Cancer Genomics and Immune Evasion

In collaboration with Drs. Tim Ley, Matthew Walters, Dan Link, and Li Ding’s laboratories we identify novel multiple myeloma (MM) and AML targets by RNA expression and proteomics and investigate the potential utility of targeting them. Changes in gene expression often accompany disease progression and immune evasion resulting in relapse.


(Dr. Michael Rettig, Dr. Ezhil Chendamarai, Tianjaou (Julia) Wang, Dr. Reyka Jaysinghe , Dr. Dilan Patel, Omar Ibrahim, and Julie Ritchey)

 
DrawImpacts_Animation_web.gif
arrow3Grey.png
 
 

To evaluate single-cell RNA-sequencing we utilize multiple computational tools developed both in-house and publicly available to annotate our dataset. Integration of 4 cases of multiple myeloma from bone marrow aspirates highlighted distinct cell type populations that show patient specific variability in the tumor cells and microenvironment. Integrating with normal bone marrow samples allows us to study unique tumor specific populations of the B and plasma cell lineage. 

 
 

Cancer Genomics

Multiple myeloma is defined by aberrant monoclonal proliferation of plasma cells in the bone marrow. This remains an incurable disease despite advances in treatment. Key genetic and epigenetic alterations that drive MM pathogenesis are known, but we are endeavoring to create a comprehensive profile of affected cellular pathways. We integrate whole-genome and whole-exome sequencing data with single-cell RNA sequencing (scRNA-seq) data across multiple treatment stages to 1) assess differential pathway enrichment between tumor subpopulations, 2) trace the clonal evolution of dominant disease mechanisms, and 3) investigate signaling interactions between surrounding cell types. Our findings advance the understanding of how MM tumor subpopulations differentially regulate cellular pathways and interact within the tumor microenvironment.(1)

We also use genomics to attempt to overcome a major limitation in the development of immunotherapies: discovery of optimal candidate targets.  These require both high expression in tumor cells as well as stringent tissue specificity. To identify potential myeloma and AML-specific target antigens, we perform unbiased searches for genes with specific expression in plasma and/or B cells using single cell RNA-seq (scRNA-seq), single cell Assay for Transposase-Accessible Chromatin (scATAC-seq), single cell proteomics, whole exome sequencing and bulk RNA-sequencing. Our strategies have enabled the discovery of myeloma-associated therapeutic target candidates, including tumor-specific neoantigens and tumor-associated antigens. Similar studies using AML samples are ongoing. These studies are enabling us to develop novel treatment options (i.e., CAR-T, bispecific antibody, vaccine, etc.) for patients with myeloma or AML.

  1. Text in this paragraph first appeared in its entirety in: Wang, et al., Blood. 134 (Supplement 1): 364

Immune Evasion

We are also studying the molecular mechanisms that underlie relapse after bone marrow or stem cell allogeneic transplantation which is currently the only curative therapy for AML. Using mouse models and human specimens we strive to identify and characterize the genomic and transcriptional variants that provide long-term, relapse-free survival. Using the former we generated allogeneic T cell-resistant (alloTR) murine AML cells in vivo by applying immunological pressure via serial alloHSCTs with MHC-matched, mHA-mismatched alloHSCT. We are searching within the transcriptome of these alloTR cells for dysregulation of minor histocompatibility antigens, immune-related genes or other metabolic pathways.

 
Immune evasion and relapse figure copy.png
 

In patients, we recently found several genes involved in major histocompatibility class II (MHCII) antigen processing and presentation are significantly downregulated after alloHSCT but not after chemotherapy. Significantly, we find that treating cell lines or patient cells with IFN-γ rapidly re-induces MHCII expression and re-sensitizes the cells to cytolysis in mixed lymphocyte reactions.

Bispecific antibodies bypass MHC restriction by creating an artificial T cell synapse between effector T cells and target cells. We study samples from patients with AML who were treated with bispecific antibodies targeting CD123 and CD3 (flotetuzumab, NCT02152956) or CD33 and CD3 (AMV564, NCT03144245).  Using 21-color flow cytometry we simultaneously characterize tumor markers and the T cell subsets in the samples.  Likewise, using a Luminex assay, we assess the concentration of 18 different serum cytokines, some of which (IL-6 and interferon-γ) play a role in cytokine release syndrome.  This led us to demonstrate that MHCII can be upregulated on human MHCII-negative blasts both in vitro and in mice by either flozetuzumab or chimeric antigen receptor T (CART) cells in an IFNγ-dependent manner.  Since MHCII upregulated blasts were killed more effectively by third-party mismatched human CD4 T cells than were untreated controls, this insight may provide a potential means to overcome relapse. We are examining this in a phase 2 clinical trial of flotetuzumab in patients with relapsed or refractory AML after allogeneic stem cell transplant (NCT04582864).

We also collaborate with other academic or industry partners to assess proprietary molecules in preclinical studies that have the potential to upregulate MHCII.

 

 Key Publications

 

  • Ley TJ, Mardis ER, Ding L, Fulton B, McLellan MD, Chen K, Dooling D, Dunford-Shore BH, McGrath S, Hickenbotham M, Cook L, Abbott R, Larson DE, Koboldt DC, Pohl C, Smith S, Hawkins A, Abbott S, Locke D, Hillier LW, Miner T, Fulton L, Magrini V, Wylie T, Glasscock J, Conyers J, Sander N, Shi X, Osborne JR, Minx P. Gordon D, Chinwalla A, Zhao Y, Ries RE, Payton JE, Westervelt P, Tomasson MH, Watson M, Baty J, Ivanovich J, Heath S, Shannon WD, Nagarajan R, Walter MJ, Link DC, Graubert TA, DiPersio JF, and Wilson RK.  DNA sequencing of a cytogenetically normal acute myeloid leukaemia genome. Nature 456(7218):66-72;2008.

  • Ding L, Ley T, Larson D, Welch J, Koboldt D, Ritchey J, Young M, Lamprecht T, McMichael J, McLellan M, Harris C, Dooling D, Fulton R, Fulton L, Wallis J, Chen K, Schmidt H, Kalicki-Veizer J, Magrini V, Cook L, McGrath S, Vickery T, Wendl M, Link D, Tomasson M, Shannon W, Payton J, Kulkarni S, Watson M, Westervelt P, Walter M, Graubert T, Mardis E, Wilson R, and DiPersio J. Clonal Evolution of Acute Myeloid Leukemia at Relapse.  Nature 481;506-10;2012.  PMID 22237025.

  • Christopher MJ, Petti AA, Rettig MP, Miller CA, Chendamarai E, Duncavage EJ, Klco JM, Helton NM, O’Laughlin M, Fronick CC, Fulton RS, Wilson RK, Wartman LD, Welch JS, Heath SE, Baty JD, Payton JE, Graubert TA, Link DC, Walter MJ, Westervelt P, Ley TJ, DiPersio JF. Downregulation of immune regulatory pathways in relapsed AML cells after allogeneic transplantation. N Engl J Med. 2018 Dec 13;379(24)2330-2341. doi: 10.1056/NEJMoa1808777. Epub 2018 Oct 31. PMID: 30380364.

  • Duncavage EJ, Jacoby MA, Chang GS, Miller CA, Edwin N, Shao J, Elliott K, Robinson J, Abel H, Fulton RS, Fronick CC, O’Laughlin M, Heath SE, Brendel K, Saba R, Pusic I, Welch JS, Uy G, Link DC, DiPersio JF, Westervelt P, Ley TJ, Trinkaus K, Graubert TA, and Walter MJ.  Mutation Clearance after Transplant for Myelodysplatic Syndrome.  N Engl J Med. 2018 Sep 13;379(11):1028-1041. doi:10.1056/NEJMoa1804714. PMID: 30207916.

  • Wong WH, Bhatt S, Trinkaus K, Pusic I, Elliott K, Mahajan N, Wan F, Switzer GE, Confer DL, DiPersio J, Pulsipher MA, Shah NN, Sees J, Bystry A, Blundell JR, Shaw BE, Druley TE.  Engraftment of rare, pathogenic donor hematopoietic mutations in unrelated hematopoietic stem cell transplantation.  Sci Transl Med. 2020 Jan 15;12(526). pii: eaax6249. doi: 10.1126/scitranslmed.aax6249.  PMID:  31941826.

  • Duncavage EJ, Schroeder MC, O’Laughlin M, Wilson R., MacMillan S, Bohannon A, Kruchowski S, Garza J, Du F, Hughes AEO, Robinson J, Hughes E, Heath SE, Baty JD, Neidich J, Christopher MJ, Jacoby MA, Uy GL, Fulton RS, Miller CA, Payton JE, Link DC, Walter MJ, Westervelt P, DiPersio JF, Ley TJ and Spencer DH.  Genome Sequencing as an Alternative to Cytogenetic Analysis in Myeloid Cancers.  N Engl J Med. 2021 Mar 11;384(10):924-935. Doi: 10.1056/NEJmoa2024534.  PMID: 33704937.

  • Liu R, Gao Q, Foltz SM, Fowles JS, Yao L, Wang JT, Cao S, Sun H, Wendl MC, Sethuraman S, Weerasinghe A,  Weerasinghe A, Rettig MP, Storrs EP, Yoon CJ, Wyczalkowski MA, McMichael JF, Kohnen DR, King J, Goldsmith SR, O’Neal JO, Fulton RS, Fronick CC, Ley TJ, Jaysinghe RG, Fiala MA, Oh ST, DiPersio JF, Vij R, Ding L.  Co-evolution of tumor and immune cells during progression of multiple myeloma.  Nat Commun 2021|

  • Rimando JC, Christopher MJ, Rettig MP, DiPersioJF. Biology of Disease Relapse in Myeloid Disease: Implication for Strategies to Prevent and Treat Disease Relapse After Stem-Cell Transplantation. J Clin Oncol. 2021 Feb 10;39(5):386-396. Doi: 10.1200/JCO.20.01587. Epub 2021 Jan 12. PMID: 33434062.

  • Yao L, Wang JT, Jayasinghe RG, O'Neal J, Tsai CF, Rettig MP, Song Y, Liu R, Zhao Y, Ibrahim OM, Fiala MA, Fortier JM, Chen S, Gehrs L, Rodrigues FM, Wendl MC, Kohnen D, Shinkle A, Cao S, Foltz SM, Zhou DC, Storrs E, Wyczalkowski MA, Mani S, Goldsmith SR, Zhu Y, Hamilton M, Liu T, Chen F, Vij R, Ding L, DiPersio JF. Single-Cell Discovery and Multiomic Characterization of Therapeutic Targets in Multiple Myeloma. Cancer Res 2023 Apr 14;83(8):1214-1233