Open Access

Progress of research on γδ T cells in colorectal cancer (Review)

  • Authors:
    • Lijuan Pan
    • Yiru Zhou
    • Yeye Kuang
    • Chan Wang
    • Weimin Wang
    • Xiaotong Hu
    • Xiabin Chen
  • View Affiliations

  • Published online on: October 4, 2024     https://doi.org/10.3892/or.2024.8819
  • Article Number: 160
  • Copyright: © Pan et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Colorectal cancer (CRC) ranks as the third most prevalent malignancy and second leading cause of cancer‑related fatalities worldwide. Immunotherapy alone or in combination with chemotherapy has a favorable survival benefit for patients with CRC. Unlike αβ T cells, which are prone to drug resistance, γδ T cells do not exhibit major histocompatibility complex restriction and can target tumor cells through diverse mechanisms. Recent research has demonstrated the widespread involvement of Vδ1T, Vδ2T, and γδ T17 cells in tumorigenesis and progression. In the present review, the influence of different factors, including immune checkpoint molecules, the tumor microenvironment and microorganisms, was summarized on the antitumor/protumor effects of these cells, aiming to provide insights for the development of more efficient and less toxic immunotherapy‑based anticancer drugs.

Introduction

Colorectal cancer (CRC) ranks as the third most prevalent malignancy and the second leading cause of cancer-related mortality worldwide (1). Previous clinical studies have demonstrated that immunotherapy monotherapy or in combination with chemotherapy confers a favorable survival benefit for patients with CRC (2). Previously, CRC immunotherapy focused primarily on αβ T cells, which exert cytotoxicity by recognizing mutant antigens in tumor cells through the major histocompatibility complex (MHC) (3). However, cancer cells typically exhibit depletion of MHC molecules, which renders tumor cells immune to αβ T-cell-mediated cell mortality (4). Another T-cell type in humans, the γδ T cell, exhibits MHC-unrestricted lytic activity against different tumor cells in vitro, suggesting the possibility for application in cancer treatment (5).

In humans, γδ T cells associated with CRC can be generally categorized into three types according to the chains on the T-cell receptor (TCR) surface: Vδ1, Vδ2 and Vδ3 T cells (6). The thymus and mucosal epithelial tissues contain the majority of Vδ1 T lymphocytes, which release various cytokines, including tumor necrosis factor-alpha (TNF-α) and interferon-gamma (IFN-γ), which have cytotoxic effects on tumor cells and are crucial in the development of numerous illnesses. Vδ2 T cells comprise 50–90% of all γδ T cells, mostly in the peripheral circulation. The TCR of Vδ2 T cells primarily utilizes Vγ9 and Vδ2, which may detect phosphorylated antigens for activation and release perforin and granzymes, resulting in cytotoxicity. Activated Vδ2 T cells can act as antigen-presenting cells (68). The proportion of Vδ3 T cells among the total γδ T cells is <1%, and Vδ3 T cells are predominantly localized in the liver and intestine (9). These cells exhibit cytotoxicity through the expression of genes encoding cytotoxic molecules such as granzyme B, perforin, granulysin, and also possess NKG2D receptors for tumor cell recognition and elimination (10). γδ T cells can be classified into regulatory γδ, γδ T17, IFN-γ+ γδ, and other functional types. The main impediment to the therapeutic application of these cells lies in the immune evasion mechanisms employed by tumor cells (11). Tumor cells can alter the function of the host immune system and create a tumor microenvironment (TME) conducive to tumor development, allowing immune evasion (12). Additionally, several studies have demonstrated a correlation between the gut microbiota and γδ T cells (13), with an imbalance in the gut microbiota potentially promoting the progression of inflammation toward CRC (14). An understanding of γδ T-cell characteristics and the mechanism of action involving the TME and the gut microbiota with γδ T cells will facilitate the development of novel anti-CRC therapeutics and establish a foundation for clinical treatment combinations.

Vδ1 T cells

The predominant infiltrating γδ T cells in CRC tissues are Vδ1 T cells (15). Vδ1 T cells have been shown to exert anticancer effects in colon cancer through the secretion of enzymes and proteins (CD107a, granzyme B and perforin) and direct interactions with cytotoxicity-related receptors and ligands (Fas, MICA/B, death receptor 4/5 and ICAM-1) (16,17). NKp46 is one of the three natural cytotoxic receptors first identified as a germline-encoded protein. The percentages of total Vδ1 and NKp46+/Vδ1 subgroups among intraepithelial lymphocytes (IELs) in CRC tumors are significantly lower than those in disease-free/healthy intestinal tissue samples. Additionally, there is a correlation between a decreased frequency of NKp46+/Vδ1 IEL subgroups in healthy intestinal tissue samples from patients with CRC and faster tumor growth and the emergence of metastatic illness (18). The liver of patients with CRC liver metastasis is infiltrated by CD69 Vδ1 T cells, which play crucial roles in limiting metastasis. These cells can also be used as a reliable prognostic marker in ‘liquid biopsy’ (19). De Vries et al (10) revealed that PD1+ Vδ1 T cells can eliminate tumor cells via the NKG2D/NKG2D-ligand interaction pathway (10). In addition to their potential as antitumor agents, Vδ1 T cells have demonstrated the ability to prevent tumor metastasis, effectively suppressing primary tumor growth and inhibiting the development of spontaneous liver and lung metastases in a xenograft model utilizing immunodeficient mice (20).

Currently, there is a paucity of research on Vδ1 T cells in CRC, likely because of the heterogeneous nature of Vδ1 T-cell populations in this malignancy (21), which poses challenges for investigation. The feasibility of categorizing Vδ1 T cells and selectively acquiring distinct subsets of Vδ1 T cells for targeted investigations may be explored in the future.

Vδ2 T cells

The reduced presence of Vδ2 T cells in patients with colitis-induced cancers can potentially be attributed to impaired recruitment of Vδ2 T cells from the peripheral circulation and sustained inflammatory processes resulting in the depletion of Vδ2 T cells (22). A potential strategy for treating tumors involves promoting the proliferation and augmenting the functionality of Vδ2 T cells. Currently, research efforts have focused predominantly on investigating the antitumor potential of Vγ9Vδ2 T cells.

Antitumor effects of Vγ9Vδ2 T cells

The recognition of tumor cells by Vγ9Vδ2 T lymphocytes is predominantly MHC-unrestricted, with CRC cell lines being recognized by ascites-derived Vγ9Vδ2 clones and regulated by both TCR-dependent and TCR-independent signals (23,24). It has been reported that Vγ9Vδ2 T cells recognize tumor cells through the CDR3δ region of the γδ-TCR (25). In a subsequent study, Zhao et al (26) engineered CDR3δ-transplanted Vγ9Vδ2 T cells capable of producing antitumor cytokines upon stimulation with tumor cell extracts. Furthermore, this antitumor effect was attenuated by the administration of anti-γδ-TCR monoclonal antibodies (26). Another study identified specific sequence and structure patterns in CDR3δ, including rearrangement within the J1 region, the presence of atypical T-cell receptor genes, the positioning of hydrophobic amino acids in CDR3δ, the distribution of CDR3δ lengths, and the number of N insertions. These factors may impact the affinity between T-cell receptors and antigens, consequently influencing T-cell activation and expansion (27).

The activation of Vγ9Vδ2 T cells can be induced by the overexpression of phospho-antigen (pAg) (18) and the interaction between NKG2D receptors and ligands in CRC (28,29). Once activated, Vγ9Vδ2 T cells can eliminate tumor cells through various mechanisms, including the engagement of death receptors/ligands with Fas ligands and tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) and the secretion of perforins, cytokines (such as TNF-α), or granzymes (30). These pAgs are mainly pyrophosphates produced in eukaryotes via the mevalonate pathway (31). Different phosphate antigens activate Vγ9Vδ2 T cells through different mechanisms. For example, bromo-hydro-pyrophosphate directly stimulates Vγ9Vδ2 T cells, whereas amino-bisphosphonates, such as pamidronate and zoledronate, indirectly activate Vγ9Vδ2 T cells by inhibiting the mevalonate pathway, thereby increasing the intracellular accumulation of isopentenyl pyrophosphate (IPP) (32,33). IPP accumulates in numerous types of cancer, and the resulting disordered metabolic processes render cancer cells susceptible to Vγ9Vδ2 T-cell-mediated mortality (6). Reportedly, interleukin-2 (IL-2) stimulates the production of the adaptor molecule DAP10, increasing the surface expression of NKG2D (34). Similarly, Smyth et al (35) reported that the cytotoxicity of IL-12-induced cells toward tumor cells is contingent upon the interaction between NKG2D and its corresponding ligand. Pei et al (36) reported that CD137 co-stimulation can overcome the inhibitory effect of endogenous IL-10 (hIL-10 and vIL-10) on the antitumor activity of Vγ9Vδ2 T cells, thereby enhancing the efficacy of this specific subset in tumor therapy. However, according to Zhang et al (37), soluble NKG2DLs impair the cytotoxicity of γδ T cells to tumor cells. Therefore, increasing the expression of NKG2DLs within tumors or employing targeted delivery of synthetic adhesives to tumors may be an effective approach for enhancing the antitumor efficacy of γδ T cells.

Dual effects of drug treatment on Vγ9Vδ2 T-cell toxicity

Evidence from three lines of investigation demonstrated that chemotherapy enhances the susceptibility of colonic cancer initiating stem cells (CICs) to Vγ9Vδ2 T-cell toxicity. Pioneering work by Mattarollo et al (38) demonstrated that the combination of Vγ9Vδ2 T cells and chemotherapeutic agents yields a high level of cytotoxicity in cell lines derived from solid tumors. IL-17-producing γδ T cells play a decisive role in immune responses against cancer induced by chemotherapy in mice (39). Simultaneous or immediate in vivo activation of Vγ9Vδ2 T cells or adoptive transfer of in vitro-activated Vγ9Vδ2 T lymphocytes following treatment with the chemotherapeutic drugs 5-fluorouracil (5-FU) and doxorubicin (DXR) significantly increased antitumor activity (7).

Vγ9Vδ2 T-cell elimination post-chemotherapy in CICs is mediated through the activation of NKG2D and TRAIL (7). 5-FU and DXR significantly increase the expression of DR5 (TRAIL-R2) in colon cancer stem cells (CSCs). Additionally, the anti-NKG2D mAb effectively suppresses the cytotoxicity of Vγ9Vδ2 T cells against colon CSCs, whereas neither anti-CD3 nor anti-TCR antibodies nor mevastatin (a 3-hydroxy-3-methylglutaryl coenzyme A reductase inhibitor that prevents endogenous pAg accumulation) demonstrate significant inhibitory effects (34). The expression of NKG2D ligands on tumor cells can be induced by various drugs, including proteasomes, histone deacetylases, heat shock proteins and apoptosis inhibitors (4044), thereby increasing the toxicity of Vγ9Vδ2 T cells and inhibiting tumor development. In addition, Benelli et al (45) developed a Cet-ZA antibody-drug conjugate (ADC) that targets CRC cells and enhances Vδ2 T-cell cytotoxicity through the TCR pathway (Fig. 1). Although numerous studies are underway, drug toxicity and targeting remain challenges.

γδ T17 cells

γδ T17 cells represent a prominent source of IL-17 within the TME. Activated inflammatory dendritic cells (inf-DCs) can induce γδ T17 cells to generate TNF-α, IL-8 and GM-CSF, while immunosuppressive polymorphonuclear myeloid-derived suppressor cells (PMN-MDSCs) accumulate in tumors. The regulatory axis of inf-DC-γδT17-PMN-MDSCs in human CRC establishes a connection between MDSC-mediated immunosuppression and tumor-induced inflammation, highlighting the potential role of γδ T17 cells in the progression of human CRC (46).

The percentage of tumor-infiltrating γδ T17 cells positively correlates with the progression of TNM stage and other clinicopathological characteristics, including tumor size, tumor invasion, lymphatic and vascular invasion, lymph node metastasis and the serum carcinoembryonic antigen level (46,47). Furthermore, inf-DC, PMN-MDSC, IL-23 and IL-17 levels in tumor tissue are significantly related to the proportion of tumor-infiltrating γδ T17 cells (46,48). Following acute intestinal injury, IL-23R+RORγT+γδ T cells located in the colonic lamina propria serve as pivotal sources of initial protective IL-17 within the intestines, playing an indispensable role in preserving and enhancing the integrity of the intestinal mucosal epithelial barrier (49). The dual role of γδ T17 cells in tumors poses a challenge for developing immunotherapies targeting this specific cell subset. Further comprehensive investigations are warranted to elucidate their functional pathways within the TME and identify pivotal breakthroughs.

Discrimination of pro- and antitumor intestinal γδ T-cell subsets

Using human CRC samples and mouse CRC models, Reis et al (50) discovered that in premalignant or nontumor colons, most γδ T cells exhibit cytotoxic markers, whereas tumor-infiltrating γδ T cells display protumorigenic characteristics. The aforementioned observation is linked to distinct TCR-Vγδ gene expression patterns in both humans and mice.

The γδ T cells that produce IFN-γ, particularly the Vγ1+ and Vγ7+ cells, exhibit antitumor activity that is dependent on Glut1 expression (50). These findings suggest a link between diabetes and cancer. A study conducted by Mu et al (51) on tumor immune monitoring in diabetic patients via γδ T cells also demonstrated that elevated glucose levels can impair the antitumor activity of Vγ9Vδ2 T cells through lactate-induced inhibition of AMPK activation, resulting in a reduced ability to secrete lytic granules and increased susceptibility to cancer in individuals with type 2 diabetes. IL-17-producing γδ T17 cells express Vγ6 (according to the Vγ nomenclature of Heilig and Tonegawa) and Vγ4 TCR chains (52), which rely on oxidative phosphorylation, continuously proliferate in lipid-rich environments, such as tumors, and promote tumor progression, indicating that this may be another mechanism connecting cancer and obesity (53). In addition, the balance of tissue recovery mechanisms may involve cytokines or molecules other than IL-17 produced by Vγ4+ or Vγ6+ cells, which can also promote the formation of tumors (50). This effect is because the generation of IL-17 by γδ T cells in the gut is also related to tissue healing (49,50) (Table I).

Table I.

Classification of γδ T cells.

Table I.

Classification of γδ T cells.

Classification basisCell typeCritical functionsSupplement(Refs.)
δ chainVδ1 T cells• Secrete cytokines: TNF-α, IFN-γ;• Are the mainly invasive γδ(6,7,10,
• Secrete enzymes and proteins: CD107a, granzyme B, and perforin;T cells in rectal cancer tissue;1518,21)
• Express cytotoxicity-related receptors and ligands: Fas, MICA/B, death receptor 4/5 and ICAM-1, NKp46, NKG2D• Have heterogeneity in tumors, are less researched
Vδ2 T cells• Release perforin, cytokines and granzyme;• Comprise 50 to 90% of all(68,25,
• Act as antigen present cells;γδ T cells;2830,
• Recognize tumor cells through the CDR3δ region of γδ-TCR;• Numerous studies have been conducted on63,76,79)
• Bind to death ligands expressed by tumor cells: Fas ligands, TRAILs, NKG2DLs;Vγ9Vδ2 T cells
• Express immunosuppression related genes: B7-H3, PD-1, Tim-3
Vδ3 T cells• Secrete granzyme B, perforin, granulysin; Express NKG2D receptors• Are the lowest, less than 1%(9,10)
Function IFN-γ-producing• Produce IFN-γ• Mainly include Vγ1+ and Vγ7+ cells;
γδ T cells • Antitumor activity was dependent on Glut1 expression(50)
IL-17-producing• Produce IL-17• Mainly include Vγ4+ and Vγ6+ cells;
γδ T cells • Protumor activity was dependent on lipid content(52,53)

TCR sequencing research has shown that γδ T cells with antitumor characteristics include polyclonal Vγ7+ and Vγ1+ cells, and a minority of tumor-promoting cells that produce IL-17 are Vγ4+ cells; most are clonally expanded Vγ6Vδ1+ cells (54). Although Vγ6+ cells are the predominant progenitor subset in tumors, Vγ4+ cells appear to be able to compensate when Vγ6+ cells are damaged, similar to Vγ7+ cells (mostly gut-specific γδ T cells) and Vγ1+ cells (with broad tissue distribution); elimination of Vγ1+ cells from the tumor is required when performing antitumor functional analyses of Vγ7+ cells (54,55).

Immune checkpoint genes that act on γδ T cells

The utilization of synthetic immune checkpoint inhibitors has emerged as a prominent area of research in the field of CRC immunotherapy and has demonstrated remarkable efficacy, especially in patients with microsatellite instability (MSI)-high CRC (56). These agents target immune checkpoints, such as the programmed cell death protein 1 (PD-1)/programmed cell death-Ligand 1 (PD-L1) pathway, which tumors utilize to evade detection by the immune system. By obstructing this interaction, these inhibitors can augment the immune response against cancer cells (57). Despite the potential for adverse effects, immune checkpoint inhibitors have been shown to have a greater safety profile than chemotherapy (5860). Several studies on γδ T cells have identified immune checkpoint genes, which are expected to be used to screen drugs for the treatment of CRC.

Inhibitory effect of the downregulation of Ten Eleven Translocation 1 (Tet1) on γδ T cells

Tie et al (61) reported that hypercholesterolemia leads to oxidative stress in hematopoietic stem cells (HSCs), accelerating HSC senescence and impairing the regenerative capacity of HSCs (61). Tet1 is a direct target of miR101c, and mechanistic studies have revealed that hypercholesterolemia induces oxidative stress that is mediated by miR101c, which causes Tet1 to be downregulated in HSCs. This effect causes genes essential for natural killer T (NKT) and γδ T-cell development to undergo an increase in DNA hypermethylation and histone alterations (Fig. 2). Consequently, the quantity and functionality of terminally differentiated NKT and γδ T cells within the thymus, colonic submucosa, and early stages of tumorigenesis are reduced. This impairment compromises immune surveillance against colonic tumors, which can be ameliorated by restoring Tet 1 expression (62).

Inhibition of Vδ2 T-cell cytotoxicity by B7-H3

Despite a significant reduction in the proportion of γδ T cells in both peripheral blood mononuclear cells and tumor areas among patients with colon cancer, there is an increase in the proportion of B7-H3+γδ T lymphocytes. It is postulated that B7-H3 functions as a negative immune checkpoint molecule, modulating the activity and biological function of γδ T cells in colon cancer. It has been revealed that blocking or reducing B7-H3 leads to enhanced proliferation, inhibition of apoptosis, and upregulation of activation markers (CD25 and CD69) in Vδ2 T cells. Conversely, the B7-H3 agonist 4H7 exerts the opposite effect. In the presence of IL-2 and zoledronic acid, Vδ2 T cells treated with MIH35 (a specific inhibitory antibody against B7-H3) or B7-H3 siRNA presented increased cell viability, a reduced rate of apoptosis, and increased expression of the signaling molecules CD25 and CD69 (63).

The inhibition of Vδ2 T cells by B7-H3 is mediated mainly by the suppression of T-bet and a decrease in IFN-γ and perforin/granzyme B expression, which involves STAT3 activation and a reduction in ULBP2 expression (11,63). Cryptotanshinone, an inhibitor of STAT3 phosphorylation, can reverse the decrease in ULBP2 expression and attenuate the B7-H3 overexpression-induced elimination of colon cancer cells by Vδ2 T cells (11). The B7-H3-mediated STAT3/ULBP2 axis may be a potential target for enhancing the efficiency of γδ T-cell-based colon cancer immunotherapy (11,63).

Diverse impacts of BTN/BTNL on γδ T cells

In mice, Btnl proteins are predominantly expressed on the epithelial cells lining the intestinal villi (64). Previously, the expression of Btnl1 in intestinal villi in the early stage of life was shown to selectively promote the maturation and proliferation of Vγ7+ IELs in tissues (65), revealing its antitumor potential, whereas the expression of Btnl2 in tumor cells specifically recruits IL-17-producing γδ T cells that promote tumorigenesis (66).

The Butinophil-3A (BTN3A, also known as CD277) protein subfamily plays a crucial role in the antitumor process of γδ T cells by serving as a pivotal mediator of pAg signal transduction (67). The BTN3A molecular subfamily is part of the B7 costimulatory molecular family and includes the BTN3A1, BTN3A2 and BTN3A3 subtypes (68). The three subtypes can stimulate Vγ9Vδ2 T cells following treatment with the 20.1 agonist mAb, activating the cells through mechanisms involving mobility reduction (67) and BTN3A molecular polymerization (69). However, BTN3A1 cannot mediate the activation of Vγ9Vδ2 T cells without BTN3A2 or BTN3A3; Cano et al (70) also demonstrated that BTN3A-mediated cytotoxicity of Vγ9Vδ2 T cells toward cancer cells must involve BTN2A1.

De Gassart et al (71) developed a humanized monoclonal antibody, ICT01, which has sub-nanomolar affinity for all three subtypes of BTN3A. Its activity depends on BTN3A and BTN2A (Fig. 3). The activation of Vγ9Vδ2 T cells by ICT01 eliminates multiple tumor cell lines and primary tumor cells (71). It has been reported that periplakin and RhoB are pivotal in activating Vγ9Vδ2 T cells, mediated by BTN3A (72). Additionally, Vγ9Vδ2 T cells exhibit cytotoxicity against CRC cell lines upon exposure to zoledronate, which is also related to the expression of BTN3A1 in the membrane and cytoskeleton and its redistribution in cells (32). Due to the absence of a B30.2 intracellular domain, the BTN3A2 subtype fails to activate Vγ9Vδ2 T cells when pAgs accumulate. Consequently, it can be considered a decoy receptor, and its increased expression in acute myeloid leukemia primitive cells or other tumors may constitute an immune escape mechanism recognized by Vγ9Vδ2 T cells (72).

Human intestinal epithelial cells express BTNL3 and BTNL8, and the concurrent expression of BTNL3+BTNL8 induces a selective TCR-dependent response in Vγ4+ cells of the human colon (65). According to the analysis by Blazquez et al (72), the homing and maintenance of BTNL3 and BTNL8 in the semi-activated state in human intestinal Vγ4+ γδ T cells may be relevant to the pathogenesis of intestinal autoimmune disorders, such as ulcerative colitis and inflammatory bowel disease. Chronic intestinal inflammation can promote the formation of colorectal tumors (73,74), revealing the correlation between BTNL3 and BTNL8 and CRC. Lebrero-Fernández et al (75) reported significantly lower levels of BTNL3 and BTNL8 expression in colon cancer tissues than in adjacent normal tissues, providing further support for this notion.

Other immune checkpoint genes

The expression of PD-1 can serve as a partial indicator of γδ T-cell function and impact patient prognosis. However, the upregulation of PD-1 alone is insufficient to fully characterize the functional phenotype of γδ T cells in cancer, necessitating comprehensive evaluation of other markers and indicators (76). In academic research, PD-1 is frequently investigated in conjunction with Tim-3 (77), whereas in clinical practice, the combination of PD-1 and CTLA-4 antibodies has demonstrated successful outcomes in the treatment of CRC (78). As a crucial negative regulator of Vγ9Vδ2 T-cell activation, Tim-3 was found to downregulate the expression of perforin and granzyme B in Vγ9Vδ2 T cells via an ERK1/2 signaling pathway-dependent mechanism, thereby attenuating the cytotoxicity of Vγ9Vδ2 T cells to colon cancer cells (79) (Fig. 4). The inhibitory receptors CTLA-4, LAG-3 and TIGIT have been demonstrated to be present on the surface of T cells (8082). However, the specific mechanism underlying their interaction with γδ T cells remains unclear, particularly in treating CRC. Further investigations into the mechanisms underlying these immune checkpoint genes and the development of diverse immune checkpoint inhibitors for combination therapy may represent promising approaches to enhance the current landscape of CRC treatment.

Obstruction of the antitumor process of γδ T cells via the TME

The CRC TME is a complex communication system comprising cancer cells and various other cell types (including endothelial cells, immune cells and cancer-associated fibroblasts). This intricate communication relies on a dysregulated regulatory network comprising chemokines, cytokines, growth factors and their corresponding receptors. Consequently, this dynamic interaction gives rise to an inflammatory TME that facilitates tumorigenesis and progression (83).

In the TME, CRC can be divided into ‘hot’ and ‘cold’ subtypes. Hot tumors are characterized by the presence of activated immune cells that exhibit proinflammatory cytokine signaling, and immune checkpoint inhibitors have shown promising efficacy in inhibiting the growth of such tumors (84). By contrast, cold tumors typically express receptors and ligands associated with immunosuppression and are encompassed by populations of immunosuppressive cells, including regulatory T cells (Tregs), MDSCs and tumor-associated macrophages (85). T Immunosuppressive cells can express IL-10 and TGF-β, thereby impeding the infiltration and functionality of effector T cells, including γδ T cells, while facilitating immune evasion (33,85,86). Modification of the immune microenvironment is essential for treating this type of tumor, including converting a cold tumor into a hot tumor or enhancing effector cell function to achieve effective immunotherapy (84).

Hu et al (87) reported that TGF-β1 derived from human CRC could induce CD39γδ T cells from paired normal colon tissue to differentiate into CD39γδ Tregs and that differentiated CD39γδ Tregs could exert adenosine-mediated immunosuppressive activity (87). Another study revealed that the polarization of CD39γδ Tregs is also related to arachidonic acid. Owing to the abnormal activation of the phospholipase a2-IVa/arachidonic acid metabolic pathway, the content of tumor-infiltrating CD39γδ Tregs in right-sided CRC is markedly greater than that in left-sided CRC, indicating a poor prognosis (88). Inhibiting the production and function of CD39γδ Tregs may represent a promising strategy to improve the prognosis of patients with CRC. In addition, hypoxia is a characteristic shared by numerous solid tumors (89). Exosomes undergo alterations in the hypoxic TME and can enhance the inhibitory impact of MDSCs on γδ T cells through a regulatory axis involving miR-21/PTEN/PD-L1 (90). Combining immunotherapy with strategies to increase the tumor oxygen content may improve the treatment outcome for patients with CRC.

The microbiota is involved in the antitumor process of γδ T cells

Various studies have demonstrated the profound impact of intestinal microbes on DNA damage, DNA methylation, chromatin structure, and noncoding RNA expression in colon epithelial cells (91). Furthermore, alterations in certain genes and pathways induced by intestinal microbes are closely associated with the CRC development and influence the functionality of γδ T cells in this context. According to previous reports, certain bacteria and their metabolites, including Bacteroides fragilis, Lactobacillus acidophilus, desulfurizing Vibrio and Citrobacter, have been found to assist γδ T cells in combating tumors. Conversely, specific gut bacteria, such as Clostridia and enterotoxigenic Bacteroides fragilis, may accelerate the development of CRC by activating γδ T cells that promote tumor growth (13). Li et al (92) reported that phosphatidylethanolamine and phosphatidylcholine, metabolites of Desulfovibrio, induced the proliferation of IL-17A-producing γδ T cells, which aggravated intestinal injury. Some probiotics can protect the normal intestinal mucosa in CRC by producing short-chain fatty acids, such as acetate and propionate (93). Propionate can directly act on γδ T17 cells and inhibit IL-17 production in a histone deacetylase-dependent manner (94), whereas Akkermansia can reduce the number of IL-17-producing γδ T cells in mice (95), thereby improving intestinal inflammation. Hydroxymethyl-butyl pyrophosphate produced by microorganisms can act as a pAg to activate γδ T cells (96). A study conducted by Roselli et al (97) demonstrated that the combination of L. acidophilus and B. longum effectively impeded the progression of colitis through the modulation of the γδ T-cell population. The α-GalCer produced by Bacteroides fragilis, Bacteroides vulgatus, Prevotella copri and other unidentified bacteria can exert antitumor effects by indirectly inducing the production of IFN-γ by γδ T cells through the activation of invariant NKT cells (98) (Fig. 5). These studies suggested that the gut microbiota actively participates in the antitumor process of γδ T cells, assuming distinct roles. However, most of these studies have focused primarily on the cellular level. Consequently, whether clinical intervention targeting specific gut microbiota components can effectively decelerate tumor progression remains uncertain. In addition, several studies have demonstrated that the gut microbiota can serve as a reliable biomarker for the non-invasive diagnosis of CRC (99101). However, the selection of appropriate biomarkers and the development of highly sensitive detection methods pose limitations for its clinical application, and further research is needed to achieve breakthroughs.

The challenges and prospects of translating research on γδ T cells in CRC into clinical application

In recent years, clinical studies on the application of γδ T cells in CRC immunotherapy have focused primarily on their role in MSI-high CRC and microsatellite-stable (MSS) CRC. MSI CRCs can be categorized into MSI-H and MSI-L groups according to the level of instability, with MSI-L and MSS often grouped together in clinical studies. Given their greater mutation load and neoantigen exposure, MSI-H tumors are more readily recognized and targeted by the immune system, making conventional immune checkpoint inhibitors more effective for treating MSI-H CRC than MSS CRC (56). However, MSS tumors account for the majority of CRC cases, underscoring the pressing need for the development of innovative immunotherapies targeting patients with CRC with MSS tumors (102,103).

Recently, Stary et al (104) reported that the dysfunctional cytotoxic potential of Vδ1+ T cells can be restored by in vitro activation in MSS CRC, suggesting the possibility of reactivating these cells to exert potent antitumor effects. This discovery offers potential for the advancement of immunotherapies targeting γδ T cells in MSS CRC and holds promise for future development. Additionally, numerous studies have provided further evidence supporting the clinical investigation of γδ T cells in CRC. Stary et al (104) conducted single-cell RNA sequencing and TCR sequencing on γδ T cells from human CRC specimens and revealed that Vδ1+ cells derived from MSS CRC contribute to tumor immune evasion by upregulating exhaustion-associated genes while downregulating effector genes. Furthermore, it was discovered that this dysfunction can be reversed through modulation of the TIGIT-NECTIN axis (104). Wu et al (105) found that knocking out QPCTL in cancer cells could promote their escape from Vγ9Vδ2 T-cell elimination through genetic screening and experimental validation, suggesting that QPCTL may be a new entry point to solve CRC immune evasion. A study conducted by Xu et al (106) demonstrated the safety and efficacy of allogeneic Vγ9Vδ2 T-cell immunotherapy in prolonging the survival of patients with advanced lung or liver cancer, which could also have implications for the treatment of CRC. Additionally, ongoing investigations are exploring the utilization of bispecific T-cell engager (BiTE) technology, chimeric antigen receptor (CAR) modification and synthetic phosphorylated antigens (107110). However, the clinical application of BiTE and CAR modification technologies in CRC is hindered by off-target effects, cytokine release syndrome and neurotoxicity (111,112). The optimization of targeting technology and the development of combination drugs will significantly expedite the implementation process of these two novel therapies for CRC. Although there are significant obstacles to overcome in harnessing the potential of γδ T cells for CRC treatment, ongoing research and clinical development efforts are paving the way for potentially transformative immunotherapies that could provide new hope for patients with this prevalent cancer.

Conclusions

γδ T cells have emerged as pivotal players in the immunotherapy landscape of CRC, exerting their antitumor effects independently of MHC restrictions and exhibiting the ability to recognize and respond to tumor cells that may have evaded conventional αβ T-cell surveillance. With the discovery that Vδ2 T cells can recognize pAgs and be activated by them to exert antitumor activity, Vδ2 T cells have emerged as a prominent research focus in recent years. Numerous researchers have dedicated efforts to investigating pAgs and their associated activation pathways, enhancing our understanding of Vδ2 T cells. Several immune checkpoint genes have been identified during investigations into the mechanism of action between Vδ2 T cells and tumor cells. The combination of PD-1 and CTLA-4 inhibitors has demonstrated efficacy in treating CRC. Moreover, ongoing research is exploring additional immune checkpoint genes as promising therapeutic targets for CRC in the future. The finding that the cytotoxic potential of dysfunctional Vδ1 T cells in tumors can be reactivated in vitro holds promise for the treatment of MSS CRC unresponsive to immune checkpoint inhibitors. In addition, utilizing BiTE and CAR modification technology, along with integrating multiple approaches, significantly enhances CRC therapy efficacy. However, given the intricate regulatory network of the TME, research on the specific regulatory mechanism is lacking. In the future, combining drugs that target diverse cellular components within the TME to impede the progression of CRC may be possible. Recent studies have shown that gut microbes and their metabolites also interact with γδ T cells and tumors. However, the diversity of gut microbes and the lack of methods for sample collection, storage and analysis pose obstacles to related research. Therefore, the mode of interaction between the gut microbiota and γδ T cells in CRC and the microbial flora involved in this process remains unclear. In the future, analyses of the gut microbial species involved in the antitumor process of γδ T cells can be initiated to discover new methods for treating or diagnosing CRC.

Acknowledgements

Not applicable.

Funding

The present study was supported by the National Natural Science Foundation of China (grant no. 81972716).

Availability of data and materials

Not applicable.

Authors' contributions

XH and XC conceived and designed the study. LP, YZ and WW prepared and wrote the manuscript. YK and CW edited the manuscript. All authors read and approved the final version of the manuscript. Data authentication is not applicable.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Glossary

Abbreviations

Abbreviations:

CRC

colorectal cancer

MHC

major histocompatibility complex

TCR

T-cell receptor

TNF-α

tumor necrosis factor alpha

IFN-γ

interferon gamma

TME

tumor microenvironment

IEL

intraepithelial lymphocyte

pAg

phospho-antigen

TRAIL

tumor necrosis factor-related apoptosis-inducing ligand

IPP

isopentenyl pyrophosphate

IL-2

interleukin-2

CICs

cancer initiating stem cells

5-FU

5-fluorouracil

DXR

doxorubicin

CSCs

cancer stem cells

ADC

antibody-drug conjugates

inf-DCs

inflammatory dendritic cells

PMN-MDSCs

polymorphonuclear myeloid-derived suppressor cells

MSI

microsatellite instability

PD-1

programmed cell death protein 1

PD-L1

programmed cell death-Ligand 1

HSCs

hematopoietic stem cells

Tet1

Ten Eleven Translocation 1

NKT

natural killer T cell

Tregs

regulatory T cells

MSS

microsatellite-stable

BiTE

bispecific T-cell engager

CAR

chimeric antigen receptor

References

1 

Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA and Jemal A: Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 68:394–424. 2018. View Article : Google Scholar : PubMed/NCBI

2 

Golshani G and Zhang Y: Advances in immunotherapy for colorectal cancer: A review. Therap Adv Gastroenterol. 13:17562848209175272020. View Article : Google Scholar : PubMed/NCBI

3 

Morazan-Fernandez D, Mora J and Molina-Mora JA: In Silico pipeline to identify Tumor-specific antigens for cancer immunotherapy using exome sequencing data. Phenomics. 3:130–137. 2023. View Article : Google Scholar : PubMed/NCBI

4 

Wu X, Li T, Jiang R, Yang X, Guo H and Yang R: Targeting MHC-I molecules for cancer: Function, mechanism, and therapeutic prospects. Mol Cancer. 22:1942023. View Article : Google Scholar : PubMed/NCBI

5 

Rodrigues NV, Correia DV, Mensurado S, Nobrega-Pereira S, deBarros A, Kyle-Cezar F, Tutt A, Hayday AC, Norell H, Silva-Santos B and Dias S: Low-Density lipoprotein uptake inhibits the activation and antitumor functions of human Vgamma9Vdelta2 T cells. Cancer Immunol Res. 6:448–457. 2018. View Article : Google Scholar : PubMed/NCBI

6 

Suzuki T, Hayman L, Kilbey A, Edwards J and Coffelt SB: Gut γδ T cells as guardians, disruptors, and instigators of cancer. Immunol Rev. 298:198–217. 2020. View Article : Google Scholar : PubMed/NCBI

7 

Todaro M, Orlando V, Cicero G, Caccamo N, Meraviglia S, Stassi G and Dieli F: Chemotherapy sensitizes colon cancer initiating cells to Vγ9Vδ2 T Cell-mediated cytotoxicity. PLoS One. 8:e651452013. View Article : Google Scholar : PubMed/NCBI

8 

Lo Presti E, Pizzolato G, Gulotta E, Cocorullo G, Gulotta G, Dieli F and Meraviglia S: Current advances in γδ T Cell-based tumor immunotherapy. Front Immunol. 8:14012017. View Article : Google Scholar : PubMed/NCBI

9 

Arias-Badia M, Chang R and Fong L: γδ T cells as critical Anti-tumor immune effectors. Nat Cancer. 5:1145–1157. 2024. View Article : Google Scholar : PubMed/NCBI

10 

de Vries NL, van de Haar J, Veninga V, Chalabi M, Ijsselsteijn ME, van der Ploeg M, van den Bulk J, Ruano D, van den Berg JG, Haanen JB, et al: γδ T cells are effectors of immunotherapy in cancers with HLA class I defects. Nature. 613:743–750. 2023. View Article : Google Scholar : PubMed/NCBI

11 

Lu H, Ma Y, Wang M, Shen J, Wu H, Li J, Gao N, Gu Y, Zhang X, Zhang G, et al: B7-H3 confers resistance to Vγ9Vδ2 T cell-mediated cytotoxicity in human colon cancer cells via the STAT3/ULBP2 axis. Cancer Immunol Immunother. 70:1213–1226. 2021. View Article : Google Scholar : PubMed/NCBI

12 

Lo Presti E, Pizzolato G, Corsale AM, Caccamo N, Sireci G, Dieli F and Meraviglia S: γδ T cells and tumor microenvironment: From immunosurveillance to tumor evasion. Front Immunol. 9:13952018. View Article : Google Scholar : PubMed/NCBI

13 

Lin P, Yan Y, Zhang Z, Dong Q, Yi J, Li Q, Zhang A and Kong X: The γδ T cells dual function and crosstalk with intestinal flora in treating colorectal cancer is a promising area of study. Int Immunopharmacol. 123:1107332023. View Article : Google Scholar : PubMed/NCBI

14 

Zhu LQ, Zhang L, Zhang J, Chang GL, Liu G, Yu DD, Yu XM, Zhao MS and Ye B: Evodiamine inhibits high-fat Diet-induced Colitis-associated cancer in mice through regulating the gut microbiota. J Integr Med. 19:56–65. 2021. View Article : Google Scholar : PubMed/NCBI

15 

Rong L, Li K, Li R, Liu HM, Sun R and Liu XY: Analysis of tumor-infiltrating gamma delta T cells in rectal cancer. World J Gastroenterol. 22:3573–3580. 2016. View Article : Google Scholar : PubMed/NCBI

16 

Ma R, Yuan D, Guo Y, Yan R and Li K: Immune effects of γδ T cells in colorectal cancer: A review. Front Immunol. 11:16002020. View Article : Google Scholar : PubMed/NCBI

17 

Wu D, Wu P, Wu X, Ye J, Wang Z, Zhao S, Ni C, Hu G, Xu J, Han Y, et al: Ex vivo expanded human circulating Vδ1 γδT cells exhibit favorable therapeutic potential for colon cancer. Oncoimmunology. 4:e9927492015. View Article : Google Scholar : PubMed/NCBI

18 

Mikulak J, Oriolo F, Bruni E, Roberto A, Colombo FS, Villa A, Bosticardo M, Bortolomai I, Lo Presti E, Meraviglia S, et al: NKp46-expressing human gut-resident intraepithelial Vδ1 T cell subpopulation exhibits high antitumor activity against colorectal cancer. JCI Insight. 4:e1258842019. View Article : Google Scholar : PubMed/NCBI

19 

Bruni E, Cimino MM, Donadon M, Carriero R, Terzoli S, Piazza R, Ravens S, Prinz I, Cazzetta V, Marzano P, et al: Intrahepatic CD69+Vδ1 T cells re-circulate in the blood of patients with metastatic colorectal cancer and limit tumor progressionn. J Immunother Cancer. 10:e0045792022. View Article : Google Scholar : PubMed/NCBI

20 

Devaud C, Rousseau B, Netzer S, Pitard V, Paroissin C, Khairallah C, Costet P, Moreau JF, Couillaud F, Dechanet-Merville J and Capone M: Anti-metastatic potential of human Vδ1(+) γδ T cells in an orthotopic mouse xenograft model of colon carcinoma. Cancer Immunol Immunother. 62:1199–1210. 2013. View Article : Google Scholar : PubMed/NCBI

21 

Bernard NJ: Expanding Vδ1 T cells. Nat Immunol. 24:13962023. View Article : Google Scholar : PubMed/NCBI

22 

Lo Presti E, Mocciaro F, Mitri RD, Corsale AM, Di Simone M, Vieni S, Scibetta N, Unti E, Dieli F and Meraviglia S: Analysis of colon-infiltrating γδ T cells in chronic inflammatory bowel disease and in colitis-associated cancer. J Leukoc Biol. 108:749–760. 2020. View Article : Google Scholar : PubMed/NCBI

23 

Bouet-Toussaint F, Cabillic F, Toutirais O, Le Gallo M, Thomas de la Pintiere C, Daniel P, Genetet N, Meunier B, Dupont-Bierre E, Boudjema K, et al: Vgamma9Vdelta2 T cell-mediated recognition of human solid tumors. Potential for immunotherapy of hepatocellular and colorectal carcinomas. Cancer Immunol Immunother. 57:531–539. 2008. View Article : Google Scholar : PubMed/NCBI

24 

Iovino F, Meraviglia S, Spina M, Orlando V, Saladino V, Dieli F, Stassi G and Todaro M: Immunotherapy targeting colon cancer stem cells. Immunotherapy. 3:97–106. 2011. View Article : Google Scholar : PubMed/NCBI

25 

Adams EJ, Strop P, Shin S, Chien YH and Garcia KC: An autonomous CDR3delta is sufficient for recognition of the nonclassical MHC class I molecules T10 and T22 by gammadelta T cells. Nat Immunol. 9:777–784. 2008. View Article : Google Scholar : PubMed/NCBI

26 

Zhao H, Xi X, Cui L and He W: CDR3δ-grafted γ9δ2T cells mediate effective antitumor reactivity. Cell Mol Immunol. 9:147–154. 2012. View Article : Google Scholar : PubMed/NCBI

27 

Vyborova A, Janssen A, Gatti L, Karaiskaki F, Yonika A, van Dooremalen S, Sanders J, Beringer DX, Straetemans T, Sebestyen Z and Kuball J: γ9δ2 T-Cell expansion and phenotypic profile are reflected in the CDR3δ repertoire of healthy adults. Front Immunol. 13:9153662022. View Article : Google Scholar : PubMed/NCBI

28 

Silva-Santos B and Strid J: Working in ‘NK Mode’: Natural Killer Group 2 Member D and natural cytotoxicity receptors in Stress-surveillance by γδ T cells. Front Immunol. 9:8512018. View Article : Google Scholar : PubMed/NCBI

29 

Kong Y, Cao W, Xi X, Ma C, Cui L and He W: The NKG2D ligand ULBP4 binds to TCRgamma9/delta2 and induces cytotoxicity to tumor cells through both TCRgammadelta and NKG2D. Blood. 114:310–317. 2009. View Article : Google Scholar : PubMed/NCBI

30 

Todaro M, D'Asaro M, Caccamo N, Iovino F, Francipane MG, Meraviglia S, Orlando V, La Mendola C, Gulotta G, Salerno A, et al: Efficient killing of human colon cancer stem cells by gammadelta T lymphocytes. J Immunol. 182:7287–7296. 2009. View Article : Google Scholar : PubMed/NCBI

31 

Hoeres T, Smetak M, Pretscher D and Wilhelm M: Improving the efficiency of Vγ9Vδ2 T-Cell immunotherapy in cancer. Front Immunol. 9:8002018. View Article : Google Scholar : PubMed/NCBI

32 

Zocchi MR, Costa D, Vene R, Tosetti F, Ferrari N, Minghelli S, Benelli R, Scabini S, Romairone E, Catellani S, et al: Zoledronate can induce colorectal cancer microenvironment expressing BTN3A1 to stimulate effector γδ T cells with antitumor activity. Oncoimmunology. 6:e12780992017. View Article : Google Scholar : PubMed/NCBI

33 

Park JH and Lee HK: Function of γδ T cells in tumor immunology and their application to cancer therapy. Exp Mol Med. 53:318–327. 2021. View Article : Google Scholar : PubMed/NCBI

34 

Ramutton T, Buccheri S, Dieli F, Todaro M, Stassi G and Meraviglia S: γδ T cells as a potential tool in colon cancer immunotherapy. Immunotherapy. 6:989–999. 2014. View Article : Google Scholar : PubMed/NCBI

35 

Smyth MJ, Swann J, Kelly JM, Cretney E, Yokoyama WM, Diefenbach A, Sayers TJ and Hayakawa Y: NKG2D recognition and perforin effector function mediate effective cytokine immunotherapy of cancer. J Exp Med. 200:1325–1335. 2004. View Article : Google Scholar : PubMed/NCBI

36 

Pei Y, Xiang Z, Wen K, Tu CR, Wang X, Zhang Y, Mu X, Liu Y and Tu W: CD137 costimulation enhances the antitumor activity of Vγ9Vδ2-T cells in IL-10-Mediated immunosuppressive tumor microenvironment. Front Immunol. 13:8721222022. View Article : Google Scholar : PubMed/NCBI

37 

Zhang T, Wang J, Zhao A, Xia L, Jin H, Xia S and Shi T: The way of interaction between Vγ9Vδ2 T cells and tumor cells. Cytokine. 162:1561082023. View Article : Google Scholar : PubMed/NCBI

38 

Mattarollo SR, Kenna T, Nieda M and Nicol AJ: Chemotherapy and zoledronate sensitize solid tumour cells to Vgamma9Vdelta2 T cell cytotoxicity. Cancer Immunol Immunother. 56:1285–1297. 2007. View Article : Google Scholar : PubMed/NCBI

39 

Ma Y, Aymeric L, Locher C, Mattarollo SR, Delahaye NF, Pereira P, Boucontet L, Apetoh L, Ghiringhelli F, Casares N, et al: Contribution of IL-17-producing gamma delta T cells to the efficacy of anticancer chemotherapy. J Exp Med. 208:491–503. 2011. View Article : Google Scholar : PubMed/NCBI

40 

Jinushi M, Vanneman M, Munshi NC, Tai YT, Prabhala RH, Ritz J, Neuberg D, Anderson KC, Carrasco DR and Dranoff G: MHC class I chain-related protein A antibodies and shedding are associated with the progression of multiple myeloma. Proc Natl Acad Sci USA. 105:1285–1290. 2008. View Article : Google Scholar : PubMed/NCBI

41 

Vales-Gomez M, Chisholm SE, Cassady-Cain RL, Roda-Navarro P and Reyburn HT: Selective induction of expression of a ligand for the NKG2D receptor by proteasome inhibitors. Cancer Res. 68:1546–1554. 2008. View Article : Google Scholar : PubMed/NCBI

42 

Diermayr S, Himmelreich H, Durovic B, Mathys-Schneeberger A, Siegler U, Langenkamp U, Hofsteenge J, Gratwohl A, Tichelli A, Paluszewska M, et al: NKG2D ligand expression in AML increases in response to HDAC inhibitor valproic acid and contributes to allorecognition by NK-cell lines with single KIR-HLA class I specificities. Blood. 111:1428–1436. 2008. View Article : Google Scholar : PubMed/NCBI

43 

Armeanu S, Bitzer M, Lauer UM, Venturelli S, Pathil A, Krusch M, Kaiser S, Jobst J, Smirnow I, Wagner A, et al: Natural killer Cell-mediated lysis of hepatoma cells via specific induction of NKG2D ligands by the histone deacetylase inhibitor sodium valproate. Cancer Res. 65:6321–6329. 2005. View Article : Google Scholar : PubMed/NCBI

44 

Jones AB, Rocco A, Lamb LS, Friedman GK and Hjelmeland AB: Regulation of NKG2D stress ligands and its relevance in cancer progression. Cancers (Basel). 14:23392022. View Article : Google Scholar : PubMed/NCBI

45 

Benelli R, Costa D, Salvini L, Tardito S, Tosetti F, Villa F, Zocchi MR and Poggi A: Targeting of colorectal cancer organoids with zoledronic acid conjugated to the anti-EGFR antibody cetuximab. J Immunother Cancer. 10:e0056602022. View Article : Google Scholar : PubMed/NCBI

46 

Wu P, Wu D, Ni C, Ye J, Chen W, Hu G, Wang Z, Wang C, Zhang Z, Xia W, et al: gammadeltaT17 cells promote the accumulation and expansion of myeloid-derived suppressor cells in human colorectal cancer. Immunity. 40:785–800. 2014. View Article : Google Scholar : PubMed/NCBI

47 

Corsale AM, Di Simone M, Lo Presti E, Dieli F and Meraviglia S: γδ T cells and their clinical application in colon cancer. Front Immunol. 14:10988472023. View Article : Google Scholar : PubMed/NCBI

48 

Grivennikov SI, Wang K, Mucida D, Stewart CA, Schnabl B, Jauch D, Taniguchi K, Yu GY, Osterreicher CH, Hung KE, et al: Adenoma-linked barrier defects and microbial products drive IL-23/IL-17-mediated tumour growth. Nature. 491:254–258. 2012. View Article : Google Scholar : PubMed/NCBI

49 

Lee JS, Tato CM, Joyce-Shaikh B, Gulen MF, Cayatte C, Chen Y, Blumenschein WM, Judo M, Ayanoglu G, McClanahan TK, et al: Interleukin-23-Independent IL-17 production regulates intestinal epithelial permeability. Immunity. 43:727–738. 2015. View Article : Google Scholar : PubMed/NCBI

50 

Reis BS, Darcy PW, Khan IZ, Moon CS, Kornberg AE, Schneider VS, Alvarez Y, Eleso O, Zhu C, Schernthanner M, et al: TCR-Vγδ usage distinguishes protumor from antitumor intestinal γδ T cell subsets. Science. 377:276–284. 2022. View Article : Google Scholar : PubMed/NCBI

51 

Mu X, Xiang Z, Xu Y, He J, Lu J, Chen Y, Wang X, Tu CR, Zhang Y, Zhang W, et al: Glucose metabolism controls human γδ T-cell-mediated tumor immunosurveillance in diabetes. Cell Mol Immunol. 19:944–956. 2022. View Article : Google Scholar : PubMed/NCBI

52 

Agerholm R and Bekiaris V: Evolved to protect, designed to destroy: IL-17-producing γδ T cells in infection, inflammation, and cancer. Eur J Immunol. 51:2164–2177. 2021. View Article : Google Scholar : PubMed/NCBI

53 

Lopes N, McIntyre C, Martin S, Raverdeau M, Sumaria N, Kohlgruber AC, Fiala GJ, Agudelo LZ, Dyck L, Kane H, et al: Distinct metabolic programs established in the thymus control effector functions of γδ T cell subsets in tumor microenvironments. Nat Immunol. 22:179–192. 2021. View Article : Google Scholar : PubMed/NCBI

54 

Mensurado S and Silva-Santos B: Battle of the γδ T cell subsets in the gut. Trends Cancer. 8:881–883. 2022. View Article : Google Scholar : PubMed/NCBI

55 

Silva-Santos B, Mensurado S and Coffelt SB: γδ T cells: Pleiotropic immune effectors with therapeutic potential in cancer. Nat Rev Cancer. 19:392–404. 2019. View Article : Google Scholar : PubMed/NCBI

56 

Cai L, Chen A and Tang D: A new strategy for immunotherapy of Microsatellite-stable (MSS)-type advanced colorectal cancer: Multi-pathway combination therapy with PD-1/PD-L1 inhibitors. Immunology. Mar 22–2024.doi: 10.1111/imm.13785 (Epub ahead of print). View Article : Google Scholar

57 

Han Y, Liu D and Li L: PD-1/PD-L1 pathway: Current researches in cancer. Am J Cancer Res. 10:727–742. 2020.PubMed/NCBI

58 

Postow MA, Sidlow R and Hellmann MD: Immune-related adverse events associated with immune checkpoint blockade. N Engl J Med. 378:158–168. 2018. View Article : Google Scholar : PubMed/NCBI

59 

Reck M, Rodriguez-Abreu D, Robinson AG, Hui R, Csoszi T, Fulop A, Gottfried M, Peled N, Tafreshi A, Cuffe S, et al: Pembrolizumab versus chemotherapy for PD-L1-positive Non-Small-Cell lung cancer. N Engl J Med. 375:1823–1833. 2016. View Article : Google Scholar : PubMed/NCBI

60 

Robert C, Long GV, Brady B, Dutriaux C, Maio M, Mortier L, Hassel JC, Rutkowski P, McNeil C, Kalinka-Warzocha E, et al: Nivolumab in previously untreated melanoma without BRAF mutation. N Engl J Med. 372:320–330. 2015. View Article : Google Scholar : PubMed/NCBI

61 

Tie G, Messina KE, Yan J, Messina JA and Messina LM: Hypercholesterolemia induces oxidant stress that accelerates the ageing of hematopoietic stem cells. J Am Heart Assoc. 3:e0002412014. View Article : Google Scholar : PubMed/NCBI

62 

Tie G, Yan J, Khair L, Messina JA, Deng A, Kang J, Fazzio T and Messina LM: Hypercholesterolemia increases colorectal cancer incidence by reducing production of NKT and γδ T cells from hematopoietic stem cells. Cancer Res. 77:2351–2362. 2017. View Article : Google Scholar : PubMed/NCBI

63 

Lu H, Shi T, Wang M, Li X, Gu Y, Zhang X, Zhang G and Chen W: B7-H3 inhibits the IFN-γ-dependent cytotoxicity of Vγ9Vδ2 T cells against colon cancer cells. Oncoimmunology. 9:17489912020. View Article : Google Scholar : PubMed/NCBI

64 

Bas A, Swamy M, Abeler-Dorner L, Williams G, Pang DJ, Barbee SD and Hayday AC: Butyrophilin-like 1 encodes an enterocyte protein that selectively regulates functional interactions with T lymphocytes. Proc Natl Acad Sci USA. 108:4376–4381. 2011. View Article : Google Scholar : PubMed/NCBI

65 

Di Marco Barros R, Roberts NA, Dart RJ, Vantourout P, Jandke A, Nussbaumer O, Deban L, Cipolat S, Hart R, Iannitto ML, et al: Epithelia use Butyrophilin-like molecules to shape organ-Specific γδ T cell compartments. Cell. 167:203–218.e17. 2016. View Article : Google Scholar : PubMed/NCBI

66 

Du Y, Peng Q, Cheng D, Pan T, Sun W, Wang H, Ma X, He R, Zhang H, Cui Z, et al: Cancer Cell-expressed BTNL2 facilitates tumour immune escape via engagement with IL-17A-producing γδ T cells. Nat Commun. 13:2312022. View Article : Google Scholar : PubMed/NCBI

67 

Harly C, Guillaume Y, Nedellec S, Peigne CM, Monkkonen H, Monkkonen J, Li J, Kuball J, Adams EJ, Netzer S, et al: Key implication of CD277/butyrophilin-3 (BTN3A) in cellular stress sensing by a major human γδ T-cell subset. Blood. 120:2269–2279. 2012. View Article : Google Scholar : PubMed/NCBI

68 

Chen S, Li Z, Huang W, Wang Y and Fan S: Prognostic and therapeutic significance of BTN3A proteins in tumors. J Cancer. 12:4505–4512. 2021. View Article : Google Scholar : PubMed/NCBI

69 

Palakodeti A, Sandstrom A, Sundaresan L, Harly C, Nedellec S, Olive D, Scotet E, Bonneville M and Adams EJ: The molecular basis for modulation of human Vγ9Vδ2 T cell responses by CD277/butyrophilin-3 (BTN3A)-specific antibodies. J Biol Chem. 287:32780–32790. 2012. View Article : Google Scholar : PubMed/NCBI

70 

Cano CE, Pasero C, De Gassart A, Kerneur C, Gabriac M, Fullana M, Granarolo E, Hoet R, Scotet E, Rafia C, et al: BTN2A1, an immune checkpoint targeting Vγ9Vδ2 T cell cytotoxicity against malignant cells. Cell Rep. 36:1093592021. View Article : Google Scholar : PubMed/NCBI

71 

De Gassart A, Le KS, Brune P, Agaugue S, Sims J, Goubard A, Castellano R, Joalland N, Scotet E, Collette Y, et al: Development of ICT01, a first-in-class, anti-BTN3A antibody for activating Vγ9Vδ2 T cell-mediated antitumor immune response. Sci Transl Med. 13:eabj08352021. View Article : Google Scholar : PubMed/NCBI

72 

Blazquez JL, Benyamine A, Pasero C and Olive D: New insights into the regulation of γδ T cells by BTN3A and Other BTN/BTNL in tumor immunity. Front Immunol. 9:16012018. View Article : Google Scholar : PubMed/NCBI

73 

Seiwert N, Adam J, Steinberg P, Wirtz S, Schwerdtle T, Adams-Quack P, Hovelmeyer N, Kaina B, Foersch S and Fahrer J: Chronic intestinal inflammation drives colorectal tumor formation triggered by dietary heme iron in vivo. Arch Toxicol. 95:2507–2522. 2021. View Article : Google Scholar : PubMed/NCBI

74 

Santiago L, Castro M, Sanz-Pamplona R, Garzon M, Ramirez-Labrada A, Tapia E, Moreno V, Layunta E, Gil-Gomez G, Garrido M, et al: Extracellular granzyme A promotes colorectal cancer development by enhancing gut inflammation. Cell Rep. 32:1078472020. View Article : Google Scholar : PubMed/NCBI

75 

Lebrero-Fernandez C, Wenzel UA, Akeus P, Wang Y, Strid H, Simren M, Gustavsson B, Borjesson LG, Cardell SL, Ohman L, et al: Altered expression of Butyrophilin (BTN) and BTN-like (BTNL) genes in intestinal inflammation and colon cancer. Immun Inflamm Dis. 4:191–200. 2016. View Article : Google Scholar : PubMed/NCBI

76 

Liu J, Wu M, Yang Y, Wang Z, He S, Tian X and Wang H: γδ T cells and the PD-1/PD-L1 axis: A love-hate relationship in the tumor microenvironment. J Transl Med. 22:5532024. View Article : Google Scholar : PubMed/NCBI

77 

Wu K, Feng J, Xiu Y, Li Z, Lin Z, Zhao H, Zeng H, Xia W, Yu L and Xu B: Vδ2 T cell subsets, defined by PD-1 and TIM-3 expression, present varied cytokine responses in acute myeloid leukemia patients. Int Immunopharmacol. 80:1061222020. View Article : Google Scholar : PubMed/NCBI

78 

Pan T, Yang H, Wang WY, Rui YY, Deng ZJ, Chen YC, Liu C and Hu H: Neoadjuvant immunotherapy with ipilimumab plus nivolumab in mismatch repair Deficient/Microsatellite Instability-High colorectal cancer: A preliminary report of case series. Clin Colorectal Cancer. 23:104–110. 2024. View Article : Google Scholar : PubMed/NCBI

79 

Li X, Lu H, Gu Y, Zhang X, Zhang G, Shi T and Chen W: Tim-3 suppresses the killing effect of Vγ9Vδ2 T cells on colon cancer cells by reducing perforin and granzyme B expression. Exp Cell Res. 386:1117192020. View Article : Google Scholar : PubMed/NCBI

80 

Guo C, Dai X, Du Y, Xiong X and Gui X: Preclinical development of a novel CCR8/CTLA-4 bispecific antibody for cancer treatment by disrupting CTLA-4 signaling on CD8 T cells and specifically depleting tumor-resident Tregs. Cancer Immunol Immunother. 73:2102024. View Article : Google Scholar : PubMed/NCBI

81 

Aggarwal V, Workman CJ and Vignali DAA: LAG-3 as the third checkpoint inhibitor. Nat Immunol. 24:1415–1422. 2023. View Article : Google Scholar : PubMed/NCBI

82 

Perales O, Jilaveanu L, Adeniran A, Su DG, Hurwitz M, Braun DA, Kluger HM and Schoenfeld DA: TIGIT expression in renal cell carcinoma infiltrating T cells is variable and inversely correlated with PD-1 and LAG3. Cancer Immunol Immunother. 73:1922024. View Article : Google Scholar : PubMed/NCBI

83 

Bhat AA, Nisar S, Singh M, Ashraf B, Masoodi T, Prasad CP, Sharma A, Maacha S, Karedath T, Hashem S, et al: Cytokine- and chemokine-induced inflammatory colorectal tumor microenvironment: Emerging avenue for targeted therapy. Cancer Commun (Lond). 42:689–715. 2022. View Article : Google Scholar : PubMed/NCBI

84 

Shu Y and Zheng S: The current status and prospect of immunotherapy in colorectal cancer. Clin Transl Oncol. 26:39–51. 2024. View Article : Google Scholar : PubMed/NCBI

85 

Chen DS and Mellman I: Elements of cancer immunity and the Cancer-immune set point. Nature. 541:321–330. 2017. View Article : Google Scholar : PubMed/NCBI

86 

Yi Y, He HW, Wang JX, Cai XY, Li YW, Zhou J, Cheng YF, Jin JJ, Fan J and Qiu SJ: The functional impairment of HCC-infiltrating γδ T cells, partially mediated by regulatory T cells in a TGFβ- and IL-10-dependent manner. J Hepatol. 58:977–983. 2013. View Article : Google Scholar : PubMed/NCBI

87 

Hu G, Wu P, Cheng P, Zhang Z, Wang Z, Yu X, Shao X, Wu D, Ye J, Zhang T, et al: Tumor-infiltrating CD39+γδ Tregs are novel immunosuppressive T cells in human colorectal cancer. Oncoimmunology. 6:e12773052017. View Article : Google Scholar : PubMed/NCBI

88 

Zhan Y, Zheng L, Liu J, Hu D, Wang J, Liu K, Guo J, Zhang T and Kong D: PLA2G4A promotes Right-sided colorectal cancer progression by inducing CD39+γδ Treg polarization. JCI Insight. 6:e1480282021. View Article : Google Scholar : PubMed/NCBI

89 

Chen Z, Han F, Du Y, Shi H and Zhou W: Hypoxic microenvironment in cancer: Molecular mechanisms and therapeutic interventions. Signal Transduct Target Ther. 8:702023. View Article : Google Scholar : PubMed/NCBI

90 

Li L, Cao B, Liang X, Lu S, Luo H, Wang Z, Wang S, Jiang J, Lang J and Zhu G: Microenvironmental oxygen pressure orchestrates an anti- and pro-tumoral γδ T cell equilibrium via tumor-derived exosomes. Oncogene. 38:2830–2843. 2019. View Article : Google Scholar : PubMed/NCBI

91 

Allen J and Sears CL: Impact of the gut microbiome on the genome and epigenome of colon epithelial cells: Contributions to colorectal cancer development. Genome Med. 11:112019. View Article : Google Scholar : PubMed/NCBI

92 

Li Y, Wang Y, Shi F, Zhang X, Zhang Y, Bi K, Chen X, Li L and Diao H: Phospholipid metabolites of the gut microbiota promote hypoxia-induced intestinal injury via CD1d-dependent γδ T cells. Gut Microbes. 14:20969942022. View Article : Google Scholar : PubMed/NCBI

93 

Casanova MR, Azevedo-Silva J, Rodrigues LR and Preto A: Colorectal cancer cells increase the production of short chain fatty acids by propionibacterium freudenreichii impacting on cancer cells survival. Front Nutr. 5:442018. View Article : Google Scholar : PubMed/NCBI

94 

Dupraz L, Magniez A, Rolhion N, Richard ML, Da Costa G, Touch S, Mayeur C, Planchais J, Agus A, Danne C, et al: Gut microbiota-derived short-chain fatty acids regulate IL-17 production by mouse and human intestinal γδ T cells. Cell Rep. 36:1093322021. View Article : Google Scholar : PubMed/NCBI

95 

Cox LM, Maghzi AH, Liu S, Tankou SK, Dhang FH, Willocq V, Song A, Wasen C, Tauhid S, Chu R, et al: Gut microbiome in progressive multiple sclerosis. Ann Neurol. 89:1195–1211. 2021. View Article : Google Scholar : PubMed/NCBI

96 

Sandstrom A, Peigne CM, Leger A, Crooks JE, Konczak F, Gesnel MC, Breathnach R, Bonneville M, Scotet E and Adams EJ: The intracellular B30.2 domain of butyrophilin 3A1 binds phosphoantigens to mediate activation of human Vγ9Vδ2 T cells. Immunity. 40:490–500. 2014. View Article : Google Scholar : PubMed/NCBI

97 

Roselli M, Finamore A, Nuccitelli S, Carnevali P, Brigidi P, Vitali B, Nobili F, Rami R, Garaguso I and Mengheri E: Prevention of TNBS-induced colitis by different Lactobacillus and Bifidobacterium strains is associated with an expansion of gammadeltaT and regulatory T cells of intestinal intraepithelial lymphocytes. Inflamm Bowel Dis. 15:1526–1536. 2009. View Article : Google Scholar : PubMed/NCBI

98 

Ustjanzew A, Sencio V, Trottein F, Faber J, Sandhoff R and Paret C: Interaction between bacteria and the immune system for cancer immunotherapy: The α-GalCer alliance. Int J Mol Sci. 23:58962022. View Article : Google Scholar : PubMed/NCBI

99 

Baxter NT, Ruffin MT, Rogers MAM and Schloss PD: Microbiota-based model improves the sensitivity of fecal immunochemical test for detecting colonic lesions. Genome Medicine. 8:372016. View Article : Google Scholar : PubMed/NCBI

100 

Liang Q, Chiu J, Chen Y, Huang Y, Higashimori A, Fang J, Brim H, Ashktorab H, Ng SC, Ng SSM, et al: Fecal bacteria act as novel biomarkers for noninvasive diagnosis of colorectal cancer. Clin Cancer Res. 23:2061–2070. 2017. View Article : Google Scholar : PubMed/NCBI

101 

Yu J, Feng Q, Wong SH, Zhang D, Liang QY, Qin Y, Tang L, Zhao H, Stenvang J, Li Y, et al: Metagenomic analysis of faecal microbiome as a tool towards targeted non-invasive biomarkers for colorectal cancer. Gut. 66:70–78. 2017. View Article : Google Scholar : PubMed/NCBI

102 

Ooki A, Shinozaki E and Yamaguchi K: Immunotherapy in colorectal cancer: Current and future strategies. J Anus Rectum Colon. 5:11–24. 2021. View Article : Google Scholar : PubMed/NCBI

103 

Heregger R, Huemer F, Steiner M, Gonzalez-Martinez A, Greil R and Weiss L: Unraveling resistance to immunotherapy in MSI-High colorectal cancer. Cancers (Basel). 15:50902023. View Article : Google Scholar : PubMed/NCBI

104 

Stary V, Pandey RV, List J, Kleissl L, Deckert F, Kabiljo J, Laengle J, Gerakopoulos V, Oehler R, Watzke L, et al: Dysfunctional tumor-infiltrating Vδ1 + T lymphocytes in microsatellite-stable colorectal cancer. Nat Commun. 15:69492024. View Article : Google Scholar : PubMed/NCBI

105 

Wu Z, Lamao Q, Gu M, Jin X, Liu Y, Tian F, Yu Y, Yuan P, Gao S, Fulford TS, et al: Unsynchronized butyrophilin molecules dictate cancer cell evasion of Vγ9Vδ2 T-cell killing. Cell Mol Immunol. 21:362–373. 2024. View Article : Google Scholar : PubMed/NCBI

106 

Xu Y, Xiang Z, Alnaggar M, Kouakanou L, Li J, He J, Yang J, Hu Y, Chen Y, Lin L, et al: Allogeneic Vγ9Vδ2 T-cell immunotherapy exhibits promising clinical safety and prolongs the survival of patients with late-stage lung or liver cancer. Cell Mol Immunol. 18:427–439. 2021. View Article : Google Scholar : PubMed/NCBI

107 

Yang XM, Lin XD, Shi W, Xie SX, Huang XN, Yin SH, Jiang XB, Hammock BD, Xu ZP and Lu XL: Nanobody-based bispecific T-cell engager (Nb-BiTE): A new platform for enhanced T-cell immunotherapy. Signal Transduct Target Ther. 8:3282023. View Article : Google Scholar : PubMed/NCBI

108 

Magee MS, Abraham TS, Baybutt TR, Flickinger JC Jr, Ridge NA, Marszalowicz GP, Prajapati P, Hersperger AR, Waldman SA and Snook AE: Human GUCY2C-targeted chimeric antigen receptor (CAR)-expressing T cells eliminate colorectal cancer metastases. Cancer Immunol Res. 6:509–516. 2018. View Article : Google Scholar : PubMed/NCBI

109 

Li M, Li S, Zhao R, Lv J, Zheng D, Qin L, Li S, Wu Q, Long Y, Tang Z, et al: CD318 is a target of chimeric antigen receptor T cells for the treatment of colorectal cancer. Clin Exp Med. 23:2409–2419. 2023. View Article : Google Scholar : PubMed/NCBI

110 

Nicol AJ, Tokuyama H, Mattarollo SR, Hagi T, Suzuki K, Yokokawa K and Nieda M: Clinical evaluation of autologous gamma delta T cell-based immunotherapy for metastatic solid tumours. Br J Cancer. 105:778–786. 2011. View Article : Google Scholar : PubMed/NCBI

111 

Kamrani A, Nasiri H, Hassanzadeh A, Ahmadian Heris J, Mohammadinasab R, Sadeghvand S, Sadeghi M, Valedkarimi Z, Hosseinzadeh R, Shomali N, et al: New immunotherapy approaches for colorectal cancer: Focusing on CAR-T cell, BiTE, and oncolytic viruses. Cell Commun Signal. 22:562024. View Article : Google Scholar : PubMed/NCBI

112 

Van De Vyver AJ, Marrer-Berger E, Wang K, Lehr T and Walz AC: Cytokine release syndrome by T-cell-Redirecting therapies: Can we predict and modulate patient risk? Clin Cancer Res. 27:6083–6094. 2021. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

December-2024
Volume 52 Issue 6

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Pan L, Zhou Y, Kuang Y, Wang C, Wang W, Hu X and Chen X: Progress of research on &gamma;&delta; T cells in colorectal cancer (Review). Oncol Rep 52: 160, 2024.
APA
Pan, L., Zhou, Y., Kuang, Y., Wang, C., Wang, W., Hu, X., & Chen, X. (2024). Progress of research on &gamma;&delta; T cells in colorectal cancer (Review). Oncology Reports, 52, 160. https://doi.org/10.3892/or.2024.8819
MLA
Pan, L., Zhou, Y., Kuang, Y., Wang, C., Wang, W., Hu, X., Chen, X."Progress of research on &gamma;&delta; T cells in colorectal cancer (Review)". Oncology Reports 52.6 (2024): 160.
Chicago
Pan, L., Zhou, Y., Kuang, Y., Wang, C., Wang, W., Hu, X., Chen, X."Progress of research on &gamma;&delta; T cells in colorectal cancer (Review)". Oncology Reports 52, no. 6 (2024): 160. https://doi.org/10.3892/or.2024.8819