Try a new search

Format these results:

Searched for:

person:yarmam01

in-biosketch:yes

Total Results:

21


Cross-HLA targeting of intracellular oncoproteins with peptide-centric CARs

Yarmarkovich, Mark; Marshall, Quinlen F; Warrington, John M; Premaratne, Rasika; Farrel, Alvin; Groff, David; Li, Wei; di Marco, Moreno; Runbeck, Erin; Truong, Hau; Toor, Jugmohit S; Tripathi, Sarvind; Nguyen, Son; Shen, Helena; Noel, Tiffany; Church, Nicole L; Weiner, Amber; Kendsersky, Nathan; Martinez, Dan; Weisberg, Rebecca; Christie, Molly; Eisenlohr, Laurence; Bosse, Kristopher R; Dimitrov, Dimiter S; Stevanovic, Stefan; Sgourakis, Nikolaos G; Kiefel, Ben R; Maris, John M
The majority of oncogenic drivers are intracellular proteins, thus constraining their immunotherapeutic targeting to mutated peptides (neoantigens) presented by individual human leukocyte antigen (HLA) allotypes1. However, most cancers have a modest mutational burden that is insufficient to generate responses using neoantigen-based therapies2,3. Neuroblastoma is a paediatric cancer that harbours few mutations and is instead driven by epigenetically deregulated transcriptional networks4. Here we show that the neuroblastoma immunopeptidome is enriched with peptides derived from proteins that are essential for tumourigenesis and focus on targeting the unmutated peptide QYNPIRTTF, discovered on HLA-A*24:02, which is derived from the neuroblastoma dependency gene and master transcriptional regulator PHOX2B. To target QYNPIRTTF, we developed peptide-centric chimeric antigen receptors (CARs) using a counter-panning strategy with predicted potentially cross-reactive peptides. We further hypothesized that peptide-centric CARs could recognize peptides on additional HLA allotypes when presented in a similar manner. Informed by computational modelling, we showed that PHOX2B peptide-centric CARs also recognize QYNPIRTTF presented by HLA-A*23:01 and the highly divergent HLA-B*14:02. Finally, we demonstrated potent and specific killing of neuroblastoma cells expressing these HLAs in vitro and complete tumour regression in mice. These data suggest that peptide-centric CARs have the potential to vastly expand the pool of immunotherapeutic targets to include non-immunogenic intracellular oncoproteins and widen the population of patients who would benefit from such therapy by breaking conventional HLA restriction.
PMID: 34732890
ISSN: 1476-4687
CID: 5324062

Targeting of intracellular oncoproteins with peptide-centric CARs

Yarmarkovich, Mark; Marshall, Quinlen F; Warrington, John M; Premaratne, Rasika; Farrel, Alvin; Groff, David; Li, Wei; di Marco, Moreno; Runbeck, Erin; Truong, Hau; Toor, Jugmohit S; Tripathi, Sarvind; Nguyen, Son; Shen, Helena; Noel, Tiffany; Church, Nicole L; Weiner, Amber; Kendsersky, Nathan; Martinez, Dan; Weisberg, Rebecca; Christie, Molly; Eisenlohr, Laurence; Bosse, Kristopher R; Dimitrov, Dimiter S; Stevanovic, Stefan; Sgourakis, Nikolaos G; Kiefel, Ben R; Maris, John M
The majority of oncogenic drivers are intracellular proteins, constraining their immunotherapeutic targeting to mutated peptides (neoantigens) presented by individual human leukocyte antigen (HLA) allotypes1. However, most cancers have a modest mutational burden that is insufficient for generating responses using neoantigen-based therapies2,3. Neuroblastoma is a paediatric cancer that harbours few mutations and is instead driven by epigenetically deregulated transcriptional networks4. Here we show that the neuroblastoma immunopeptidome is enriched with peptides derived from proteins essential for tumorigenesis. We focused on targeting the unmutated peptide QYNPIRTTF discovered on HLA-A*24:02, which is derived from the neuroblastoma-dependency gene and master transcriptional regulator PHOX2B. To target QYNPIRTTF, we developed peptide-centric chimeric antigen receptors (PC-CARs) through a counter panning strategy using predicted potentially cross-reactive peptides. We further proposed that PC-CARs can recognize peptides on additional HLA allotypes when presenting a similar overall molecular surface. Informed by our computational modelling results, we show that PHOX2B PC-CARs also recognize QYNPIRTTF presented by HLA-A*23:01, the most common non-A2 allele in people with African ancestry. Finally, we demonstrate potent and specific killing of neuroblastoma cells expressing these HLAs in vitro and complete tumour regression in mice. These data suggest that PC-CARs have the potential to expand the pool of immunotherapeutic targets to include non-immunogenic intracellular oncoproteins and allow targeting through additional HLA allotypes in a clinical setting.
PMCID:10665195
PMID: 37938771
ISSN: 1476-4687
CID: 5607932

Discovery and CAR T targeting of lineage-restricted neuroblastoma oncoproteins. [Meeting Abstract]

Yarmarkovich, Mark; Warrington, John M.; Marshall, Quinlen F.; Shen, Helena; Li, Wei; Beasley, Matt; Di Marco, Moreno; Stevanovic, Stefan; Sgourakis, Nikolaos G.; Dimitrov, Dimiter; Smith, Peter; Maris, John M.
ISI:000680263502139
ISSN: 0008-5472
CID: 5324142

Identification of SARS-CoV-2 Vaccine Epitopes Predicted to Induce Long-Term Population-Scale Immunity

Yarmarkovich, Mark; Warrington, John M; Farrel, Alvin; Maris, John M
Here we propose a SARS-CoV-2 vaccine design concept based on identification of highly conserved regions of the viral genome and newly acquired adaptations, both predicted to generate epitopes presented on major histocompatibility complex (MHC) class I and II across the vast majority of the population. We further prioritize genomic regions that generate highly dissimilar peptides from the human proteome and are also predicted to produce B cell epitopes. We propose sixty-five 33-mer peptide sequences, a subset of which can be tested using DNA or mRNA delivery strategies. These include peptides that are contained within evolutionarily divergent regions of the spike protein reported to increase infectivity through increased binding to the ACE2 receptor and within a newly evolved furin cleavage site thought to increase membrane fusion. Validation and implementation of this vaccine concept could specifically target specific vulnerabilities of SARS-CoV-2 and should engage a robust adaptive immune response in the vast majority of the population.
PMID: 32835302
ISSN: 2666-3791
CID: 5324052

High throughput pMHC-I tetramer library production using chaperone-mediated peptide exchange

Overall, Sarah A; Toor, Jugmohit S; Hao, Stephanie; Yarmarkovich, Mark; Morozov, Giora I; Nguyen, Son; Japp, Alberto Sada; Gonzalez, Nicolas; Moschidi, Danai; Betts, Michael R; Maris, John M; Smibert, Peter; Sgourakis, Nikolaos G
Peptide exchange technologies are essential for the generation of pMHC-multimer libraries used to probe diverse, polyclonal TCR repertoires in various settings. Here, using the molecular chaperone TAPBPR, we develop a robust method for the capture of stable, empty MHC-I molecules comprising murine H2 and human HLA alleles, which can be readily tetramerized and loaded with peptides of choice in a high-throughput manner. Alternatively, catalytic amounts of TAPBPR can be used to exchange placeholder peptides with high affinity peptides of interest. Using the same system, we describe high throughput assays to validate binding of multiple candidate peptides on empty MHC-I/TAPBPR complexes. Combined with tetramer-barcoding via a multi-modal cellular indexing technology, ECCITE-seq, our approach allows a combined analysis of TCR repertoires and other T cell transcription profiles together with their cognate antigen specificities in a single experiment. The new approach allows TCR/pMHC interactions to be interrogated easily at large scale.
PMCID:7170893
PMID: 32312993
ISSN: 2041-1723
CID: 5324022

A SARS-CoV-2 Vaccination Strategy Focused on Population-Scale Immunity

Yarmarkovich, Mark; Warrington, John M; Farrel, Alvin; Maris, John M
Here we propose a vaccination strategy for SARS-CoV-2 based on identification of both highly conserved regions of the virus and newly acquired adaptations that are presented by MHC class I and II across the vast majority of the population, are highly dissimilar from the human proteome, and are predicted B cell epitopes. We present 65 peptide sequences that we expect to result in a safe and effective vaccine which can be rapidly tested in DNA, mRNA, or synthetic peptide constructs. These include epitopes that are contained within evolutionarily divergent regions of the spike protein reported to increase infectivity through increased binding to the ACE2 receptor, and within a novel furin cleavage site thought to increase membrane fusion. This vaccination strategy specifically targets unique vulnerabilities of SARS-CoV-2 and should engage a robust adaptive immune response in the vast majority of the human population. Funding: This work was supported by a St. Baldrick's-Stand Up To Cancer Pediatric Dream Team Translational Research Grant (SU2C-AACR-DT2727) and the Beau Biden Cancer Moonshot Pediatric Immunotherapy Discovery and Development Networ (NCI Grant U54 CA232568). Stand Up To Cancer is a program of the Entertainment Industry Foundation administered by the American Association for Cancer Research. This work was also supported by NIH R35 CA220500 and the Giulio D'Angio Endowed Chair and the Quod Erat Demonstrandum (QED) program at the Science Center in Philadelphia.
PMCID:7366814
PMID: 32714112
ISSN: 1556-5068
CID: 5324042

A SARS-CoV-2 Vaccination Strategy Focused on Population-Scale Immunity

Yarmarkovich, Mark; Warrington, John M; Farrel, Alvin; Maris, John M
Here we propose a vaccination strategy for SARS-CoV-2 based on identification of both highly conserved regions of the virus and newly acquired adaptations that are presented by MHC class I and II across the vast majority of the population, are highly dissimilar from the human proteome, and are predicted B cell epitopes. We present 65 peptide sequences that we expect to result in a safe and effective vaccine which can be rapidly tested in DNA, mRNA, or synthetic peptide constructs. These include epitopes that are contained within evolutionarily divergent regions of the spike protein reported to increase infectivity through increased binding to the ACE2 receptor, and within a novel furin cleavage site thought to increase membrane fusion. This vaccination strategy specifically targets unique vulnerabilities of SARS-CoV-2 and should engage a robust adaptive immune response in the vast majority of the human population.
PMID: 32511347
ISSN: 2692-8205
CID: 5324032

SARS-CoV-2 multiepitope vaccine constructs designed to drive long-term immunity in the majority of the population. [Meeting Abstract]

Yarmarkovich, Mark; Warrington, John M.; Farrel, Alvin; Maris, John M.
ISI:000572825800052
ISSN: 1078-0432
CID: 5324132

Immunogenicity and Immune Silence in Human Cancer

Yarmarkovich, Mark; Farrel, Alvin; Sison, Artemio; di Marco, Moreno; Raman, Pichai; Parris, Joshua L; Monos, Dimitrios; Lee, Hongzhe; Stevanovic, Stefan; Maris, John M
Despite recent advances in cancer immunotherapy, the process of immunoediting early in tumorigenesis remains obscure. Here, we employ a mathematical model that utilizes the Cancer Genome Atlas (TCGA) data to elucidate the contribution of individual mutations and HLA alleles to the immunoediting process. We find that common cancer mutations including BRAF-V600E and KRAS-G12D are predicted to bind none of the common HLA alleles, and are thus "immunogenically silent" in the human population. We identify regions of proteins that are not presented by HLA at a population scale, coinciding with frequently mutated hotspots in cancer, and other protein regions broadly presented across the population in which few mutations occur. We also find that 9/29 common HLA alleles contribute disproportionately to the immunoediting of early oncogenic mutations. These data provide insights into immune evasion of common driver mutations and a molecular basis for the association of particular HLA genotypes with cancer susceptibility.
PMCID:7092187
PMID: 32256484
ISSN: 1664-3224
CID: 5324012

When Cold Is Hot: Immune Checkpoint Inhibition Therapy for Rhabdoid Tumors

Yarmarkovich, Mark; Maris, John M
Carcinogen-induced cancers typically have high mutation burdens and an inflamed microenvironment and thus are poised to respond to immune checkpoint inhibitors (ICIs). However, cancers with loss-of-function mutations in the SWI/SNF complex have few additional mutations yet also have an inflamed immunophenotype and should respond to ICI therapy.
PMID: 31951557
ISSN: 1878-3686
CID: 5324002