently described mouse model that recapitulates the characteristics of a MYC driven human MM disease using the Visium Spatial Gene Expression analysis (10x Genomics).

Using formalin-fixed paraffin-embedded (FFPE) BM tissues from the femur of control mice, we first characterized the healthy BM. Considering that each spot contains an averaged transcriptomic profile of 3-10 cells, we generated a single-cell reference set that included the most prevalent cell types in the BM to identify the cell type contribution per spot. The relative proportions of the estimations are consistent with percentage estimations observed by previous works (Hongzhe et al, eLife. 2023). Next, using the “cell proportion estimations per spot” we identified four different clusters of spots. A deeper characterization of those clusters revealed a positive correlation between erythroblasts and B cells, and a negative correlation between both cell types and neutrophils. Additionally, we spatially defined clusters 1 and 2, those with larger proportion of erythroblasts, in the metaphysis trabecular regions of the bone marrow. We followed the same analysis framework to characterize the spatial configuration in the MIcγ1 mice BM with a developed MM. To this end, we incorporated the MM-derived single-cell PCs in the single-cell reference set. Importantly, the pathological PCs were identified and organized in large groups in the BM space. Cell proportion estimations in these samples revealed a distribution in 7 clusters, 4 of them concentrated in the periphery of the PC hot spots and 3 mirroring the PC gradient observed in previous analysis (Figure 1A).

We next performed a targeted differential analysis to confirm previous observed markers identities of different MM processes as Cd44 (de Jong et al, Nat Immunol. 2021), a signature of dormant cells (Khoo et al, Blood. 2019), the 38 MM-associated surface-protein-encoding genes (Yao et al, Cancer Res. 2023), Mmp9 and different profiles of T cell exhaustion markers within the 3 clusters with higher PC concentration. These analyses allowed us to verify two different markers, Cd44 and Mmp9 localized in the periphery of the hot spots of PC which may explain an invasive mechanism of the cells localized in the outer areas of the MM combining an inflammatory environment, manifested by a Cd44 increment, with a degradative mechanism driven by Mmp9. Simultaneously, an untargeted differential analysis aimed to identify markers characterizing the clusters with high PC prevalence, revealed novel marker genes never described before in MM and not associated with PC proportion, as the Transmembrane Immune Signaling Adaptor (Tyrobp) in the cluster 5 (peripheral cluster), which is predictive of a poor prognosis and high tumor immune infiltration in other described tumors (Lu et al, BMC Cancer 2021) (Fig 1B).

Finally, as a proof of principle of this technology potential in human samples, spatial transcriptomic was applied to FFPE samples from BM biopsies of 7 MM patients with different degrees of PC infiltration, providing a source for validation of the results obtained in the mouse model.

In conclusion, our findings demonstrate that the application of spatial transcriptomics represents a useful tool for understanding the spatial architecture and niche specific interactions in human diseases, offering a systemic approach to dissect the role of the spatial interactions in the pathogenesis of MM.

Disclosures: Paiva: Roche Glycart AG: Honoraria, Research Funding; Bristol-Myers Squibb: Consultancy, Honoraria, Research Funding; Janssen: Consultancy, Honoraria; Sanofi: Consultancy, Honoraria, Research Funding; EngMab: Research Funding; GSK: Honoraria, Research Funding; Adaptive: Honoraria; Takeda: Honoraria, Research Funding; Amgen: Honoraria; Gilead: Honoraria; Oncopeptides: Honoraria. Martinez-Climent: Roche-Genentech: Research Funding; BMS-Celgene: Research Funding; Janssen: Research Funding; Priothera: Research Funding; Palleon: Research Funding; AstraZeneca: Research Funding.

*signifies non-member of ASH