Abstract
Steven A. Greenberg1
1 Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
Correspondence to:
Steven A. Greenberg, | email: | [email protected] |
Keywords: immunotherapy; KLRG1
Received: October 11, 2024 Accepted: December 30, 2024 Published: January 20, 2025
ABSTRACT
Recently, combination checkpoint therapy of cancer has been recognized as producing additive as opposed to synergistic benefit due in part to positively correlated effects. The potential for uncorrelated or negatively correlated therapies to produce true synergistic benefits has been noted. Whereas the inhibitory receptors PD-1, CTLA-4, TIM-3, LAG-3, and TIGIT have been collectively characterized as exhaustion receptors, another inhibitory receptor KLRG1 was historically characterized as a senescent receptor and received relatively little attention as a potential checkpoint inhibitor target. The anti-tumor effects of KLRG1 blockade has relatively recently been demonstrated in preclinical in vivo studies. Here, expression of the inhibitory receptors PD-1, CTLA-4, TIM-3, LAG-3, TIGIT, and KLRG1 was studied in publicly available gene expression datasets. Bulk RNA microarray and RNAseq, and single cell RNAseq data from healthy blood and tumor tissue samples were analyzed for Pearson correlation. CD8 T cell differentiation of memory T cells from the TEM to TEMRA states is characterized by PD-1/KLRG1 anti-correlation, with decreased PD-1 expression but increased KLRG1 expression. Single cell RNAseq analysis of tumor infiltrating CD8 T cells shows positive correlation of CTLA-4, TIM-3, LAG-3, TIGIT, GITR, 4-1BB, and OX40 with PD-1 but negative correlation of KLRG1 with PD-1. The anti-correlation of PD-1 and KLRG1 expression in human tumor infiltrating CD8 T cells suggests the potential for combination therapy supra-additive benefits of anti-PD-1 and anti-KLRG1 therapies.
Introduction
Targeting of the T cell coinhibitory receptors PD-1, CTLA-4, TIM-3, LAG-3, and TIGIT and costimulatory receptors GITR, 4-1BB, and OX40 for applications in oncology has gained widespread interest [1–3] and been tested in clinical studies. Therapeutic benefits in preclinical mouse studies with blocking antibodies was demonstrated for CTLA-4 by 1996 [4], PD-1 by 2005 [5, 6], LAG-3 by 2007 [7], TIM-3 by 2013 [8], and TIGIT by 2014 [9].
In contrast, the inhibitory receptor KLRG1 has received less interest. A benefit for KLRG1 blockade in mouse models was demonstrated in 2019 and again in 2021 [10, 11], with KLRG1 and PD-1 blockade producing supra-additive benefits in the B16F10 mouse melanoma model.
Historically, the pursuit of additional T cell inhibitory receptors has been driven by attention to co-regulated expression. Thus, studies in 2007 of the molecular signature of mouse CD8+ T cells during chronic viral infection identified coordinate regulation of all inhibitory receptors studied (including PD-1, CTLA-4, and LAG-3) except for two, KLRG1 and KLRA9 [12]. Subsequent nearest neighbor correlation studies of this data identified KLRG1 as uniquely anti-correlated with PD-1 in mouse CD8+ T cells during chronic viral infection [13]. The coordinated expression of PD-1 with TIGIT [9] has also been emphasized.
Ultimately, a view emerged that receptors with coordinated expression to each other constituted a class of exhaustion receptors that were promising therapeutic targets while receptors with low or negative correlation to PD-1 might not be attractive targets. This view was further cemented by 2011 and 2015 with nomenclature for cancer CD8 T cells characterizing CTLA-4, PD-1, LAG-3, and TIM-3 as marking “exhausted” cells, whereas KLRG1 marked senescent cells [14, 15]. Alternative views as early as 2012 have noted the arbitrary distinction between exhaustion and senescence applied to these receptors, that human CD8 T cell inhibitory receptor surface protein expression may depend on differentiation and that KLRG1 protein surface expression is much higher in CD8 effector memory and effector populations than PD-1, CTLA-4, TIM-3, and LAG-3 [16, 17].
Here, we point out the distinct anti-correlation of KLRG1 expression with PD-1 and other checkpoint receptors in human CD8 T cell tumor infiltrating lymphocytes (TILS). Combination therapy in cancer seeks to increase efficacy over monotherapy, ideally through synergy that produces more than just additive benefits of two independent treatments. However, combination therapy synergistic efficacy has very uncommonly been seen in cancer immunotherapy combination trials [18]. Previously the potential for combination therapies of anti-correlated combinations to produce clinical benefits greater than the sum of the benefits of the individual therapies has been noted [19]. As this effect has been unrealized in the field of combination inhibitory receptor blocking therapies in the treatment of cancer, the identification of anti-correlated mechanisms may yield synergistic combination therapy benefits beyond those seen to date.
Results
KLRG1 marks highly differentiated cytotoxic T cells that do not express PD-1 in healthy human blood and tumor
Across the highly differentiated T cell population in normal blood, KLRG1 gene expression was anti-correlated (negatively correlated) with PD-1 (r = −0.377) (Figure 1A). Previous authors have noted that KLRG1 protein surface expression by flow cytometry increases substantially in TEM and TEMRA subsets (75–80% of CD8+ T cells), while PD-1 expression decreases from TEM (30%) to TEMRA (10%) subsets [17] (Figure 1B).
Figure 1: Distinct expression pattern of KLRG1 on highly differentiated human T cells.
(A) Anticorrelated gene expression with PD-1 in RNAseq dataset GSE107011 for differentiated T cell subsets CD4 TEMRA, CD8 TEM, CD8 TEMRA, and γδ T cells. (B) Abstracted previously published data [17] for human blood CD8+ T cells protein surface expression showing increased KLRG1 on effector T memory (TEM) and effector (TEMRA) cells contrasting with decreasing PD-1 on TEMRA. (C–E) Characterization of PD-1+ CD8+ tumor infiltrating lymphocytes (TILS) in non-small cell lung cancer (NSCLC) datasets. (C) Abstracted data from previous publication [20] demonstrating that PD-1-high TILS are not highly differentiated TEMRA (CD27+) and (D) that KLRG1+ TILS are PD-1-. (E) Public domain single cell RNAseq (GSE99531) shows coregulated expression of CTLA-4, TIM-3, LAG-3, and TIGIT but anti-regulated KLRG1 expression in PD-1-high TILS.
A similar pattern has been reported [20] in non-small cell lung cancer (NSCLC) CD8+ tumor infiltrating lymphocytes (TILS), where PD-1-high cells are earlier stage CD27+ T cells, while KLRG1+ cells are later-stage CD27– T cells that are not PD-1-high (Figure 1C, 1D). More generally, NSCLC CD8+ PD-1-high vs. PD-1-negative TILS have coregulated expression of CTLA-4, TIM-3, LAG-3, and TIGIT, but anti-regulated expression of KLRG1 (Figure 1E).
DISCUSSION
Much effort in the field of immuno-oncology has involved the study of combination therapies, including combinations involving blockade of more than one T cell inhibitory receptor. Combination therapies may produce clinical benefit through the additive independent action of each therapy or may produce supra-additive efficacy (in excess of that predicted by combined independent action) [18, 21, 22], which has been called clinical synergy [18]. Recent studies have demonstrated that the vast majority of combination studies with anti-PD-1 therapies have produced no more, and sometimes less, than the benefit expected from additive independent action [18, 23, 24].
Thus, supra-additive efficacy remains a desired goal. Although it can result from the well-established concept of pharmacological synergy, other mechanisms can also produce supra-additive efficacy [19]. For example, collateral sensitivity (resistance to one drug confers susceptibility to another) can also produce clinical synergy, with antibiotic combinations used to overcome drug resistance a classic example. More generally, responses to combinations of monotherapies a and b are governed by
where Pab is the probability of response to the combination ab and ρ is the correlation coefficient of responses to a and b [19, 22]. Positively correlated therapies (ρ > 0) can produce less than additive benefit and negatively correlated (anti-correlated) therapies (ρ < 0) can produce supra-additive benefit.
Here, we point out that KLRG1 expression is concentrated on the most differentiated and potent CD8 T cells, unlike PD-1, and its expression is anti-correlated to PD-1 as well as all major coinhibitory and costimulatory checkpoint receptors across tumor infiltrating (TILS) CD8 T cells in a wide variety of cancers. Whereas much of the T cell inhibitory drug development efforts over the last decade have been focused on combinations of expression-correlated inhibitory receptor targets, the targeting of anti-correlated inhibitory receptors has greater potential to produce supra-additive benefit, and KLRG1 has this distinct property.
Materials and Methods
Public domain RNAseq gene expression data of bulk immune cell populations in normal blood (GSE107011) [25] was analyzed for pairwise Pearson correlation of PD-1 (PDCD1) with other T cell inhibitory receptors across the differentiated T cell subsets CD4 TEMRA, CD8 TEM (T effector memory) and TEMRA (effector), and γδ T cells, and in lung cancer tumor infiltrating lymphocytes (TILS) from dataset GSE99531. Correlation analysis was performed of single cell RNAseq gene expression from cancer tissue datasets used in a previously published analysis [10] including GSE72046 (melanoma) [26], GSE98638 (HCC) [27], GSE103322 (HNSCC) [28], and GSE89567 (astrocytoma) [29] and newly analyzed comprehensive datasets GSE102575 (melanoma) [30], GSE108989 (colorectal cancer) [31], and GSE99254 (non-small cell lung cancer) [32]. CD8 T cells were annotated as those T cells with non-zero expression of CD8A. Published graphical data was abstracted using https://apps.automeris.io/wpd/.
Data availability
Data used is publicly available as described in Methods.
CONFLICTS OF INTEREST
SG has completed the ICMJE uniform disclosure form at https://www.icmje.org/coi_disclosure.pdf and declares: SG is a founder and consultant to Abcuro, Inc. and inventor of intellectual property pertaining to KLRG1 targeting, owned and managed by the Brigham and Women’s Hospital. There are no other relationships or activities that could appear to have influenced the submitted work.
FUNDING
SG performed all analyses. No funding was used for this study.
References
1. Pauken KE, Wherry EJ. Overcoming T cell exhaustion in infection and cancer. Trends Immunol. 2015; 36:265–76. https://doi.org/10.1016/j.it.2015.02.008. [PubMed].
2. Mahoney KM, Rennert PD, Freeman GJ. Combination cancer immunotherapy and new immunomodulatory targets. Nat Rev Drug Discov. 2015; 14:561–84. https://doi.org/10.1038/nrd4591. [PubMed].
3. Anderson AC, Joller N, Kuchroo VK. Lag-3, Tim-3, and TIGIT: Co-inhibitory Receptors with Specialized Functions in Immune Regulation. Immunity. 2016; 44:989–1004. https://doi.org/10.1016/j.immuni.2016.05.001. [PubMed].
4. Leach DR, Krummel MF, Allison JP. Enhancement of antitumor immunity by CTLA-4 blockade. Science. 1996; 271:1734–36. https://doi.org/10.1126/science.271.5256.1734. [PubMed].
5. Hirano F, Kaneko K, Tamura H, Dong H, Wang S, Ichikawa M, Rietz C, Flies DB, Lau JS, Zhu G, Tamada K, Chen L. Blockade of B7-H1 and PD-1 by monoclonal antibodies potentiates cancer therapeutic immunity. Cancer Res. 2005; 65:1089–96. [PubMed].
6. Iwai Y, Terawaki S, Honjo T. PD-1 blockade inhibits hematogenous spread of poorly immunogenic tumor cells by enhanced recruitment of effector T cells. Int Immunol. 2005; 17:133–44. https://doi.org/10.1093/intimm/dxh194. [PubMed].
7. Grosso JF, Kelleher CC, Harris TJ, Maris CH, Hipkiss EL, De Marzo A, Anders R, Netto G, Getnet D, Bruno TC, Goldberg MV, Pardoll DM, Drake CG. LAG-3 regulates CD8+ T cell accumulation and effector function in murine self-and tumor-tolerance systems. J Clin Invest. 2007; 117:3383–92. https://doi.org/10.1172/JCI31184. [PubMed].
8. Baghdadi M, Nagao H, Yoshiyama H, Akiba H, Yagita H, Dosaka-Akita H, Jinushi M. Combined blockade of TIM-3 and TIM-4 augments cancer vaccine efficacy against established melanomas. Cancer Immunol Immunother. 2013; 62:629–37. https://doi.org/10.1007/s00262-012-1371-9. [PubMed].
9. Johnston RJ, Comps-Agrar L, Hackney J, Yu X, Huseni M, Yang Y, Park S, Javinal V, Chiu H, Irving B, Eaton DL, Grogan JL. The immunoreceptor TIGIT regulates antitumor and antiviral CD8(+) T cell effector function. Cancer Cell. 2014; 26:923–37. https://doi.org/10.1016/j.ccell.2014.10.018. [PubMed].
10. Greenberg SA, Kong SW, Thompson E, Gulla SV. Co-inhibitory T cell receptor KLRG1: human cancer expression and efficacy of neutralization in murine cancer models. Oncotarget. 2019; 10:1399–1406. https://doi.org/10.18632/oncotarget.26659. [PubMed].
11. Tata A, Dodard G, Fugère C, Leget C, Ors M, Rossi B, Vivier E, Brossay L. Combination blockade of KLRG1 and PD-1 promotes immune control of local and disseminated cancers. Oncoimmunology. 2021; 10:1933808. https://doi.org/10.1080/2162402X.2021.1933808. [PubMed].
12. Wherry EJ, Ha SJ, Kaech SM, Haining WN, Sarkar S, Kalia V, Subramaniam S, Blattman JN, Barber DL, Ahmed R. Molecular signature of CD8+ T cell exhaustion during chronic viral infection. Immunity. 2007; 27:670–84. https://doi.org/10.1016/j.immuni.2007.09.006. [PubMed].
13. Blackburn SD, Shin H, Haining WN, Zou T, Workman CJ, Polley A, Betts MR, Freeman GJ, Vignali DA, Wherry EJ. Coregulation of CD8+ T cell exhaustion by multiple inhibitory receptors during chronic viral infection. Nat Immunol. 2009; 10:29–37. https://doi.org/10.1038/ni.1679. [PubMed].
14. Apetoh L, Smyth MJ, Drake CG, Abastado JP, Apte RN, Ayyoub M, Blay JY, Bonneville M, Butterfield LH, Caignard A, Castelli C, Cavallo F, Celis E, et al. Consensus nomenclature for CD8+ T cell phenotypes in cancer. Oncoimmunology. 2015; 4:e998538. https://doi.org/10.1080/2162402X.2014.998538. [PubMed].
15. Akbar AN, Henson SM. Are senescence and exhaustion intertwined or unrelated processes that compromise immunity? Nat Rev Immunol. 2011; 11:289–95. https://doi.org/10.1038/nri2959. [PubMed].
16. Legat A, Speiser DE, Pircher H, Zehn D, Fuertes Marraco SA. Inhibitory Receptor Expression Depends More Dominantly on Differentiation and Activation than “Exhaustion” of Human CD8 T Cells. Front Immunol. 2013; 4:455. https://doi.org/10.3389/fimmu.2013.00455. [PubMed].
17. Baitsch L, Legat A, Barba L, Fuertes Marraco SA, Rivals JP, Baumgaertner P, Christiansen-Jucht C, Bouzourene H, Rimoldi D, Pircher H, Rufer N, Matter M, Michielin O, Speiser DE. Extended co-expression of inhibitory receptors by human CD8 T-cells depending on differentiation, antigen-specificity and anatomical localization. PLoS One. 2012; 7:e30852. https://doi.org/10.1371/journal.pone.0030852. [PubMed].
18. Schmidt EV, Chisamore MJ, Chaney MF, Maradeo ME, Anderson J, Baltus GA, Pinheiro EM, Uebele VN. Assessment of Clinical Activity of PD-1 Checkpoint Inhibitor Combination Therapies Reported in Clinical Trials. JAMA Netw Open. 2020; 3:e1920833. https://doi.org/10.1001/jamanetworkopen.2019.20833. [PubMed].
19. Schmidt EV, Sun LZ, Palmer AC, Chen C. Rationales for Combining Therapies to Treat Cancer: Independent Action, Response Correlation, and Collateral Sensitivity Versus Synergy. Annu Rev Cancer Biol. 2023; 7:247–63. https://doi.org/10.1146/annurev-cancerbio-061421-020411.
20. Thommen DS, Koelzer VH, Herzig P, Roller A, Trefny M, Dimeloe S, Kiialainen A, Hanhart J, Schill C, Hess C, Savic Prince S, Wiese M, Lardinois D, et al. A transcriptionally and functionally distinct PD-1+ CD8+ T cell pool with predictive potential in non-small-cell lung cancer treated with PD-1 blockade. Nat Med. 2018; 24:994–1004. https://doi.org/10.1038/s41591-018-0057-z. [PubMed].
21. Plana D, Palmer AC, Sorger PK. Independent Drug Action in Combination Therapy: Implications for Precision Oncology. Cancer Discov. 2022; 12:606–24. https://doi.org/10.1158/2159-8290.CD-21-0212. [PubMed].
22. Pomeroy AE, Schmidt EV, Sorger PK, Palmer AC. Drug independence and the curability of cancer by combination chemotherapy. Trends Cancer. 2022; 8:915–29. https://doi.org/10.1016/j.trecan.2022.06.009. [PubMed].
23. Palmer AC, Izar B, Hwangbo H, Sorger PK. Predictable Clinical Benefits without Evidence of Synergy in Trials of Combination Therapies with Immune-Checkpoint Inhibitors. Clin Cancer Res. 2022; 28:368–77. https://doi.org/10.1158/1078-0432.CCR-21-2275. [PubMed].
24. Palmer AC, Sorger PK. Combination Cancer Therapy Can Confer Benefit via Patient-to-Patient Variability without Drug Additivity or Synergy. Cell. 2017; 171:1678–91.e13. https://doi.org/10.1016/j.cell.2017.11.009. [PubMed].
25. Monaco G, Lee B, Xu W, Mustafah S, Hwang YY, Carré C, Burdin N, Visan L, Ceccarelli M, Poidinger M, Zippelius A, Pedro de Magalhães J, Larbi A. RNA-Seq Signatures Normalized by mRNA Abundance Allow Absolute Deconvolution of Human Immune Cell Types. Cell Rep. 2019; 26:1627–40.e7. https://doi.org/10.1016/j.celrep.2019.01.041. [PubMed].
26. Tirosh I, Izar B, Prakadan SM, Wadsworth MH 2nd, Treacy D, Trombetta JJ, Rotem A, Rodman C, Lian C, Murphy G, Fallahi-Sichani M, Dutton-Regester K, Lin JR, et al. Dissecting the multicellular ecosystem of metastatic melanoma by single-cell RNA-seq. Science. 2016; 352:189–96. https://doi.org/10.1126/science.aad0501. [PubMed].
27. Zheng C, Zheng L, Yoo JK, Guo H, Zhang Y, Guo X, Kang B, Hu R, Huang JY, Zhang Q, Liu Z, Dong M, Hu X, et al. Landscape of Infiltrating T Cells in Liver Cancer Revealed by Single-Cell Sequencing. Cell. 2017; 169:1342–56.e16. https://doi.org/10.1016/j.cell.2017.05.035. [PubMed].
28. Puram SV, Tirosh I, Parikh AS, Patel AP, Yizhak K, Gillespie S, Rodman C, Luo CL, Mroz EA, Emerick KS, Deschler DG, Varvares MA, Mylvaganam R, et al. Single-Cell Transcriptomic Analysis of Primary and Metastatic Tumor Ecosystems in Head and Neck Cancer. Cell. 2017; 171:1611–24.e24. https://doi.org/10.1016/j.cell.2017.10.044. [PubMed].
29. Venteicher AS, Tirosh I, Hebert C, Yizhak K, Neftel C, Filbin MG, Hovestadt V, Escalante LE, Shaw ML, Rodman C, Gillespie SM, Dionne D, Luo CC, et al. Decoupling genetics, lineages, and microenvironment in IDH-mutant gliomas by single-cell RNA-seq. Science. 2017; 355:eaai8478. https://doi.org/10.1126/science.aai8478. [PubMed].
30. Sade-Feldman M, Yizhak K, Bjorgaard SL, Ray JP, de Boer CG, Jenkins RW, Lieb DJ, Chen JH, Frederick DT, Barzily-Rokni M, Freeman SS, Reuben A, Hoover PJ, et al. Defining T Cell States Associated with Response to Checkpoint Immunotherapy in Melanoma. Cell. 2018; 175:998–1013.e20. https://doi.org/10.1016/j.cell.2018.10.038. [PubMed].
31. Zhang L, Yu X, Zheng L, Zhang Y, Li Y, Fang Q, Gao R, Kang B, Zhang Q, Huang JY, Konno H, Guo X, Ye Y, et al. Lineage tracking reveals dynamic relationships of T cells in colorectal cancer. Nature. 2018; 564:268–72. https://doi.org/10.1038/s41586-018-0694-x. [PubMed].
32. Guo X, Zhang Y, Zheng L, Zheng C, Song J, Zhang Q, Kang B, Liu Z, Jin L, Xing R, Gao R, Zhang L, Dong M, et al. Global characterization of T cells in non-small-cell lung cancer by single-cell sequencing. Nat Med. 2018; 24:978–85. https://doi.org/10.1038/s41591-018-0045-3. [PubMed].