Try a new search

Format these results:

Searched for:

person:krogsm01

Total Results:

86


Modulating the extracellular TCR-CD3 interaction to identify novel immunotherapy targets against melanoma [Meeting Abstract]

Natarajan, Aswin; Velmurugu, Yogambigai; Zhou, Yuan; Ge, Chenghao; Nadarajah, Vidushan; Felsovalyi, Klara; Cardozo, Timothy J.; Bracken, Clay; Zhu, Cheng; Krogsgaard, Michelle
ISI:000514869700017
ISSN: 1479-5876
CID: 4345052

Mechanisms of primary resistance to PD-1 checkpoint blockade [Meeting Abstract]

Krogsgaard, M; Moogk, D; Li, K; Yuan, Z; Osman, I; Weber, J S; Zhu, C
Although much clinical progress has been made in harnessing the immune system to recognize and target cancer, there is still a significant lack of an understanding of how tumors evade immune recognition and the mechanisms that drive tumor resistance to both T-cell and checkpoint blockade immunotherapy. Our objective is to understand how tumor-mediated signaling through inhibitory receptors, including PD-1, combines to affect the process of T-cell recognition of tumor antigen and activation signaling. This has the goal of understanding the basis of resistance to PD-1 blockade and potentially identifying new molecular targets to enable T-cells to overcome dysfunction mediated by multiple inhibitory receptors. Biomembrane Force Probe (BFP) measurements show that that the activities of TCR-proximal signaling components affect T-cell mechanosensing and sensitivity at the earliest stages of antigen recognition and are influenced by PD-1 and other inhibitory receptors via Shp-1/2 by targeting CD28 and Lck to directly suppress TCR-pMHC-CD8 binding. Phospho-proteomics and flow cytometry-based analysis of patient-derived T-cells from PD-1 responders and nonresponders identified additional mediators, signaling components and pathways associated with PD-1 checkpoint blockade resistance. Targeting these interactions and understanding the basis of resistance to PD-1 blockade would potentially allow identification of novel biomarkers of resistance or new molecular targets to enable T-cells to overcome dysfunction during PD-1 checkpoint blockade
EMBASE:626516759
ISSN: 2326-6074
CID: 3729902

Mechanisms of primary resistance to immune checkpoint inhibitors in Melanoma [Meeting Abstract]

Moogk, Duane; Wang, Lin; Li, Kaitao; Yuan, Zhou; Zhong, Shi; Yu, Zhiya; Liadi, Ivan; Rittase, William; Fang, Victoria; Dougherty, Janna; Perez-Garcia, Arianne; Varadarajan, Navin; Restifo, Nicholas P.; Frey, Alan; Osman, Iman; Weber, Jeff; Zhu, Cheng; Krogsgaard, Michelle
ISI:000455805400022
ISSN: 1479-5876
CID: 3613502

A KDR germline variant is associated with increased risk of melanoma, a pro-angiogenic phenotype and resistance to immunotherapy [Meeting Abstract]

Illa-Bochaca, Irineu; Giles, Keith; Darvishian, Farbod; Moran, Una; Zhong, Judy; Krogsgaard, Michelle; Kirchhoff, Tomas; Osman, Iman
ISI:000455805400024
ISSN: 1479-5876
CID: 3613492

Melanoma patients harbor pre-existing IgG autoantibodies targeting neuronal proteins that associate with differential clinical outcomes following checkpoint blockade [Meeting Abstract]

Hulett, T; Giles, K; Gowen, M; Simpson, D; Tchack, J; Moran, U; Dawood, Z; Pavlick, A; Hu, S; Zhong, H; Krogsgaard, M; Kirchhoff, T; Osman, I
Background Autoantibody landscapes are very specific to the individual, can remain stable for many years, and contain unique features reported in association with cancer, autoimmunity, infection, neurologic conditions, CD8+ T cell behavior, and checkpoint blockade adverse events [1-11]. The goal of this work was to determine whether pre-existing antigenspecific features in melanoma patient autoantibody landscapes would associate with clinical outcomes following checkpoint blockade. Methods Pre-treatment serum samples were collected from 117 melanoma patients prior to checkpoint blockade with anti-CTLA4 (N=60), anti-PD1 (N=38), or both in combination (N=16). All data was collected with approval of the NYU Institutional Review Board at the NYU Perlmutter Cancer Center with informed consent [11]. Serum samples were run on HuProt Human Proteome Microarrays containing >19,000 human proteins by CDI Laboratories. Raw serum IgG signal intensities were processed across staining cohorts via interquartile range normalization. Pre-existing antibody responses were defined as patient-specific IgG signals >3.5 median absolute deviations above cohort median IgG background (modified Z-score). Group statistics were computed (GraphPad Prism), and gene ontology enrichment analysis was performed (Enrichr) [12]. Results Several pre-existing antigen-specific IgG autoantibody targets were observed to have associations with good outcomes (SD/PR) or objective clinical responses (PR/CR) versus patients with progressive disease (POD). While final determination of the most predictive subsets is ongoing, many targets represent genes in an axis surrounding immune signaling pathways, hereditary neurodegenerative disease, and the ubiquitin proteasome pathway (ie, UBQLN1, UBQLN2). An exemplary example was observed in the autoantibody responses shared by >10% of all patients regardless of clinical outcome. Gene ontology enrichment analysis of these shared melanoma-patient autoantibodies versus KEGG 2019 [12] demonstrates this set of proteins is strongly enriched for neurotrophin signaling-associated proteins after multi-sample correction (P=0.004) (Table 1). Several other associations were observed cohort-wide for ontologies with tissuespecific enrichment in the brain, neurons, and neuronal processes. Conclusions In this pilot study, we found strong associations across the cohort for autoantibodies against nerve-growth-inducing neurotrophins and genes like UBQLN1 and UBQLN2 which have strong associations with amyotrophic lateral sclerosis, frontotemporal dementia, Parkinson's, and Alzheimer's - neurodegenerative diseases that are known to have incidences which correlate with melanoma [14-16]; this hints at a potential immunologic connection between the conditions, perhaps related to an antitumor / autoimmune axis involving the targets reported here. (Table Presented)
EMBASE:629890572
ISSN: 2051-1426
CID: 4227402

The myriad targets of a T cell

Natarajan, Aswin; Krogsgaard, Michelle
PMID: 30520863
ISSN: 1546-1696
CID: 3520362

Baseline antibody profiles predict toxicity in melanoma patients treated with immune checkpoint inhibitors

Gowen, Michael F; Giles, Keith M; Simpson, Danny; Tchack, Jeremy; Zhou, Hua; Moran, Una; Dawood, Zarmeena; Pavlick, Anna C; Hu, Shaohui; Wilson, Melissa A; Zhong, Hua; Krogsgaard, Michelle; Kirchhoff, Tomas; Osman, Iman
BACKGROUND:Immune checkpoint inhibitors (anti-CTLA-4, anti-PD-1, or the combination) enhance anti-tumor immune responses, yielding durable clinical benefit in several cancer types, including melanoma. However, a subset of patients experience immune-related adverse events (irAEs), which can be severe and result in treatment termination. To date, no biomarker exists that can predict development of irAEs. METHODS:We hypothesized that pre-treatment antibody profiles identify a subset of patients who possess a sub-clinical autoimmune phenotype that predisposes them to develop severe irAEs following immune system disinhibition. Using a HuProt human proteome array, we profiled baseline antibody levels in sera from melanoma patients treated with anti-CTLA-4, anti-PD-1, or the combination, and used support vector machine models to identify pre-treatment antibody signatures that predict irAE development. RESULTS:We identified distinct pre-treatment serum antibody profiles associated with severe irAEs for each therapy group. Support vector machine classifier models identified antibody signatures that could effectively discriminate between toxicity groups with > 90% accuracy, sensitivity, and specificity. Pathway analyses revealed significant enrichment of antibody targets associated with immunity/autoimmunity, including TNFα signaling, toll-like receptor signaling and microRNA biogenesis. CONCLUSIONS:Our results provide the first evidence supporting a predisposition to develop severe irAEs upon immune system disinhibition, which requires further independent validation in a clinical trial setting.
PMCID:5880088
PMID: 29606147
ISSN: 1479-5876
CID: 3025242

T cell receptor signal transduction: affinity, force and conformational change [Review]

Moogk, Duane; Natarajan, Aswin; Krogsgaard, Michelle
T cell recognition of antigen and resulting proximal signaling are key steps in the initiation of the adaptive immune response. The T cell receptor interaction with antigen drives signal initiation in an affinity-dependent manner, but many aspects of this process remain incompletely understood, including what regions are responsible for structural changes in the TCR upon antigen binding, the importance of extracellular T cell receptor interactions with CD3, how structural changes are integrated with signaling components, and the role of force in signal transduction. Advances in structural modeling of the TCR-CD3 complex and the ability to quantify the affinity and biophysical nature of these molecular interactions have significantly furthered our understanding of the mechanism of transduction of T cell antigen recognition into intracellular signaling. This knowledge is paramount to understanding how T cell perform their critical role in adaptive immune responses, and for the development and improvement of immunotherapies.
ISI:000432589700008
ISSN: 2211-3398
CID: 3140482

Predictive biomarkers of check point inhibition toxicity in metastatic melanoma [Meeting Abstract]

Gowen, M; Tchack, J; Zhou, H; Giles, K; Paschke, S; Moran, U; Fenyo, D; Tsirigos, A; Pacold, M; Pavlick, A; Krogsgaard, M; Osman, I
Background: There are no predictive biomarkers of ipilimumab (IPI) toxicity. Of metastatic melanoma (MM) patients (pts) receiving IPI (3 mg/kg), 35% require systemic therapies to treat immune-related adverse events (irAEs) and 20% must terminate treatment [1]. Here we tested the hypothesis that a pre-existing autoantibody (autoAb) profile is predictive of IPI irAEs.
Method(s): We measured autoAb levels in pre- and post-treatment sera from MM pts who received IPI (3 mg/kg) monotherapy on a proteome microarray containing ~ 20,000 unique full-length human proteins (HuProt array, CDI Laboratories). Clinical data were prospectively collected with protocol-driven follow-up. IrAEs were categorized by CTCAE guidelines as none (grade 0), mild (grade 12), or severe (grade 34). AutoAb levels were standardized using median quantile normalization and considered positive hits if > 2-SD above the peak array signal and differed by >= 2-fold with p < 0.05 between toxicity groups (Non-parametric Analysis/Wilcox test).
Result(s): Seventy-eight sera from 37 MM pts were analyzed. Antibodies against CTLA-4 were significantly elevated post IPI treatment (p < 0.0001), validating the assay. The pre-treatment levels of 190 IgG autoAbs were significantly different in pts who experienced irAEs (n = 28) compared to those with no irAEs (n = 9). Comparison of severe irAE (n = 9) and no irAE (n = 9) groups revealed 129 IgG auto- Abs that significantly differed in pre-treatment sera. Localization and pathway analysis (UniProt, KEGG, Reactome) showed 81/190 (43%) of the autoAbs targeted nuclear and mitochondrial antigens and were enriched in metabolic pathways (p = 0.015). AutoAbs associated with irAEs did not correlate with treatment response.
Conclusion(s): AutoAbs to antigens enriched in metabolic pathways prior to treatment may predict IPI-induced toxicity in MM. The subcellular localization of targeted antigens could explain the autoimmune toxicities associated with IPI. Studies in larger cohorts and in pts receiving other checkpoint inhibitors and/or combination therapies are essential to determine the validity of the data. If validated, our results would support the discovery of the first toxicity predictor in cancer immunotherapy
EMBASE:627350799
ISSN: 1479-5876
CID: 3831892

Mechanisms of primary resistance to cancer immunotherapies [Meeting Abstract]

Moogk, D; Li, K; Yuan, Z; Zhong, S; Yu, Z; Liadi, I; Rittase, W; Fang, V; Dougherty, J; Perez-Garcia, A; Osman, I; Varadarajan, N; Restifo, N P; Frey, A; Zhu, C; Krogsgaard, M
Background: Although much clinical progress has been made in harnessing the immune system to recognize and target cancer, there is still a significant lack of an understanding of how tumors evade immune recognition and the mechanisms that drive tumor resistance to both T cell and checkpoint blockade immunotherapy. Our objective is to understand how tumor-mediated signaling through inhibitory receptors, including PD-1, combine to affect the process of T cell recognition of tumor antigen and activation signaling, with the goal of understanding the basis of resistance to PD-1 blockade and the potential identification of new molecular targets to enable T cells to overcome dysfunction mediated by multiple inhibitory receptors.
Methods and Results: We show that Lck activity affects T cell sensitivity and influences the probability of inducing effector function [1]. Under non-activating conditions, Lck and Shp-1 phosphorylation and activity vary based on CD8+ memory T cell phenotype. Shp-1 interaction with Lck under non-activation conditions can also vary, as suggested by our results showing decreased Shp-1 S591 phosphorylation, which affects Shp-1 localization and correlates with increased Shp-1 colocalization with Lck. Further, we showed that Shp-1 directly influences Lck activity under non-activating conditions, as inhibition of Shp-1 leads to increased Lck activity. Importantly, inhibition of Shp- 1/2, a major mediator of PD-1 signaling, targeting CD28 and Lck [2], prior to activation leads to increased T cell cytotoxic effector function. Our proteomics-based analysis of patient T cells identified both mediators of PD-1 signaling and signaling components and pathways associated with blockade resistance. It has generally been thought that TCR and CD8 binding depend mainly on their ectodomain interactions with pMHC. We have shown, however, that Lck-CD8 binding [3] and Lck activity [4] are required for upregulated CD8 binding to prebound TCR-pMHC complex. Therefore, the cytoplasmic associations of Lck with CD8 and Zap-70, as well as CD3 with Zap-70 may influence formation and stability of the TCRpMHCCD8 complex. To determine the mechanistic basis of PD-1 inhibition of TCR-pMHCCD8 binding we utilized 2D affinity combined with Biomembrane Force Probe (BFP) measurements[5, 6] and showed that PD-1 directly suppresses TCR pMHCCD8 binding. Our data also revealed that TCR-pMHC binding was independent of PD-1-PD-L1, but TCRpMHCCD8 binding was suppressed by PD-1-PD-L1 binding demonstrating negative cooperativity, as fewer bonds formed than the sum of bonds formed by each interaction alone.
Conclusion(s): Together, our results show that the activities of TCRproximal signaling components affect T cell mechanosensing and sensitivity at the earliest stages of antigen recognition and are influenced by PD-1. Targeting these interactions may enhance tumor-specific T cell sensitivity for cancer immunotherapy and understanding the basis of resistance to PD-1 blockade to potentially allow identification of new molecular targets to enable T cells to overcome dysfunction mediated by multiple inhibitory receptors
EMBASE:627349888
ISSN: 1479-5876
CID: 3831912