Cannabis Sativa

TNFRSF9
Available structures
PDBOrtholog search: PDBe RCSB
Identifiers
AliasesTNFRSF9, 4-1BB, CD137, CDw137, ILA, tumor necrosis factor receptor superfamily member 9, TNF receptor superfamily member 9
External IDsOMIM: 602250 MGI: 1101059 HomoloGene: 1199 GeneCards: TNFRSF9
Orthologs
SpeciesHumanMouse
Entrez
Ensembl
UniProt
RefSeq (mRNA)

NM_001561

NM_001077508
NM_001077509
NM_011612

RefSeq (protein)

NP_001552

NP_001070976
NP_001070977
NP_035742

Location (UCSC)Chr 1: 7.92 – 7.94 MbChr 4: 151 – 151.03 Mb
PubMed search[3][4]
Wikidata
View/Edit HumanView/Edit Mouse

CD137, a member of the tumor necrosis factor (TNF) receptor family, is a type 1 transmembrane protein, expressed on surfaces of leukocytes and non-immune cells.[5] Its alternative names are tumor necrosis factor receptor superfamily member 9 (TNFRSF9), 4-1BB, and induced by lymphocyte activation (ILA). It is of interest to immunologists as a co-stimulatory immune checkpoint molecule, and as a potential target in cancer immunotherapy.

Expression[edit]

CD137 is only expressed on the cell surface after T cell activation. When T cells are activated by Antigen Presenting Cells (APCs), CD137 becomes embedded in CD4+ and CD8+ T cells.

CD137 is a costimulatory molecule functioning to stimulate T cell proliferation, dendritic cell maturation, and promotion of B cell antibody secretion.[6] As a T cell co-stimulator, T cell receptor (TCR) and CD28 signaling causes expression of CD137 on T cell membranes. When CD137 then reacts with the CD137 ligand, it leads to CD137 upregulation.[6] This is a form of self regulation or positive feedback cycle. When CD137 interacts with its ligand, it leads to T cell cytokine production and T cell proliferation, among other signaling pathway responses.

Other cells that express CD137 include both immune cells (i.e. monocytes, natural killer cells, dendritic cells, follicular dendritic cells (FDCs), and regulatory T cells) and non-immune cells (i.e. chondrocytes, neurons, astrocytes, microglia and endothelial cells).[6]

Regulation of the immune system[edit]

CD137 and its ligand both induce signaling cascades upon interaction, a phenomenon known as bidirectional signal transduction. The CD137/ligand complex is also involved in regulation of the immune system. The CD137 ligand is a type-II transmembrane glycoprotein expressed on APCs.[7] The CD137 ligand is normally expressed at low levels, but can have increased expression in presence of pathogen associated molecular patterns (PAMPs) or proinflammatory immune responses like IL-1 secretion.

Cross-linking CD137 and active T cells can not only result in T cell proliferation via increased IL-2 secretion, but surviving cells also contribute to expanding immune system memory and augmenting T cell cytolytic activity.[7]

Atherosclerosis[edit]

Inflammation[edit]

Atherosclerosis is a disease, linked to Cardiovascular Disease (CVD), and associated with cardiac inflammation, in the form of lesions in the walls of the atrial chambers and other vasculature.[8] Treatments designed to target the CD137 molecules expressed on immune cell surfaces often lead to T cell proliferation as CD137 stimulation allows for the T cells to continue through the cell cycle. In this way, CD137 is often referred to as an immune checkpoint. This proliferation eventually leads to other immune cell responses and secretion of proinflammatory cytokines which result in exaggerated inflammatory responses that exacerbate atherosclerosis.[8] Ongoing studies are researching CD137 as a biomarker for atherosclerosis as well as CD137 antagonists as potential therapeutics to reduce the symptoms associated with the condition.

Endothelial cells[edit]

The mechanism connecting CD137 bidirectional signaling to the promotion of atherosclerosis is related to CD137 mediation of epithelial cell damage. When the CD137/CD137L complex interacts with endothelial cells, including those lining vascular structures, it induces the upregulation of molecules that promote inflammation and damage. For instance, increases in adhesion molecules, including vascular adhesion molecule-1 or intracellular adhesion molecule-1, on epithelial cells causes recruitment of immune cells like macrophages and neutrophils.[9] When they arrive, these cells initiate proinflammatory responses including cytokine secretion. In chronic cases, this results in excessive inflammation of the epithelial tissue, leading to cell damage and the formation of atherosclerotic inflammatory lesions.[9]

Interactions[edit]

CD137 has been shown to interact with TRAF2.[10][11]

As a drug target[edit]

Cancer immunotherapy[edit]

CD137 is also involved in cancer having been found upregulated in cancerous cell lines. CD137/ligand stimulation has been found to lead to stronger anti-tumor responses due to cytotoxic T cell activation and is being examined as a possible anticancer therapy.[12]

Current cancer immunotherapy treatments use monoclonal antibodies (mAbs) to target and kill cancer cells. Cancer cells upregulate cell surface CD137, however the reason behind this remains unclear. What is known is the fact that mAbs targeting CD137 are successful in fighting cancer as they can not only mark cancer cells, but they allow for CD8+ T cell activation and increased IFN-gamma secretion as per CD137’s function as a costimulatory molecule.[13] This enables the affected individual’s immune system to actively target and kill cancer cells that express CD137 on their cell surfaces. Currently, Utomilumab is the only mAb targeting CD137 on the market.[14] Urelumab trials were temporarily halted due to risk of liver toxicity. Utomilumab trials resulted in the drug’s being cleared for therapeutic use.

Utomilumab[edit]

Utomilumab (PF-05082566) targets this receptor to stimulate a more intense immune system attack on cancers.[15] It is a fully human IgG2 monoclonal antibody.[16] It is in early clinical trials.[15] As of June 2016, 5 clinical trials are active.[17]

See also[edit]

References[edit]

  1. ^ a b c GRCh38: Ensembl release 89: ENSG00000049249Ensembl, May 2017
  2. ^ a b c GRCm38: Ensembl release 89: ENSMUSG00000028965Ensembl, May 2017
  3. ^ "Human PubMed Reference:". National Center for Biotechnology Information, U.S. National Library of Medicine.
  4. ^ "Mouse PubMed Reference:". National Center for Biotechnology Information, U.S. National Library of Medicine.
  5. ^ Thum E, Shao Z, Schwarz H (January 2009). "CD137, implications in immunity and potential for therapy". Frontiers in Bioscience. 14 (11): 4173–4188. doi:10.2741/3521. PMID 19273343.
  6. ^ a b c Thum E, Shao Z, Schwarz H (January 2009). "CD137, implications in immunity and potential for therapy". Frontiers in Bioscience. 14 (11): 4173–4188. doi:10.2741/3521. PMID 19273343.
  7. ^ a b Thum E, Shao Z, Schwarz H (January 2009). "CD137, implications in immunity and potential for therapy". Frontiers in Bioscience. 14 (11): 4173–4188. doi:10.2741/3521. PMID 19273343.
  8. ^ a b Söderström LÅ, Tarnawski L, Olofsson PS (May 2018). "CD137: A checkpoint regulator involved in atherosclerosis". Atherosclerosis. 272: 66–72. doi:10.1016/j.atherosclerosis.2018.03.007. PMID 29571029.
  9. ^ a b Yuan W, Xu C, Li B, Xia H, Pan Y, Zhong W, et al. (June 2021). "Contributions of Costimulatory Molecule CD137 in Endothelial Cells". Journal of the American Heart Association. 10 (11): e020721. doi:10.1161/JAHA.120.020721. PMC 8483511. PMID 34027676.
  10. ^ Jang IK, Lee ZH, Kim YJ, Kim SH, Kwon BS (January 1998). "Human 4-1BB (CD137) signals are mediated by TRAF2 and activate nuclear factor-kappa B". Biochemical and Biophysical Research Communications. 242 (3): 613–620. doi:10.1006/bbrc.1997.8016. PMID 9464265.
  11. ^ Arch RH, Thompson CB (January 1998). "4-1BB and Ox40 are members of a tumor necrosis factor (TNF)-nerve growth factor receptor subfamily that bind TNF receptor-associated factors and activate nuclear factor kappaB". Molecular and Cellular Biology. 18 (1): 558–565. doi:10.1128/MCB.18.1.558. PMC 121523. PMID 9418902.
  12. ^ Makkouk A, Chester C, Kohrt HE (February 2016). "Rationale for anti-CD137 cancer immunotherapy". European Journal of Cancer. 54: 112–119. doi:10.1016/j.ejca.2015.09.026. PMID 26751393.
  13. ^ Chu DT, Bac ND, Nguyen KH, Tien NL, Thanh VV, Nga VT, et al. (April 2019). "An Update on Anti-CD137 Antibodies in Immunotherapies for Cancer". International Journal of Molecular Sciences. 20 (8): 1822. doi:10.3390/ijms20081822. PMC 6515339. PMID 31013788.
  14. ^ Jhajj HS, Lwo TS, Yao EL, Tessier PM (January 2023). "Unlocking the potential of agonist antibodies for treating cancer using antibody engineering". Trends in Molecular Medicine. 29 (1): 48–60. doi:10.1016/j.molmed.2022.09.012. PMC 9742327. PMID 36344331.
  15. ^ a b "Pfizer cancer drug shows promise in combo with Merck's Keytruda". Reuters. May 2016.
  16. ^ Thall A (May 2016). Phase 1 Study of Utomilumab (PF-05082566) In Combination with Rituximab in Patients with CD20+ NHL (PDF). New Therapies in Hematology. Bologna, Italy. Study B1641001).
  17. ^ "PF-05082566 clinical trials". clinicaltrials.gov.

External links[edit]

Further reading[edit]

Leave a Reply