Next Article in Journal
The Effect of Landscape Environmental Factors on Gene Flow of Red Deer (Cervus canadensis xanthopygus) in the Southern of the Greater Khingan Mountains, China
Previous Article in Journal
Expression of a Siglec-Fc Protein and Its Characterization
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Communication

Electrostatic Complementarities of Glioblastoma-Resident T-Cell Receptors and Cancer Testis Antigens Linked to Poor Outcomes and High Levels of Sphingosine Kinase-2 Expression

1
Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
2
College of Medicine, University of Central Florida, Orlando, FL 32827, USA
3
Department of Pediatrics, Oregon Health and Science University Hospital, Portland, OR 97239, USA
4
Internal Medicine, Oregon Health and Science University Hospital, Portland, OR 97239, USA
5
Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
*
Author to whom correspondence should be addressed.
Biology 2023, 12(4), 575; https://doi.org/10.3390/biology12040575
Submission received: 10 February 2023 / Revised: 4 April 2023 / Accepted: 5 April 2023 / Published: 10 April 2023
(This article belongs to the Section Immunology)

Abstract

:

Simple Summary

The chemical complementarity of glioblastoma, tumor-resident T-cell receptors and cancer testis antigens were associated with a worse outcome. Additionally, the high expression of immune marker and low expression of apoptosis genes were associated with a high T-cell receptor–cancer testis antigen chemical complementarity and a worse outcome. In sum, T-cell receptor recombination reads from exome files have the potential to aid in glioblastoma prognoses and may provide opportunities to detect unproductive immune responses.

Abstract

Introduction. Glioblastoma (GBM) is the most aggressive primary brain tumor in adults. Despite a growing understanding of glioblastoma pathology, the prognosis remains poor. Methods. In this study, we used a previously extensively benchmarked algorithm to retrieve immune receptor (IR) recombination reads from GBM exome files available from the cancer genome atlas. The T-cell receptor complementarity determining region-3 (CDR3) amino acid sequences that represent the IR recombination reads were assessed and used for the generation of chemical complementarity scores (CSs) that represent potential binding interactions with cancer testis antigens (CTAs), which is an approach particularly suited to a big data setting. Results. The electrostatic CSs representing the TRA and TRB CDR3s and the CTAs, SPAG9, GAGE12E, and GAGE12F, indicated that an increased electrostatic CS was associated with worse disease-free survival (DFS). We also assessed the RNA expression of immune marker genes, which indicated that a high-level expression of SPHK2 and CIITA genes also correlated with high CSs and worse DFS. Furthermore, apoptosis-related gene expression was revealed to be lower when the TCR CDR3-CTA electrostatic CSs were high. Conclusion. Adaptive IR recombination reads from exome files have the potential to aid in GBM prognoses and may provide opportunities to detect unproductive immune responses.

1. Introduction

Glioblastoma (GBM) remains incurable and has benefited little from new medical treatments since the current treatment approach was established in 2002 [1], although there have been some surgical innovations since that time. Because of the very limited success of current approaches, more recently emphasis has been placed on establishing GBM subtypes with the expectation of moving more in the direction of personalized medical treatments. In particular, the four subtypes of proneuronal, neural, classical, and mesenchymal [2] GBM have gained widespread acceptance with regard to consistent mutation profiles. Meanwhile, immune checkpoint blockade (ICB) has been a disappointment since the first attempts in 2013, with several exceptions related to subdividing GBM categories and with an eye towards one exemplary case of a mutator phenotype that results in possibly complete but, at least, very long-term remissions [3].
Several reasons have been proposed for the lack of improvement using ICB. Glioblastomas contain relatively few T-cells and an abundance of tumor-associated macrophages (TAMs), which enhance tumor development and promote treatment resistance. In contrast, other aggressive tumors have high T-cell infiltration and tend to be more responsive to ICB. Another explanation for GBM’s lack of responsiveness to ICB could be due to chemotherapy and corticosteroids, which are commonly used to manage GBM symptoms. For example, the immunosuppression caused by chemotherapy could negatively impact the effectiveness of ICB. Additionally, corticosteroids are commonly prescribed to manage cerebral edema and are anti-inflammatory, which would likely decrease lymphocyte tumor infiltration. GBM tumors can also adapt to ICB by upregulating different checkpoint protein ligands [4].
The above considerations indicate a need for additional immunological parameters for personalized treatments, including ICB treatments and possibly other immunotherapies or even anti-inflammatory therapies [5]. Specifically, for GBM, cancer testis antigens (CTAs) have come to light as possible targets for immunotherapy [6], with one very relevant indication that such CTAs can be specifically detected in GBM patients and not in healthy controls [7]. CTAs are generally thought to be commonly expressed in many cancers due to increased levels of gene demethylation in cancer cells, either indirectly, due to the inability to reassemble heterochromatic regions representing the CTA genes because of rapid cell division, or due to other, as yet not fully appreciated stem cell-like features of cancer cells that specifically reduce heterochromatin in regions of CTA genes. Thus, in this report, we have applied a recently developed algorithm for assessing the chemical complementarity between immune receptors and CTAs [8] to assess the relationship between such complementarity and DFS and GBM-related gene expression, with results revealing a novel connection between sphingosine kinase-2 (SPHK2) expression and an apparently failed immune response.

2. Methods

2.1. Recovery of the TCGA-GBM (phs000178) IR Recombination Reads

The recovery of the TRA and TRB recombination reads from the GBM exome (WXS) files was per NIH dbGaP approved protocol number 6300, and the WXS file mining for these recombination reads has been extensively described [9,10,11]. The software for recombination read recovery, including readme files, is available at https://github.com/bchobrut-USF/blanck_group and a container version is available at https://hub.docker.com/r/bchobrut/vdj. An updated version of the software, with some added technical conveniences, is available at https://github.com/kcios/2021. The data representing the entire collection of TRA and TRB recombination reads extracted from the TCGA-GBM WXS files are available in supporting online material (SOM) Table S1. The nucleotide sequences themselves are not available due to controlled-access restrictions; however, these reads will be made available by the corresponding author to investigators with proper dbGaP controlled-access approvals.

2.2. Construction and Use of the Adaptive Match Web Tool

The chemical complementarity scoring is based on [8] and was facilitated by the construction of an original web tool, adaptivematch.com [5,12,13], which is publicly accessible and has been extensively benchmarked in [13]. The electrostatic complementarity scores (CSs) are generated by aligning the adaptive IR CDR3 AAs with a candidate antigen peptide sequence. The scoring process is described extensively in [8], including in the SOM of ref. [8], which includes algorithmic details and an mp4 video representing a user-friendly description. Basically, a positive charge in one entity directly across from a negative charge in the other entity would increase the score significantly, whereas proximity without direct alignment would represent a reduced contribution to a complementarity score increase. The adaptivematch.com web tool includes instructions for preparation of input files. Additionally, three example input files and two example output files are provided in the SOM as Tables S2–S6, representing CS calculations with the CDR3s traceable to the TCGA-GBM WXS files.

2.3. Gene Expression Analysis

The TCGA-GBM gene expression assessments were performed using RNAseq values available via cbioportal.org. The RNAseq values were plotted against the electrostatic CSs to determine Pearson’s correlation coefficients and p-values. Again, this process is facilitated by adaptivematch.com, with the uploading of the csv files as detailed in the web tool instructions for preparation of input files containing RNAseq values and case IDs. For this study, the RNAseq values represented the Firehose legacy version of the TCGA-GBM dataset.

2.4. Survival Analyses

The Kaplan–Meier analyses were verified using the cbioportal.org web tool and GraphPad Prism as described [14,15,16].

2.5. Analysis of the Clinical Proteomic Tumor Analysis Consortium (CPTAC, phs001287) Dataset

To assess the correlation of CIITA RNAseq values with the CSs for TCR CDR3-SPAG9 Fragment 6, first, the adaptive IR recombination reads were obtained from the GBM subset of the CPTAC-3 study, by downloading the RNAseq files representing that dataset to USF research computing, via dbGaP approved protocol number 31752, and processing the RNAseq files with the software described and provided above, via the github links. Then, the CIITA RNAseq values were obtained by merging the files for each case ID for open access, “STAR counts”, available via the genomic data commons web tool.

3. Results

To determine whether the electrostatic CSs (Methods) for GBM tumor-resident TCR CDR3s and CTAs could represent survival distinctions, we tested a set of CTAs, with results indicating that the upper 50th percentile of the TCR CDR3-CTA CSs for SPAG9, GAGE12F, and GAGE12G represented a worse DFS probability (Figure 1; Table S7). To further investigate these results, we assessed the RNA expression of a panel of immune marker genes for the correlation of their expression levels with the electrostatic CSs, with results indicating that CIITA and SPHK2 expression correlated with the TCR CDR3-CTA CSs (Figure 2), while expression of the B-cell marker gene CD19 inversely correlated with the CSs (Table 1).
We next considered the possibility that GBM tumor samples representing higher TCR CDR3-CTA CSs and worse DFS would also have a lower level of expression of apoptosis-related genes. We thus tested a panel of 22 apoptosis-effector genes and, most strikingly, the results indicated that AIFM3, which has a highly brain-specific expression pattern (http://genome.ucsc.edu/cgi-bin/hgGene?hgg_gene=ENST00000399167.6&hgg_chrom=chr22&hgg_start=20965107&hgg_end=20981360&hgg_type=knownGene&db=hg38), inversely correlated with the high TCR CDR3-CTA CSs (Figure 3, Table 2).
We next divided the SPAG9 AA sequences into approximately 18 equal segments and tested each segment for a correlation of the TCR CDR3-SPAG9 fragment CSs with survival, with the results indicating that the SPAG9 Fragment 6 (KHIEVQVAQETRNVSTGSAENEEKSEVQAIIESTPELDMDKDLSGYKGSSTPTKGIENKA) in particular reproduced the strong inverse association of the TCR CDR3-CTA CSs with DFS and reproduced the same immune marker and apoptosis marker correlations, or inverse correlations (Figure 4; Table 3 and Table 4).
We next considered the possibility that the immune gene expression markers could represent survival distinctions for the entire GBM dataset rather than only for the samples with TRA and TRB recombination read recoveries. We focused on CIITA, in the immune marker case, because CIITA represented the most statistically significant correlations with the TCR CDR3-CTA CSs. The results indicated that upper and lower 50th percentiles for CIITA RNAseq values indicated a DFS distinction, with worse DFS probability associated with high CIITA RNAseq values (Figure 5).
Finally, we sought to replicate the correlation of the TCR CDR3-SPAG9 Fragment 6 CS with the CIITA gene expression using the CPTAC GBM dataset. In this case, the CSs were based on TCR recombination reads obtained from GBM RNAseq files, rather than WXS files, which were the source of the TCR recombination reads for the TCGA-GBM dataset analyses above. The results indicated that the CPTAC GBM TCR CDR3-SPAG9 Fragment 6 CSs did indeed correlate with the expression of CIITA (R value = 0.388, p-value = 0.0002). As indicated in Methods, this assessment was facilitated by the use of the adaptivematch.com web tool. The input files for the web tool for this CPTAC-related analysis are in the SOM as Tables S11–S13. The output data are available in Table S14.

4. Discussion

The data above indicated that a higher electrostatic CS for TCR-CDR3-GBM CTAs and higher CIITA expression negatively correlated with GBM DFS probabilities, which emphasizes the need to more fully understand the potential negative impacts of the immune system on outcomes. While it is clear that an anti-tumor immune response in certain settings will facilitate tumor eradication, it is equally clear that certain inflammatory settings predispose to cancer growth. AIFM3, which is expressed almost exclusively in the brain, was expressed at a higher level in tumors where the TCR CDR3-CTA CSs were low, which is consistent with the idea that a high TCR CDR3-CTA CS is not representative of tumor cell killing by T-cells, i.e., lower DFS probabilities were consistent with the highly likely lack of apoptosis in the tumor samples. Thus, the important question becomes, is the inflammatory setting indicated by high TCR CDR3-CTA CSs supporting rather than reducing tumor growth?
The above results have two important limitations. First, there is no other reported approach, other than the in silico approach reported here, that supports the in vivo interaction of a TCR CDR3 with the CTAs that were identified with this in silico immunoinformatics approach. Second, there was no identification of the source of CIITA, which was identified as a pan-GBM dataset biomarker for a worse DFS probability. However, while additional work is needed to support the in silico indications of TCR and CTA binding, the algorithm used here will likely have use for patient risk stratification. As for CIITA, generally, when CIITA is expressed by tumor cells, which is very common in melanoma, it has a strongly anti-apoptotic function [17], consistent with the worse outcome reported here. However, CIITA, of course, could also be expressed by microenvironment antigen-presenting cells, consistent with a strong TCR CDR3–CTA interaction.
The data above also indicated that a higher level of SPHK2 was associated with the higher TCR CDR3-CTA CSs. Sphingosine 1-phosphate (S1P) plays a role in various cellular processes, including cell survival, proliferation, and apoptosis. Both mammalian sphingosine kinases, SPHK2 and sphingosine kinase 1 (SPHK1), generate S1P from a sphingosine precursor [18]. Both SPHK2 and SPHK1 have the capacity to facilitate tumorigenesis. However, SPHK2 has a more complex role, which is dependent on its subcellular localization. When SPHK2 is translocated to the plasma membrane, pro-proliferative signaling occurs, while translocation to internal organelles enhances anti-proliferative functions [18]. The cellular localization of SPHK2 to internal organelles is mediated by cytoplasmic dynein 1. It has been shown that GBM demonstrates lower cellular cytoplasmic dynein 1, and in turn, higher SPHK2 in the plasma membrane. In addition, it should be kept in mind that sphingosine-1-phosphate is a chemoattractant for T-cells [19,20], raising the question of whether SPHK2-generated sphingosine-1-phosphate in turn leads to a high infiltration of T-cells and ultimately to a pro-tumor, inflammatory environment?

5. Conclusions

A significant portion of GBM patients is likely to have only non-functional adaptive immune responses that may be actively supporting tumor growth.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/biology12040575/s1, Table S1: Data representing the entire collection of TRA and TRB recombination reads extracted from the TCGA-GBM WXS files; Table S2: adaptivematch input file, CTAs; Table S3: adaptivematch input file, GBM CDR3s; Table S4: adaptivematch input file, GBM DFS data. Table S5: adaptive match output file, summary (Note, electrostatic indicated as “ncpr” in this output file); Table S6: adaptive match output file, raw electrostatic complementarity scores (Note, electrostatic indicated as “ncpr” in this output file); Table S7: Figure 1 and Figure 4A case IDs used for KM analyses; Table S8: Figure 2 input and output data (SPHK2 and CIITA correlated with CDR3-SPAG9 CSs); Table S9: Figure 3 input and output data; Table S10: Figure 5 case IDs used for KM analyses; Table S11: Adaptivematch (AM) input file, CPTAC TRA, TRB CDR3s; Table S12: Adaptivematch (AM) input file, spag9 fragment 6; Table S13: Adaptivematch (AM) input file, CPTAC CIITA gene expression; Table S14: adaptivematch output file, CIITA RNAseq_CS_correlation, for the CPTAC dataset.

Author Contributions

M.A.A.: Conceptualization: Formal analysis; Methodology; Visualization; Writing—review & editing. K.J.C.: Methodology; Software. D.B.K.: Methodology; Software; Visualization. B.E.M.: Conceptualization; Formal analysis; Methodology; Visualization; Writing—review & editing. J.J.S.: Methodology; Software. A.R.P.: Methodology; Software. A.C.: Methodology; Visualization. B.I.C.: Formal analysis; Methodology; Software; Visualization. G.B.: Methodology; Project administration; Resources; Supervision; Writing—review & editing. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

IRB Ethical review and approval were waived for this study due to the de-identified features of the data. However, as noted above, the data access and project approvals were required by the dbGaP. The dbGaP project approval number is 6300.

Informed Consent Statement

Not applicable, as the analysis of these data were non-human subject results, as deemed by dbGaP project approval 6300.

Data Availability Statement

The data presented in this study are available in the supporting online materials or are publicly available at cbioportal.org.

Acknowledgments

The authors thank USF research computing; Corinne Walters for extensive administrative assistance with the database approvals; and the taxpayers of the State of Florida.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

AAAmino acid
BBBBlood–brain barrier
CDR3Complementarity determining region-3
CNSCentral nervous system
CPTACClinical Proteomic Tumor Analysis Consortium
CSComplementarity score
CTACancer testis antigen
dbGaPDatabase of genotypes and phenotypes
DFSDisease-free survival
GBMGlioblastoma
ICBImmune checkpoint blockade
IR(Adaptive) immune receptor
KMKaplan–Meier
NIHNational Institutes of Health
TCRT-cell receptor
TCGAThe cancer genome atlas

References

  1. Wu, W.; Klockow, J.L.; Zhang, M.; Lafortune, F.; Chang, E.; Jin, L.; Wu, Y.; Daldrup-Link, H.E. Glioblastoma multiforme (GBM): An overview of current therapies and mechanisms of resistance. Pharmacol. Res. 2021, 171, 105780. [Google Scholar] [CrossRef] [PubMed]
  2. Davis, M.E. Glioblastoma: Overview of Disease and Treatment. Clin. J. Oncol. Nurs. 2016, 20 (Suppl. S5), S2–S8. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Rong, L.; Li, N.; Zhang, Z. Emerging therapies for glioblastoma: Current state and future directions. J. Exp. Clin. Cancer Res. 2022, 41, 142. [Google Scholar] [CrossRef] [PubMed]
  4. Yu, M.W.; Quail, D.F. Immunotherapy for Glioblastoma: Current Progress and Challenges. Front. Immunol. 2021, 12, 1637. [Google Scholar] [CrossRef] [PubMed]
  5. Patel, A.R.; Patel, D.N.; Tu, Y.N.; Yeagley, M.; Chobrutskiy, A.; Chobrutskiy, B.I.; Blanck, G. Chemical complementarity between immune receptor CDR3s and candidate cancer antigens correlating with reduced survival: Evidence for outcome mitigation with corticosteroid treatments. J. Biomol. Struct. Dyn. 2022, 1–9. [Google Scholar] [CrossRef] [PubMed]
  6. Ma, R.; Rei, M.; Woodhouse, I.; Ferris, K.; Kirschner, S.; Chandran, A.; Gileadi, U.; Chen, J.-L.; Pinho, M.P.; Ariosa-Morejon, Y.; et al. Decitabine increases neoantigen and cancer testis antigen expression to enhance T-cell–mediated toxicity against glioblastoma. Neuro-Oncology 2022, 24, 2093–2106. [Google Scholar] [CrossRef] [PubMed]
  7. Shraibman, B.; Barnea, E.; Kadosh, D.M.; Haimovich, Y.; Slobodin, G.; Rosner, I.; López-Larrea, C.; Hilf, N.; Kuttruff, S.; Song, C.; et al. Identification of Tumor Antigens Among the HLA Peptidomes of Glioblastoma Tumors and Plasma. Mol. Cell. Proteom. 2019, 18, 1255–1268. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  8. Chobrutskiy, B.I.; Chobrutskiy, A.; Zaman, S.; Yeagley, M.; Huda, T.I.; Blanck, G. High-throughput, sliding-window algorithm for assessing chemical complementarity between immune receptor CDR3 domains and cancer mutant peptides: TRG-PIK3CA interactions and breast cancer. Mol. Immunol. 2021, 135, 247–253. [Google Scholar] [CrossRef] [PubMed]
  9. Gill, T.R.; Samy, M.D.; Butler, S.N.; Mauro, J.A.; Sexton, W.J.; Blanck, G. Detection of Productively Rearranged TcR-α V–J Sequences in TCGA Exome Files: Implications for Tumor Immunoscoring and Recovery of Antitumor T-cells. Cancer Inform. 2016, 15, 23–28. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  10. Tong, W.L.; Tu, Y.N.; Samy, M.D.; Sexton, W.J.; Blanck, G. Identification of immunoglobulin V(D)J recombinations in solid tumor specimen exome files: Evidence for high level B-cell infiltrates in breast cancer. Hum. Vaccines Immunother. 2017, 13, 501–506. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  11. Chobrutskiy, B.I.; Zaman, S.; Tong, W.L.; Diviney, A.; Blanck, G. Recovery of T-cell receptor V(D)J recombination reads from lower grade glioma exome files correlates with reduced survival and advanced cancer grade. J. Neuro-Oncol. 2018, 140, 697–704. [Google Scholar] [CrossRef] [PubMed]
  12. Huda, T.I.; Diaz, M.J.; Gozlan, E.C.; Chobrutskiy, A.; Chobrutskiy, B.I.; Blanck, G. Immunogenomics Parameters for Patient Stratification in Alzheimer’s Disease. J. Alzheimer’s Dis. 2022, 88, 619–629. [Google Scholar] [CrossRef] [PubMed]
  13. Eakins, R.A.; Chobrutskiy, A.; Teer, J.K.; Patel, D.N.; Hsiang, M.; Huda, T.I.; Zaman, S.; Sexton, W.J.; Coppola, D.; Falasiri, S.; et al. Chemical complementarity between tumor resident, T-cell receptor CDR3s and MAGEA3/6 correlates with increased melanoma survival: Potential relevance to MAGE vaccine auto-reactivity. Mol. Immunol. 2022, 150, 58–66. [Google Scholar] [CrossRef] [PubMed]
  14. Yeagley, M.; Chobrutskiy, B.I.; Gozlan, E.C.; Medikonda, N.; Patel, D.N.; Falasiri, S.; Callahan, B.M.; Huda, T.; Blanck, G. Electrostatic Complementarity of T-Cell Receptor-Alpha CDR3 Domains and Mutant Amino Acids Is Associated with Better Survival Rates for Sarcomas. Pediatr. Hematol. Oncol. 2021, 38, 251–264. [Google Scholar] [CrossRef] [PubMed]
  15. Chobrutskiy, A.; Chobrutskiy, B.I.; Zaman, S.; Hsiang, M.; Blanck, G. Chemical features of blood-borne TRG CDR3s associated with an increased overall survival in breast cancer. Breast Cancer Res. Treat. 2020, 185, 591–600. [Google Scholar] [CrossRef] [PubMed]
  16. Hsiang, M.; Chobrutskiy, B.I.; Diaz, M.; Huda, T.I.; Creadore, S.; Zaman, S.; Cios, K.J.; Gozlan, E.C.; Blanck, G. Chemical complementarity between immune receptors and cancer mutants, independent of antigen presentation protein binding, is associated with increased survival rates. Transl. Oncol. 2021, 14, 101069. [Google Scholar] [CrossRef] [PubMed]
  17. Gourley, T.S.; Chang, C.-H. Cutting Edge: The Class II Transactivator Prevents Activation-Induced Cell Death by Inhibiting Fas Ligand Gene Expression. J. Immunol. 2001, 166, 2917–2921. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  18. Neubauer, H.A.; Tea, M.N.; Zebol, J.R.; Gliddon, B.L.; Stefanidis, C.; Moretti, P.A.B.; Pitman, M.R.; Costabile, M.; Kular, J.; Stringer, B.W.; et al. Cytoplasmic dynein regulates the subcellular localization of sphingosine kinase 2 to elicit tumor-suppressive functions in glioblastoma. Oncogene 2019, 38, 1151–1165. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  19. Allende, M.L.; Dreier, J.L.; Mandala, S.; Proia, R.L. Expression of the Sphingosine 1-Phosphate Receptor, S1P1, on T-cells Controls Thymic Emigration. J. Biol. Chem. 2004, 279, 15396–15401. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  20. Kim, C.H. The greater chemotactic network for lymphocyte trafficking: Chemokines and beyond. Curr. Opin. Hematol. 2005, 12, 298–304. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Kaplan–Meier (KM) analyses comparing disease-free survival (DFS) probabilities for case IDs representing the upper and lower 50th percentile TCR CDR3-CTA complementarity scores (CSs). (A) KM analysis of case IDs representing the upper (black line, n = 17) and lower (grey line, n = 18) 50th percentile TRA or TRB CDR3-SPAG9 CS groups. (B) KM analysis of case IDs representing the upper (black line, n = 19) and lower (grey line, n = 21) 50th percentile TRA or TRB CDR3-GAGE12F CS groups. (C) KM analysis of case IDs representing the upper (black line, n = 22) and lower (grey line, n = 22) 50th percentile TRA or TRB CDR3-GAGE12G CS groups.
Figure 1. Kaplan–Meier (KM) analyses comparing disease-free survival (DFS) probabilities for case IDs representing the upper and lower 50th percentile TCR CDR3-CTA complementarity scores (CSs). (A) KM analysis of case IDs representing the upper (black line, n = 17) and lower (grey line, n = 18) 50th percentile TRA or TRB CDR3-SPAG9 CS groups. (B) KM analysis of case IDs representing the upper (black line, n = 19) and lower (grey line, n = 21) 50th percentile TRA or TRB CDR3-GAGE12F CS groups. (C) KM analysis of case IDs representing the upper (black line, n = 22) and lower (grey line, n = 22) 50th percentile TRA or TRB CDR3-GAGE12G CS groups.
Biology 12 00575 g001
Figure 2. Correlation of SPHK2 and CIITA gene expression with TCR CDR3-SPAG9 electrostatic CSs. (See also Table 1; Table S8).
Figure 2. Correlation of SPHK2 and CIITA gene expression with TCR CDR3-SPAG9 electrostatic CSs. (See also Table 1; Table S8).
Biology 12 00575 g002
Figure 3. Inverse correlation of AIFM3 gene expression and electrostatic CSs for TCR CDR3-CTAs, SPAG9, GAGE12G, and GAGE12F. (See also Table 2; Table S9).
Figure 3. Inverse correlation of AIFM3 gene expression and electrostatic CSs for TCR CDR3-CTAs, SPAG9, GAGE12G, and GAGE12F. (See also Table 2; Table S9).
Biology 12 00575 g003
Figure 4. KM analysis comparing DFS for case IDs representing the upper and lower 50th percentile for the TCR CDR3-SPAG9 Fragment 6 electrostatic CSs. (A) The case IDs represent the upper 50th percentile electrostatic CS group (black line, n = 21) and lower 50th percentile electrostatic CS group (grey line, n = 23). (B,C) Scatter plots showing a correlation of the expression of SPHK2 and CIITA genes and electrostatic CSs for the TCR CDR3-SPAG9 Fragment 6. See also Table 3; Table S7.
Figure 4. KM analysis comparing DFS for case IDs representing the upper and lower 50th percentile for the TCR CDR3-SPAG9 Fragment 6 electrostatic CSs. (A) The case IDs represent the upper 50th percentile electrostatic CS group (black line, n = 21) and lower 50th percentile electrostatic CS group (grey line, n = 23). (B,C) Scatter plots showing a correlation of the expression of SPHK2 and CIITA genes and electrostatic CSs for the TCR CDR3-SPAG9 Fragment 6. See also Table 3; Table S7.
Biology 12 00575 g004
Figure 5. KM analysis comparing DFS for case IDs representing the upper (black line) and lower 50th percentile (grey line) CIITA RNAseq values for the entire TCGA-GBM dataset. (logrank p-value = 0.013; Table S10).
Figure 5. KM analysis comparing DFS for case IDs representing the upper (black line) and lower 50th percentile (grey line) CIITA RNAseq values for the entire TCGA-GBM dataset. (logrank p-value = 0.013; Table S10).
Biology 12 00575 g005
Table 1. Immune marker gene expression correlations with GBM TCR CDR3-CTA electrostatic CSs for SPAG9, GAGE12F, and GAGE12G.
Table 1. Immune marker gene expression correlations with GBM TCR CDR3-CTA electrostatic CSs for SPAG9, GAGE12F, and GAGE12G.
(See also Figure 2)
CTAImmune Marker GenePearson’s Coefficientp-Value
GAGE12FCIITA0.4860.0160
GAGE12GCIITA0.4860.0160
SPAG9CD19−0.4320.0348
SPAG9CIITA0.4270.0373
SPAG9SPHK20.4080.0477
GAGE12GSPHK20.4060.0492
GAGE12FSPHK20.4060.0492
GAGE12FCD19−0.3770.0691
GAGE12GCD19−0.3770.0691
Table 2. Apoptosis-effector gene expression, inverse correlations with GBM TCR CDR3 CTA CSs for SPAG9, GAGE12F, and GAGE12G.
Table 2. Apoptosis-effector gene expression, inverse correlations with GBM TCR CDR3 CTA CSs for SPAG9, GAGE12F, and GAGE12G.
CTAApoptosis GenePearson’s Coefficientp-Value
GAGE12FCOX7A2L−0.4380.0324
GAGE12GCOX7A2L−0.4380.0324
SPAG9COX7A2L−0.4260.0378
SPAG9AIFM3−0.4250.0385
SPAG9UQCRC2−0.4080.0480
GAGE12GUQCRC2−0.3950.0561
GAGE12FUQCRC2−0.3950.0561
GAGE12GAIFM3−0.3700.0747
GAGE12FAIFM3−0.3700.0747
Table 3. Immune marker gene expression correlations with GBM TCR CDR3-SPAG9 Fragment 6 electrostatic CSs.
Table 3. Immune marker gene expression correlations with GBM TCR CDR3-SPAG9 Fragment 6 electrostatic CSs.
Immune Marker GenePearson’s Coefficientp-Value
CIITA0.4650.0222
CD19−0.3960.0553
SPHK20.3400.104
Table 4. Apoptosis effector gene expression, inverse correlations with GBM TCR CDR3-SPAG9 Fragment 6 electrostatic CSs.
Table 4. Apoptosis effector gene expression, inverse correlations with GBM TCR CDR3-SPAG9 Fragment 6 electrostatic CSs.
Apoptosis Effector GenePearson’s Coefficientp-Value
COX7A2L−0.4730.0195
AIFM3−0.4510.0268
UQCRC2−0.4290.0363
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Arias, M.A.; Cios, K.J.; Kacsoh, D.B.; Montgomery, B.E.; Song, J.J.; Patel, A.R.; Chobrutskiy, A.; Chobrutskiy, B.I.; Blanck, G. Electrostatic Complementarities of Glioblastoma-Resident T-Cell Receptors and Cancer Testis Antigens Linked to Poor Outcomes and High Levels of Sphingosine Kinase-2 Expression. Biology 2023, 12, 575. https://doi.org/10.3390/biology12040575

AMA Style

Arias MA, Cios KJ, Kacsoh DB, Montgomery BE, Song JJ, Patel AR, Chobrutskiy A, Chobrutskiy BI, Blanck G. Electrostatic Complementarities of Glioblastoma-Resident T-Cell Receptors and Cancer Testis Antigens Linked to Poor Outcomes and High Levels of Sphingosine Kinase-2 Expression. Biology. 2023; 12(4):575. https://doi.org/10.3390/biology12040575

Chicago/Turabian Style

Arias, Miguel A., Konrad J. Cios, Dorottya B. Kacsoh, Bailey E. Montgomery, Joanna J. Song, Anishaa R. Patel, Andrea Chobrutskiy, Boris I. Chobrutskiy, and George Blanck. 2023. "Electrostatic Complementarities of Glioblastoma-Resident T-Cell Receptors and Cancer Testis Antigens Linked to Poor Outcomes and High Levels of Sphingosine Kinase-2 Expression" Biology 12, no. 4: 575. https://doi.org/10.3390/biology12040575

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop