Next Article in Journal
Oxidative Stress and Oocyte Cryopreservation: Recent Advances in Mitigation Strategies Involving Antioxidants
Previous Article in Journal
Circular RNA hsa_circ_0051040 Promotes Hepatocellular Carcinoma Progression by Sponging miR-569 and Regulating ITGAV Expression
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Human Vδ2 T Cells and Their Versatility for Immunotherapeutic Approaches

Department of Microbiology Immunology and Tropical Medicine, George Washington University, Washington, DC 20052, USA
*
Author to whom correspondence should be addressed.
Cells 2022, 11(22), 3572; https://doi.org/10.3390/cells11223572
Submission received: 12 October 2022 / Revised: 6 November 2022 / Accepted: 9 November 2022 / Published: 11 November 2022
(This article belongs to the Section Cell and Gene Therapy)

Abstract

:
Gamma/delta (γδ) T cells are innate-like immune effectors that are a critical component linking innate and adaptive immune responses. They are recognized for their contribution to tumor surveillance and fight against infectious diseases. γδ T cells are excellent candidates for cellular immunotherapy due to their unique properties to recognize and destroy tumors or infected cells. They do not depend on the recognition of a single antigen but rather a broad-spectrum of diverse ligands through expression of various cytotoxic receptors. In this manuscript, we review major characteristics of the most abundant circulating γδ subpopulation, Vδ2 T cells, their immunotherapeutic potential, recent advances in expansion protocols, their preclinical and clinical applications for several infectious diseases and malignancies, and how additional modulation could enhance their therapeutic potential.

1. Background

γδ T cells are innate-like immune effectors with a critical role linking innate and adaptive immune responses owing to their unique properties [1,2]. Their defining feature is a T-cell receptor (TCR) comprised of variable γ and δ chains that recognizes non-peptide antigens in the absence of Major Histocompatibility Complex (MHC) molecules and elicit non-redundant functions when compared to conventional αβ T cells [1,2]. Human γδ T cells account for 0.5–10% of circulating T cells and are classified into two major subpopulations based on the δ chain usage, Vδ1 and Vδ2 T cells. The Vδ2 chain is present in the majority of circulating γδ T cells and is almost always paired with the Vγ9 chain (Vγ9Vδ2 T cells) [3,4].
γδ T cells are further characterized by membrane receptors and cytokine production capability that denotes a high degree of polyfunctionality as regulatory and effector cells [1]. γδ T cells shape the total immune response by secreting regulatory cytokines and interacting directly with other immune cell populations [5]. Each effector class plays a direct role in immune regulation and surveillance by modulating CD4+ T, CD8+ T, B, and dendritic cells (DCs) according to the challenge and the subset of γδ T cells involved in the response [6,7]. γδ T cells are also potent cytotoxic effectors, executing direct cytolytic activity mediated by the release of perforin, granzymes and Interferon- γ (IFN-γ) [1,8,9,10]. γδ T cell-mediated induction of apoptosis may occur through the expression of tumor necrosis factor receptor superfamily members TNF- related apoptosis-inducing ligand (TRAIL) or FasL, both of which have been demonstrated to be important for mediating the resolution of inflammation and clearing tumor cells [11,12]. In addition, expression of CD16 (FcγRIII) confers an additional mechanism for mediating antibody dependent cellular cytotoxicity (ADCC) [11,12,13,14].
Due to their unique biology, i.e., antigen recognition and effector functions, and our capacity to specifically induce their activation, γδ T cells are excellent candidates for cellular immunotherapy. Recognition and killing of tumors may not depend on the expression of a single antigen but diverse broad-spectrum ligands in cancer cells thanks to the expression of various cytotoxicity receptors [15]. Although the specific activating ligand for Vδ1 T cells remains elusive, an approved protocol for the clinical expansion of Vδ1 T cells developed by Silva-Santo’s group has provided new insights into the treatment of solid tumors and hematological malignancies [15,16]. A better understanding of Vδ2 T cell immunobiology has propelled most of the existing clinical trials. However, in the absence of a clinical protocol to expand Vδ2 cells ex vivo most clinical approaches have relied on the combined use of amino-bisphosphonates (N-BPs) and IL-2 or IL-15 as the preferred method to produce enough numbers (i.e., >90% Vδ2 T cells of total culture) for clinical application and further discussed below [8]. The potential to generate “super-killer” effector cells directed towards enhanced recognition and clearance is starting to be more widely investigated [17,18,19].
In this manuscript, we review major characteristics of Vδ2 T cells, their immunotherapeutic use for different adoptive cell therapy clinical applications in the context of malignancies and infectious diseases, discuss recent advances in expansion protocols and how additional modulation of Vδ2 T cell inherent properties could enhance their therapeutic potential.

2. Phosphoantigens for Ex Vivo Vδ2 T Cell Activation

The Vδ2 TCR specifically recognizes small “self” and “non-self” pyrophosphate antigens generally called phosphoantigens (p-Ags) [20,21,22]. “Non-self” p-Ags are microbial metabolites derived from the non-mevalonate or methyl-erythritol (MEP) pathway, including (E)-4-Hydroxy-3-methyl-but-2-enyl pyrophosphate (HMBPP) that triggers the most potent Vδ2 T cell activation [22,23]. Several laboratories have synthesized HMBPP with similar effects than the naturally derived HMBPP on Vδ2 T cells [24,25,26]. The use of HMBPP in combination with IL-2 leads to Vδ2 T cell proliferation and up-regulation of markers like CD16, CD25, or CD69 and secretion of pro-inflammatory cytokines [27,28]. Further combination of synthetic HMBPP in the presence of IL-21 enhances the antitumor cytolytic activity of Vδ2 T cells [23,29,30]. A second, less potent activator, isopentenyl pyrophosphate (IPP) is a prenyl pyrophosphate common to both the MEP and the Mevalonate (MVA) pathway that accumulates during periods of cellular stress [31,32].
The MVA pathway is an essential metabolic pathway common to all eukaryotic cells with critical roles in multiple cellular processes including synthesis of sterol derivatives such as cholesterol and non-sterol isoprenoids such as dolichol or heme-A [33]. Knowledge of the MVA pathway led to the development of several drugs such as statins and N-BPs that target different steps of the pathway [22,34,35,36,37,38] (Figure 1). Statins are widely used for the treatment of hypercholesterolemia, and they function by inhibiting the HMG-CoA reductase, which is the rate-limiting enzyme in the MVA pathway [39,40]. On the contrary, N-BPs are potent inhibitors of the synthesis of farnesyl pyrophosphate (FPP) and function by inhibiting the enzyme farnesyl pyrophosphate synthase [41]. N-BPs are the first line of treatment for osteoporosis and bone-related metastases [8,42]. These are small molecules with similar structure to prenyl pyrophosphates [43]. The use of N-BPs such as pamidronate (PAM), alendronate (ALN) or zoledronate (ZOL) has a dual effect, inducing accumulation of the natural antigen for Vδ2 T cells, IPP, and disrupting the production of FPP groups used for downstream processes such as protein prenylation. In the tumor setting, this inhibition provokes the disruption of important signal transduction pathways that regulate the proliferation, invasive properties, and pro-angiogenic activity of human tumor cells [44,45,46].
Although the specific mode of p-Ag presentation and recognition is not completely understood, the discovery of the involvement of transmembrane butyrophilin (BTN) molecules constituted an incredible breakthrough and exponentially increased our potential to exploit their intrinsic properties [47,48,49,50,51]. BTNs belong to the immunoglobulin superfamily of proteins with shared homology to B7 co-stimulatory molecules. Humans express seven distinct BTNs encoded by: BTN1A1, BTN2A1, BTN2A2, BTN2A3, BTN3A1, BTN3A2, and BTN3A3 as well as five additional butyrophilin-like (BTNL) proteins: BTNL2, BTNL3, BTNL8, BTNL9 and BTNL10 [52]. Each protein includes extracellular IgV and IgC domains and, with the exception of BTNL2 and BTN3A2, cytoplasmic B30.2 domains that are critical for intracellular signaling [49,53]. Specifically, the B30.2 domain on BTN3A1 binds to p-Ag leading to conformational changes that allow recognition by the Vγ9Vδ2 TCR and subsequent activation of Vδ2 cells [54,55]. Rigau M. et al., recently identified that, in addition to BTN3A1, BTN2A1 plays an important role as a ligand that binds to the Vγ9 TCR γ chain [48]. It is believed that once p-Ag binds to BTN3A1 through its intracellular domain, the BTN2A1–BTN3A1 complex engages the γδ TCR via two distinct binding sites: BTN2A1 binding to Vγ9 region, whereas another ligand (possibly BTN3A1) binding to the Vδ2 chain [48]. The use of monoclonal antibodies against BTN3 (CD277) can directly stimulate Vδ2 T cell responses and can be used to expand Vδ2 T cells both in vitro and in vivo [56,57]. The anti-BTN3 clone 20.1 specifically binds to an epitope within the extracellular IgV domain which generates or possibly stabilizes an “activating” conformation of dimeric BTN3A1. Additional agonistic antibodies such as ICT101 have been shown to promote Vδ2 T cell recognition of target cells, including through p-Ag independent mechanisms that require interaction between BTN3A and BTN2A isoforms [47,58]. The MHC-unrestricted nature of antigen recognition represents an advantage to develop allogeneic immunotherapies using Vδ2 T cells. However, there are also challenges associated with such approaches including culture conditions and interindividual variability.
A previous study from our group showed that optimal in vitro expansion of Vδ2 T cells was achieved in the presence of MHC-II+ cells, and more specifically in cocultures with DCs [59]. A more recent study showed that depletion of αβ TCR+ cells during a two-step expansion protocol that included coculture with a genetically modified K562 cell line, induced an average fold expansion of around 230,000-fold [60].
An additional challenge for developing an adoptive cell therapy, includes interindividual Vδ2 subpopulation heterogeneity that may possibly require stratifying donors in terms of biological sex, age, ethnicity or even level of exercise to develop an optimal protocol that will provide sufficient numbers of cytotoxic Vδ2 T cells [61,62,63,64]. Several studies have investigated the variability of in vitro expansion rates among healthy individuals. Ryan PL. et al., elegantly showed a heterogeneous composition of proliferative versus cytotoxic Vδ2 functional subpopulations that was variable among healthy individuals [6]. In a follow up study, our group corroborated these findings in people living with HIV [64]. Interestingly, donor variability may be influenced by donor lifestyle, where exercise prior to PBMCs isolation has been shown to increase ex vivo expansion of γδ T cells [63]. In addition, the success of an expansion cannot be predicted based upon initial γδ T cell percentages [65], highlighting the complexity of finding optimal donors for expansion.

3. Clinical Applications of Vδ2 T Cell Immunotherapy

3.1. Immunotherapy for Infectious Diseases

Compared to oncology trials, discussed below, there is a dearth of clinical data for the therapeutic use of Vδ2 T cells to treat infectious diseases. To date, only a single pilot study has been published in which people living with HIV who had not yet started antiretroviral therapy (ART) received a short-course of ZOL and IL-2 [66]. The authors observed partial restoration in Vδ2 T cell functionality, including potential adjuvant effects boosting dendritic cell maturation and HIV-specific CD8 T cell responses. Data taken from preclinical animal models suggests that Vδ2 T cell immunotherapy may be unable to control early HIV replication despite possessing potent anti-HIV capabilities [67,68,69]. In contrast to acute infection, treating chronic SIV-infected Chinese rhesus macaques with HMBPP and IL-2 resulted in robust Vδ2 T cell expansion and pro-inflammatory cytokine production, which boosted both anti-HIV cellular and humoral responses [70]. Finally, our group has provided a proof-of-concept that Vδ2 T cells may be incorporated into current strategies for HIV cure within virally suppressed people living with HIV [71,72]. One approach utilized in HIV cure strategies encompasses reactivation of latent virus using small compounds generally called latency reversing agents and boosting immune responses [73,74,75,76,77,78]. We demonstrated that autologous expanded Vδ2 T cells mediated killing of latently HIV-infected CD4+ T cell reservoirs upon reactivation. Additional work from our lab showed that the frequency of cytotoxic Vδ2 + CD16+ cells inversely correlated with the recovery of replication-competent HIV from CD4+ T cell reservoirs [72]. This work corroborated the identification of proliferative versus cytotoxic Vδ2 T cell profiles in uninfected donors and people living with HIV [6,64]. Vδ2 T cell immunotherapeutic potential to cure HIV as well as other infectious diseases remains an intriguing avenue of research and has been previously reviewed by our group and others [76,79].
In several preclinical studies, administration of N-BPs or adoptive transfer of ex vivo expanded Vδ2 T cells demonstrated a therapeutic effect against both influenza and flavivirus infections (e.g., West Nile Virus and Hepatitis C Virus) [80,81,82,83]. Similar strategies were found to mediate the control of Mycobacterium tuberculosis infection in non-human primates [84,85]. Targeting Vδ2 T cell responses through vaccination is another possibility that has shown potential. Although historically neglected in vaccine research, the effect of the Mycobacterium bovis derived Bacillus Calmette-Guérin (BCG) vaccine on Vδ2 T cells has been well documented. BCG vaccination induces Vδ2 T cell expansion which is accompanied by increased IFN-γ production and changes in effector memory phenotypes that may confer protection to future infection [86,87,88,89]. Despite the lack of clinical validation in humans, these findings provide further evidence of the therapeutic potential of Vδ2 T cells against a wide breadth of pathogenic infections.

3.2. Immunotherapy for Cancer

The critical importance of γδ T cell responses in immunosurveillance of both malignancies and pathogenic infection has been well established [90,91,92]. The ability of γδ T cells to exert potent cytotoxic functions in recognition of both self and foreign antigens, as well as stress ligands underscores their potential as a highly dynamic immunotherapeutic approach. Antitumor functions of γδ T cells have been extensively reviewed elsewhere and are only briefly mentioned here [1,8,93,94,95,96]. In addition to the TCR, γδ T cells recognize tumor cells through a variety of innate-like receptors involved in the recognition of stress molecules. Interestingly, receptor utilization for tumor killing is dependent on the specific tumor and microenvironment [97]. Stress-induced molecules upregulated in transformed cells including MHC class I polypeptide-related sequence A (MICA) or MICB bind to natural killer group 2 member D (NKG2D) [98]. Additional NK receptors like DNAM-1, natural cytotoxicity-triggering receptors NKp30 or NKp44 and TNF receptors are also involved in tumor recognition [99]. Expression of CD16 also enables γδ T cells to exert antibody-dependent cellular cytotoxicity upon engagement of membrane-surface antigens on the target cell bound to specific antibodies [100,101]. Upon ligation, γδ T cells produce cytotoxic molecules such as granzymes, perforins, and they can exert their killing functions through engagement of TNF-related apoptosis-inducing ligand (TRAIL) and Fas ligand (FasL) [102]. In addition to the direct cytotoxicity, γδ T cells provide necessary signals to induce responses from other cytotoxic effectors including production of cytokines that enables dendritic cell maturation as well as enhancement of CD8 T cell or NK cell killing [93,103,104].
The earliest clinical application of γδ T cells began with attempts to directly expand autologous Vδ2 T cells in vivo for individuals with late-stage malignancies that had exhausted previous lines of therapy. This came after observations that short course PAM and IL-2 could not only activate, but also boost Vδ2 T cell antitumor functions [36,105]. Several strategies have been utilized to harness Vδ2 T cells as an immunotherapy, differing on the source of donor cells, activating stimuli, and conditions for activation. Most of the early studies attempted Vδ2 T cell activation by directly administering N-BPs (e.g., PAM or ZOL) or synthetic P-Ag (e.g., BrHPP or 2M3B1PP) with low dose IL-2 to patients being treated for either solid tumors or hematological malignancies [106,107,108,109,110,111,112,113]. The use of synthetic P-Ags has the advantage of directly activating Vδ2 T cells rather than indirectly through inducing the accumulation of IPP upon treatment with N-BPs. In addition, synthetic P-Ags are more potent and require nanomolar concentrations, although its use has not been pursued probably due to the decreased number of Vδ2 T cells after prolonged use [113,114]. On the contrary, several studies have demonstrated the antitumor capacity of N-BPs by not only activating Vδ2 T cells, but also promoting apoptosis of sensitized cells [8,115]. In vivo treatment was generally well tolerated with only several observations of dose-limiting toxicities or high-grade adverse events possibly due to IL-2 toxicity. Despite robust expansion of peripheral Vδ2 T cells in many participants, anti-tumor activity was insufficient to significantly improve outcomes. Notably, repeated exposure to N-BPs or synthetic P-Ag throughout the course of treatment led to an increasingly exhausted phenotype of Vδ2 T cells, which may have limited any clinical benefit [66,107,108].
In an effort to improve efficacy and mitigate the aforementioned challenges observed in previous studies, researchers began manipulating autologous Vδ2 T cells ex vivo followed by reinfusion of expanded cells into the individual undergoing treatment, and studies are summarized in Table 1 [112,114,116,117,118,119,120,121,122,123,124,125,126,127,128,129]. In addition, a list of ongoing clinical trials targeting hematological malignancies have been recently reviewed [130]. The advantages to this approach include the ability to characterize the phenotype and anti-tumor potency of expanded Vδ2 T cells prior to administration to the participant. The phenotype of the expanded cell product has been characterized in several studies. The majority of ex vivo expanded γδ T-cells were CD27- CD45RA- cells [113,114,118,123,124,131] and had high NKG2D expression on their surface [123,124,125,131]. In several studies, expanded cells did not express PD-1 or CTLA-4, but did have detectable expression of TIM-3 and LAG-3 [123,125].
In addition, adoptively transferred cells could be radiolabeled to assess their ability to traffic to and penetrate solid tumors. Using indium [111] oxine, Nicol AJ et al., demonstrated that ex vivo expanded Vδ2 T cells are capable of penetrating metastatic sites within a participant with melanoma and another with lung adenocarcinoma [121]. Interestingly, the administered Vδ2 T cells displayed a similar pattern of tissue trafficking in each participant that received In-111 labeled cells. This was noted by rapid migration to the lungs during the first hour, which began to wane concurrently with their increased appearance in the liver and spleen by the fourth hour; a behavior that mimics adoptively transferred tumor-specific cytotoxic αβ T cells [132]. Similar to direct in vivo expansion, this strategy proved to be feasible and safe. Unfortunately, clinical outcomes varied between individual participants based on cancer type, disease stage, and previous treatment regimen. These factors may inherently limit the therapeutic use of autologous Vδ2 T cells within this context. Further studies are required to determine if there is a cancer-specific mechanism that impairs Vδ2 T cell proliferative and cytotoxic potential as well as individual effects of concurrent or previous therapies (e.g., chemotherapy, radiation, surgical resection, or additional immunotherapies).
In order to overcome the inherent limitations of autologous cells, we may exploit the MHC-independent mechanisms by which γδ T cells recognize and kill target cells. In the context of transplanting donor derived cells into a recipient (e.g., hematopoietic stem cell transplant), the genetic compatibility of the pair is paramount due to the risk of developing life-threatening Graft v. Host disease [133]. Studies on haploidentical HSCT revealed a positive association between allogeneic donor γδ T cells and disease-free survival in patients with late-stage hematological malignancies. This paved the way for separate trials investigating the direct infusion of ex vivo expanded allogeneic Vδ2 T cells [126,127,129,134,135]. These early stage trials published by Alnaggar. et al. and Xu et al. were pivotal in demonstrating the clinical proof of concept for an allogeneic Vδ2 T cell therapy using a solid tumor setting. Most notably, efficacy was demonstrated in cases by extended overall survival in the absence of severe adverse events for patients with lung or liver cancer. Positive changes in autologous αβ T cell and NK cell phenotypes were also observed, suggesting the infused cells may boost existing anti-tumor responses. Several of these observations were validated in more recent studies that explored allogeneic Vδ2 T cells to treat brain, pancreatic, or hepatic cancers [125,128,136]. Safety was consistent across each study, but efficacy remained mixed possibly due to factors such as previous or cotreatment, tumor burden, and systemic v. localized infusion. Despite these shortcomings, the robust responses observed in some individuals provides hope and justification for further investigation into allogeneic Vδ2 T cell-based immunotherapies for the treatment of cancer.

3.3. Lessons Learned from the Clinic

Regardless of the type of Vδ2 T cell therapy implementation, a common theme of variable clinical responses has been observed between previous trials. One of the more convincing arguments is based on evidence that suggests there are inherent interpersonal differences within Vδ2 T cell populations that directly impact their therapeutic potential. The Vδ2 T cell compartment is thought to be shaped by interaction with endogenous P-Ag during gestation as well as post-natal exposure to commensal bacteria [137,138,139]. Despite the prevalence of public CDR3γ9 clonotypes within Vδ2 T cell populations in adults, the influence of early life events on selecting clone lineages or skewing effector differentiation cannot be discounted. When factoring in the effects of age, gender, and previous P-Ag mediated activation, work by Ryan et al., and further work by our group, demonstrated that humans possess distinct subpopulations of Vδ2 T cells that differ in their cytotoxic and proliferative capabilities [6,64]. The variability of Vδ2 T cell responses within the clinic may be related to the polyclonal or homogeneous expansion of proliferative or cytotoxic subpopulations of Vδ2 T cells with mixed potency against tumor and infected cells [6].
The identification of an optimal donor profile or biomarker of highly responsive Vδ2 T cells may also lead to improved therapies. Examples of such potential biomarkers include CD16 expression associated with anti-HIV activity as demonstrated by our group [64,72] as well as increased NKG2A expression linked to hyperresponsive anti-tumor function as recently shown by Cazzetta et al. [19]. Additionally, overcoming donor heterogeneity may require optimizing the stimuli and culture conditions used within Vδ2 T cell expansion protocols, since currently we lack an accepted clinical protocol to specifically expand Vδ2 T cells. The different methods for Vδ2 T cell expansion are summarized in Table 2. Overcoming the variability of clinical responses is a critical step to make this approach widely available and a reality for treating a variety of diseases regardless of the origin.

3.4. Enhancing Vδ2 T Cell Cytotoxic Effector Functions

The development of a clinical protocol may comprise strategic utilization of several approaches to produce adequate numbers of highly cytotoxic Vδ2 T effector cells. An increasing number of studies focused on γδ T cell immunotherapy has provided further insight into developing expansion protocols where specific interventions may be used to modulate Vδ2 T cell proliferation, phenotype, and function. To provide the proper context for interpreting these studies, it is important to understand which receptors mediate γδ T cell to recognition of target cells, their downstream effects on γδ T cell responses, and whether modulation of the expression of such receptors during expansion is possible.
γδ T cells recognize malignant cells through the engagement of innate activating receptors like NKG2D, and natural cytotoxicity receptors (NCRs) [1,155,156,157,158]. Therefore, inducing their expression may enhance recognition and possibly killing. NKG2D acts as a costimulatory signal upon recognition of stress markers, MHC class I chain related protein A and B (MICA, MICB) and UL16 binding proteins (ULBPs). Activation through this pathway provokes the secretion of proinflammatory cytokines TNF-α and IFN-γ as well as direct cytolytic activity mediated by the release of perforins and granzymes [159,160,161].
Several strategies could be used to upregulate NKG2D ligands on the surface of cancer cells. Temozolomide, used to treat brain tumors such as glioblastoma or Gemcitabine for bladder and breast cancer, increases the expression of NKG2D ligands making tumor cells more susceptible to recognition and lysis by γδ T cells [162,163]. Treatment with the histone deacetylase inhibitor, Valproic acid (VPA), significantly enhanced the expression of the NKG2D ligands MICA, MICB and ULBP-2, in a pancreatic carcinoma cell line (Panc89) and prostate carcinoma cell line (PC-3) [164]. Given the importance of NKG2D/ligands in cancer recognition and cytotoxicity, an expansion protocol that increases NKG2D expression on Vδ2 cells may be desirable. Dr. Kabelitz’s group demonstrated a differential capacity of HDACi to induce NKG2D expression on Vδ2 T cells, which could be an additional intervention for developing an optimized clinical expansion protocol [164].
Vδ2 cells can also express NKG2A which induces inhibitory signals through engagement with HLA-E/CD94 [165,166]. Interestingly, a recent study by Cazzetta, V. et al. demonstrated that expression of NKG2A identified a highly cytotoxic Vδ2 T cell subset against a variety of cancer cells from different origins. Furthermore, Vδ2 T cells expressing high levels of NKG2A supplied with remarkably higher cytokine production and cytotoxic potential compared to Vδ2 T cells that did not express NKG2A [19]. Since HLA-E is expressed virtually on all human tissues and allows for self-recognition, increased expression of HLA-E in several tumors constitute a mechanism of tumor escape that evades immune effector responses [167,168,169,170]. Therefore, extra attention should be given to changes in NKG2A expression during Vδ2 cell expansion and any attempts to select for this population will require additional measures to ensure the inhibitory nature of this receptor does not limit clinical efficacy.
γδ T cell activity is also regulated by immune checkpoint receptors, such as program death-1 (PD-1), cytotoxic T-lymphocyte associated protein 4 (CTLA-4), lymphocyte-activation gene 3 (LAG-3), and T cell Immunoglobulin domain and mucin domain 3 (TIM-3) [171,172,173,174]. Blocking the inhibitory signaling mediated by these receptors with therapeutic monoclonal antibodies (mAbs) has proven beneficial for improving immune responses to refractory malignancies such as melanoma, non-small cell lung cancer, and renal cell carcinoma. This led to the development of FDA-approved mAbs targeting PD1/PD-L1, CTLA-4, and more recently LAG-3 [175,176]. The effects of immune checkpoint blockade (ICB) have primarily been studied within CD8 T cells, but several preliminary studies suggest a positive benefit may extend to γδ T cell-based immunotherapies. Although blockade of PD-1 does not appear to improve proliferation in vitro, it does result in increased IFN-γ production. Recent studies have shown that PD-1 checkpoint blockade enhanced anti-tumor immunity of adoptively transferred Vδ2 T cells in preclinical models and clinical trials [177,178,179]. Similarly, overexpression of TIM-3 on γδ T cells coincides with reduced expression of perforin and granzymeB and therefore decreased cytotoxicity against colon cancer cells [180]. Despite the lack of direct clinical evidence for ICB targeting γδ T cells, one study involving the CTLA-4 targeting mAb ipilimumab revealed a positive correlation between Vδ2 T cell frequencies and survival outcome for individuals who received the treatment [181]. These observations necessitate further investigation into harnessing ICB in order to improve γδ T cell immunotherapies. Activated Vδ2 T cells transiently upregulate the expression of PD-1 during ex vivo expansion with N-BPs [7,140,171,182]. TIM-3 and CTLA-4 expression has also been shown to be significantly up-regulated on Vδ2 T cells during expansion with ZOL which may contribute to a degree of cytotoxic dysfunction and increased susceptibility to apoptosis in a portion of expanded cells [183]. Importantly, effective blockade of PD-1 and TIM-3 during expansion may rescue γδ T cell cytotoxicity and effector function [183]. Despite the lack of ICB studies on γδ T cells, these collective preliminary findings suggest that blocking inhibitory receptors expressed on γδ T cells during expansion may provide an additional method of enhancing their cytotoxic functions.

3.5. Vδ2 T Cell Expansion: Combining the Cytokine Cocktail

Vδ2 T cell activation and expansion requires a secondary signal from cytokines that affect Vδ2 T cell phenotype and functions (Figure 2).
Interleukins consist of a large group of proteins that can elicit distinct reactions in cells and tissues by binding to high-affinity receptors on the cell surface [184]. They play essential roles in the activation, differentiation, proliferation, maturation, migration, and adhesion of immune cells [184]. Below, we review cytokine use for the expansion of Vδ2 T cells, and the in vitro effect on Vδ2 T cell phenotype and function.
IL-2. IL-2 has been the most frequently used cytokine for the expansion of Vδ2 T cells. IL-2 is a growth factor essential for the proliferation of T cells and the generation of effector and memory cells [185,186]. N-BPs and p-Ags, in combination with IL-2 are strong stimulators of Vδ2 T cell activation and proliferation in ex vivo cultures of human PBMCs [28,36,140,141]. In an average of 10 to 14 days, Vδ2 T cells can expand between 40–70% depending on the N-BPs used [27,28,142,143,144,145]. Expansion protocols provided an initial exposure to N-BP or p-Ag in combination with IL-2 (100 and 1000 U/mL), replenished every 2–3 days, led to increased expression of cytotoxicity and activation markers like CD16, CD56, Nkp44, NKp30, and the production of IFN-γ, granzymeB, perforin, or TNF-α [22,28,141,142,146,150,151]. The anti-tumor functions of IL-2-expanded Vδ2 T cells against several tumor cancer cell lines have been investigated including human neuroblastoma, myeloma, glioblastoma, melanoma, and renal adenocarcinoma and Burkitt´s lymphoma [29,140,141,142,146,147,148].
IL-12. IL-12 has been studied extensively as a proinflammatory cytokine inducing production of IFN-γ by αβ T cells as well as γδ T cells. Expansion of Vδ2 T cells using a combination of IL-2, IL-12, and IL-18, induced over 80% IFN-γ producing Vδ2 T cells compared to a modest increase when using IL-2 combined with IL-15 [150,151]. The combination of IL-2, IL-12, and IL-18 also resulted in potent antitumor activity against different cell lines like Ewing sarcoma (A673), rhabdomyosarcoma (RH30), and neuroblastoma (SH-SY5Y) [150]. Upon IL-2, IL-12, and IL-18 stimulation, Vδ2 T cells also induced cell cycle arrest in various tumor cells like bladder carcinoma (T24), breast cancer (MCF7) and melanoma (WM115) [150].
IL-15. IL-15 plays a fundamental role in the stimulation of innate and adaptive cells, the reactivation of memory T cells, the induction of DC maturation, and the proliferation and activation of αβ T cells, γδ T cells, NK cells and B cells [187,188]. With an average of 6 to 14 days, studies have shown that Vδ2 T cells expanded between 30–60% depending on the p-Ag or NBPs used (IPP, BrHPP or ZOL) [18,27,140,152]. p-Ag combined with IL-15 significantly augmented T cell proliferation and intracellular cytokine expression, and together with IL-12, IFN-γ production was enhanced [152]. IL-15 also induced increased killing in ZOL or BrHPP-activated Vδ2 T cells against different tumor cell lines (Daudi and U299 cells) compared to IL-2 alone [140,189]. In addition, a higher killing was accompanied by an IL-15-mediated overexpression of cytotoxic markers and molecules like CD56, NKG2A, IFN-γ, TNF-α, and higher direct killing against several adherent tumor cell lines [18,180,189]. Similarly, IL-15 in combination with IL-2 boosted the proliferation as well as the in vitro anti-tumor activity of ZOL-expanded Vδ2 T cells [140].
IL-21. IL-21 is known to improve CD8+ T and NK cell cytotoxicity [29]. IL-21 combined with IL-2 promotes p-Ag-induced Vδ2 T cell proliferation [27,28,29]. In addition, IL-21 polarizes Vδ2 T cells toward a cytotoxic effector phenotype with an increase in granzymeA, granzymeB and perforin production levels [27,28,29]. The increase in cytotoxic activity is associated with enhanced tumor cell killing, although IL-21 neither synergizes nor interferes with IL-2 [29]. In acute myeloid leukemia patients, however, there was a reduction in the efficacy of IL-21 to increase proliferation and cytotoxicity of Vδ2 T, with a lower IL-21R and higher TIM-3 expression. Blocking TIM-3 could significantly improve the efficacy of IL-21 [190,191].
IL-18. IL-18 was discovered as an IFN-γ inducing factor and different studies demonstrated that it plays essential roles in host defense against infections and tumors, and in the pathogenesis of various inflammatory diseases through the upregulation of IFN-γ production [192,193,194]. IL-18 induced increased expansion of Vδ2 T cells stimulated by ZOL and IL-2, and expanded cells displayed an effector memory phenotype, with high levels of production of IFN-γ, TNF-α and strong cytotoxicity against tumor cells like mesothelioma cells [153,154]. As mentioned above, IL-18, in combination with IL-12, also increases proliferation, anti-tumor activity and IFN-γ production [150].
TGF-β. TGF-β is a pleiotropic cytokine that play roles in adult and embryonic growth and development, in inflammation and repair including angiogenesis and regulation of host resistance mechanisms [195]. TGF-β, combined with IL-2 or IL-15, increased the proliferation and cytotoxicity of BrHPP-activated Vδ2 T cells in vitro [196]. In addition, the expression of CD54, CD103, IFN-γ, IL-9 and granzyme-B were upregulated upon expansion. However, CD56 and integrin CD11a/CD18 were downregulated suggesting that transmigration into tissues may be impaired [18,197]. TGF- β expanded Vδ2 T cells were shown to have tumor cell killing against cell lines derived from pancreatic ductal adenocarcinoma (PDAC) like Colo357, MiaPaCa2, Panc1 and Panc89, as well as mammary gland adenocarcinoma cell line (MCF7) [18]. Vδ2 T cells activated with TGF-β have also shown to have better yield, viability, cytokine release and tumor cell killing compared to IL-2 activated controls [198]. However, TGF- β expanded Vδ2 T cells, displayed a more regulatory phenotype characterized by increased FOXP3 expression, and expression of PD-1, CTLA4 and CD80/CD86 were upregulated [7,8,149]. This suppressive function can be deemed counterproductive for use in immunotherapy.
Vitamin C. Vitamin C (L-ascorbic acid) is an important vitamin in many different biological processes, and it is a potent antioxidant and an essential cofactor in many enzymatic reactions [199,200]. Dr. Kabelitz’s group pioneered the investigation of the effect of Vitamin C and its more stable and less toxic derivative, L-ascorbic acid 2-phosphate on the proliferation and effector function effect of ZOL or synthetic p-Ags. Vitamin C and its analog reduced apoptosis and increased cytokine production during primary stimulation of a 14-day period of purified Vδ2 T cells [17]. The modulatory effect of vitamin C and its analog can be harnessed in order to optimize γδ T cell generation for cellular therapy [17].

4. Conclusions and Future Perspective

Overall, current developed protocols for clinical applications in oncology have achieved individual goals. However, a specific protocol strategically combining some approaches and combinations of cytokines may help achieve obtaining a universal protocol subjected to certain modifications based on the clinical use [16]. The clinical utilization of γδ T cells for infectious diseases, including HIV cure, remains understudied compared to the cancer field. Studies from our group and others paired with an examination of Vδ2 T cell basic biology warrants further investigation into their potential as an immunotherapy, beyond its utility against cancer [64,68,69,201].
The past two decades have provided examples of incredible progress in harnessing the power of the immune system in the form of life-saving cellular therapies. Despite cell therapy gaining acknowledgment as a new pillar of medicine, we are arguably still within the field’s infancy. Through the tireless efforts of pioneering researchers, we are well positioned to take the next step in developing γδ T cell-based immunotherapies through improved expansion protocols, possibly without requiring additional engineered genetic modifications. There are still a number of outstanding questions to answer both within the lab and in the clinic, but with a convergence of expertise from different fields, γδ T cells are poised to become the next tool in our armamentarium against disease.

Author Contributions

Writing original draft, M.S., A.C., B.T.M. and N.S.-S.; Resources, N.S.-S.; Conceptualization, N.S.-S. All authors have read and agreed to the published version of the manuscript.

Funding

This work was funded by NIH Grant R01-AI125097 and R21-AI157864.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Figures were created using Biorender, accessed on 10 December 2022.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Vantourout, P.; Hayday, A. Six-of-the-best: Unique contributions of γδ T cells to immunology. Nat. Rev. Immunol. 2013, 13, 88–100. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Silva-Santos, B.; Serre, K.; Norell, H. γδ T cells in cancer. Nat. Rev. Immunol. 2015, 15, 683–691. [Google Scholar] [CrossRef] [PubMed]
  3. Wu, D.; Wu, P.; Qiu, F.; Wei, Q.; Huang, J. Human γδT-cell subsets and their involvement in tumor immunity. Cell Mol. Immunol. 2017, 14, 245–253. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Takihara, Y.; Reimann, J.; Michalopoulos, E.; Ciccone, E.; Moretta, L.; Mak, T.W. Diversity and structure of human T cell receptor delta chain genes in peripheral blood gamma/delta-bearing T lymphocytes. J. Exp. Med. 1989, 169, 393–405. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Wu, Y.L.; Ding, Y.P.; Tanaka, Y.; Shen, L.W.; Wei, C.H.; Minato, N.; Zhang, W. γδ T cells and their potential for immunotherapy. Int. J. Biol. Sci. 2014, 10, 119–135. [Google Scholar] [CrossRef] [Green Version]
  6. Ryan, P.L.; Sumaria, N.; Holland, C.J.; Bradford, C.M.; Izotova, N.; Grandjean, C.L.; Jawad, A.S.; Bergmeier, L.A.; Pennington, D.J. Heterogeneous yet stable Vδ2(+) T-cell profiles define distinct cytotoxic effector potentials in healthy human individuals. Proc. Natl. Acad. Sci. USA 2016, 113, 14378–14383. [Google Scholar] [CrossRef] [Green Version]
  7. Peters, C.; Oberg, H.H.; Kabelitz, D.; Wesch, D. Phenotype and regulation of immunosuppressive Vδ2-expressing γδ T cells. Cell Mol. Life Sci. 2014, 71, 1943–1960. [Google Scholar] [CrossRef] [Green Version]
  8. Kabelitz, D.; Serrano, R.; Kouakanou, L.; Peters, C.; Kalyan, S. Cancer immunotherapy with γδ T cells: Many paths ahead of us. Cell. Mol. Immunol. 2020, 17, 925–939. [Google Scholar] [CrossRef]
  9. Wu, J.; Groh, V.; Spies, T. T cell antigen receptor engagement and specificity in the recognition of stress-inducible MHC class I-related chains by human epithelial gamma delta T cells. J. Immunol. 2002, 169, 1236–1240. [Google Scholar] [CrossRef] [Green Version]
  10. Rincon-Orozco, B.; Kunzmann, V.; Wrobel, P.; Kabelitz, D.; Steinle, A.; Herrmann, T. Activation of V gamma 9V delta 2 T cells by NKG2D. J. Immunol. 2005, 175, 2144–2151. [Google Scholar] [CrossRef]
  11. Dalton, J.E.; Howell, G.; Pearson, J.; Scott, P.; Carding, S.R. Fas-Fas ligand interactions are essential for the binding to and killing of activated macrophages by gamma delta T cells. J. Immunol. 2004, 173, 3660–3667. [Google Scholar] [CrossRef] [Green Version]
  12. Ponomarev, E.D.; Dittel, B.N. Gamma delta T cells regulate the extent and duration of inflammation in the central nervous system by a Fas ligand-dependent mechanism. J. Immunol. 2005, 174, 4678–4687. [Google Scholar] [CrossRef] [Green Version]
  13. Wu, Y.; Wu, W.; Wong, W.M.; Ward, E.; Thrasher, A.J.; Goldblatt, D.; Osman, M.; Digard, P.; Canaday, D.H.; Gustafsson, K. Human gamma delta T cells: A lymphoid lineage cell capable of professional phagocytosis. J. Immunol. 2009, 183, 5622–5629. [Google Scholar] [CrossRef] [Green Version]
  14. Chen, Z.; Freedman, M.S. CD16+ gammadelta T cells mediate antibody dependent cellular cytotoxicity: Potential mechanism in the pathogenesis of multiple sclerosis. Clin. Immunol. 2008, 128, 219–227. [Google Scholar] [CrossRef]
  15. Di Lorenzo, B.; Simões, A.E.; Caiado, F.; Tieppo, P.; Correia, D.V.; Carvalho, T.; da Silva, M.G.; Déchanet-Merville, J.; Schumacher, T.N.; Prinz, I.; et al. Broad Cytotoxic Targeting of Acute Myeloid Leukemia by Polyclonal Delta One T Cells. Cancer Immunol. Res. 2019, 7, 552–558. [Google Scholar] [CrossRef] [Green Version]
  16. Almeida, A.R.; Correia, D.V.; Fernandes-Platzgummer, A.; da Silva, C.L.; da Silva, M.G.; Anjos, D.R.; Silva-Santos, B. Delta One T Cells for Immunotherapy of Chronic Lymphocytic Leukemia: Clinical-Grade Expansion/Differentiation and Preclinical Proof of Concept. Clin. Cancer Res. 2016, 22, 5795–5804. [Google Scholar] [CrossRef] [Green Version]
  17. Kouakanou, L.; Xu, Y.; Peters, C.; He, J.; Wu, Y.; Yin, Z.; Kabelitz, D. Vitamin C promotes the proliferation and effector functions of human γδ T cells. Cell. Mol. Immunol. 2020, 17, 462–473. [Google Scholar] [CrossRef] [Green Version]
  18. Peters, C.; Meyer, A.; Kouakanou, L.; Feder, J.; Schricker, T.; Lettau, M.; Janssen, O.; Wesch, D.; Kabelitz, D. TGF-β enhances the cytotoxic activity of Vδ2 T cells. Oncoimmunology 2019, 8, e1522471. [Google Scholar] [CrossRef] [Green Version]
  19. Cazzetta, V.; Bruni, E.; Terzoli, S.; Carenza, C.; Franzese, S.; Piazza, R.; Marzano, P.; Donadon, M.; Torzilli, G.; Cimino, M.; et al. NKG2A expression identifies a subset of human Vδ2 T cells exerting the highest antitumor effector functions. Cell Rep. 2021, 37, 109871. [Google Scholar] [CrossRef]
  20. Constant, P.; Davodeau, F.; Peyrat, M.A.; Poquet, Y.; Puzo, G.; Bonneville, M.; Fournié, J.J. Stimulation of human gamma delta T cells by nonpeptidic mycobacterial ligands. Science 1994, 264, 267–270. [Google Scholar] [CrossRef]
  21. Tanaka, Y.; Morita, C.T.; Tanaka, Y.; Nieves, E.; Brenner, M.B.; Bloom, B.R. Natural and synthetic non-peptide antigens recognized by human gamma delta T cells. Nature 1995, 375, 155–158. [Google Scholar] [CrossRef] [PubMed]
  22. Hintz, M.; Reichenberg, A.; Altincicek, B.; Bahr, U.; Gschwind, R.M.; Kollas, A.K.; Beck, E.; Wiesner, J.; Eberl, M.; Jomaa, H. Identification of (E)-4-hydroxy-3-methyl-but-2-enyl pyrophosphate as a major activator for human gammadelta T cells in Escherichia coli. FEBS Lett. 2001, 509, 317–322. [Google Scholar] [CrossRef] [Green Version]
  23. Jomaa, H.; Feurle, J.; Luhs, K.; Kunzmann, V.; Tony, H.P.; Herderich, M.; Wilhelm, M. Vgamma9/Vdelta2 T cell activation induced by bacterial low molecular mass compounds depends on the 1-deoxy-D-xylulose 5-phosphate pathway of isoprenoid biosynthesis. FEMS Immunol. Med. Microbiol. 1999, 25, 371–378. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Amslinger, S.; Kis, K.; Hecht, S.; Adam, P.; Rohdich, F.; Arigoni, D.; Bacher, A.; Eisenreich, W. Biosynthesis of terpenes. Preparation of (E)-1-hydroxy-2-methyl-but-2-enyl 4-diphosphate, an intermediate of the deoxyxylulose phosphate pathway. J. Org. Chem. 2002, 67, 4590–4594. [Google Scholar] [CrossRef] [PubMed]
  25. Reichenberg, A.; Hintz, M.; Kletschek, Y.; Kuhl, T.; Haug, C.; Engel, R.; Moll, J.; Ostrovsky, D.N.; Jomaa, H.; Eberl, M. Replacing the pyrophosphate group of HMB-PP by a diphosphonate function abrogates Its potential to activate human gammadelta T cells but does not lead to competitive antagonism. Bioorg. Med. Chem. Lett. 2003, 13, 1257–1260. [Google Scholar] [CrossRef]
  26. Fox, D.T.; Poulter, C.D. Synthesis of (E)-4-hydroxydimethylallyl diphosphate. An intermediate in the methyl erythritol phosphate branch of the isoprenoid pathway. J. Org. Chem. 2002, 67, 5009–5010. [Google Scholar] [CrossRef]
  27. Eberl, M.; Engel, R.; Beck, E.; Jomaa, H. Differentiation of human gamma-delta T cells towards distinct memory phenotypes. Cell. Immunol. 2002, 218, 1–6. [Google Scholar] [CrossRef]
  28. Vermijlen, D.; Ellis, P.; Langford, C.; Klein, A.; Engel, R.; Willimann, K.; Jomaa, H.; Hayday, A.C.; Eberl, M. Distinct cytokine-driven responses of activated blood gammadelta T cells: Insights into unconventional T cell pleiotropy. J. Immunol. 2007, 178, 4304–4314. [Google Scholar] [CrossRef] [Green Version]
  29. Thedrez, A.; Harly, C.; Morice, A.; Salot, S.; Bonneville, M.; Scotet, E. IL-21-mediated potentiation of antitumor cytolytic and proinflammatory responses of human V gamma 9V delta 2 T cells for adoptive immunotherapy. J. Immunol. 2009, 182, 3423–3431. [Google Scholar] [CrossRef] [Green Version]
  30. Green, A.E.; Lissina, A.; Hutchinson, S.L.; Hewitt, R.E.; Temple, B.; James, D.; Boulter, J.M.; Price, D.A.; Sewell, A.K. Recognition of nonpeptide antigens by human V gamma 9V delta 2 T cells requires contact with cells of human origin. Clin. Exp. Immunol. 2004, 136, 472–482. [Google Scholar] [CrossRef]
  31. Gober, H.J.; Kistowska, M.; Angman, L.; Jenö, P.; Mori, L.; De Libero, G. Human T cell receptor gammadelta cells recognize endogenous mevalonate metabolites in tumor cells. J. Exp. Med. 2003, 197, 163–168. [Google Scholar] [CrossRef] [Green Version]
  32. Gossman, W.; Oldfield, E. Quantitative structure--activity relations for gammadelta T cell activation by phosphoantigens. J. Med. Chem. 2002, 45, 4868–4874. [Google Scholar] [CrossRef]
  33. Buhaescu, I.; Izzedine, H. Mevalonate pathway: A review of clinical and therapeutical implications. Clin. Biochem 2007, 40, 575–584. [Google Scholar] [CrossRef]
  34. Greenwood, J.; Steinman, L.; Zamvil, S.S. Statin therapy and autoimmune disease: From protein prenylation to immunomodulation. Nat. Rev. Immunol. 2006, 6, 358–370. [Google Scholar] [CrossRef] [Green Version]
  35. Guerra, B.; Recio, C.; Aranda-Tavío, H.; Guerra-Rodríguez, M.; García-Castellano, J.M.; Fernández-Pérez, L. The Mevalonate Pathway, a Metabolic Target in Cancer Therapy. Front. Oncol. 2021, 11, 626971. [Google Scholar] [CrossRef]
  36. Kunzmann, V.; Bauer, E.; Wilhelm, M. Gamma/delta T-cell stimulation by pamidronate. N. Engl. J. Med. 1999, 340, 737–738. [Google Scholar] [CrossRef]
  37. Tricarico, P.M.; Girardelli, M.; Kleiner, G.; Knowles, A.; Valencic, E.; Crovella, S.; Marcuzzi, A. Alendronate, a double-edged sword acting in the mevalonate pathway. Mol. Med. Rep. 2015, 12, 4238–4242. [Google Scholar] [CrossRef] [Green Version]
  38. Ebetino, F.H.; Sun, S.; Cherian, P.; Roshandel, S.; Neighbors, J.D.; Hu, E.; Dunford, J.E.; Sedghizadeh, P.P.; McKenna, C.E.; Srinivasan, V.; et al. Bisphosphonates: The role of chemistry in understanding their biological actions and structure-activity relationships, and new directions for their therapeutic use. Bone 2022, 156, 116289. [Google Scholar] [CrossRef]
  39. Thompson, K.; Rogers, M.J. Statins prevent bisphosphonate-induced γ,δ-T-cell proliferation and activation in vitro. J. Bone Miner. Res. 2004, 19, 278–288. [Google Scholar] [CrossRef]
  40. Moghadasian, M.H. Clinical pharmacology of 3-hydroxy-3-methylglutaryl coenzyme A reductase inhibitors. Life Sci. 1999, 65, 1329–1337. [Google Scholar] [CrossRef]
  41. van Beek, E.; Pieterman, E.; Cohen, L.; Löwik, C.; Papapoulos, S. Farnesyl pyrophosphate synthase is the molecular target of nitrogen-containing bisphosphonates. Biochem. Biophys. Res. Commun. 1999, 264, 108–111. [Google Scholar] [CrossRef] [PubMed]
  42. Benzaïd, I.; Mönkkönen, H.; Stresing, V.; Bonnelye, E.; Green, J.; Mönkkönen, J.; Touraine, J.L.; Clézardin, P. High phosphoantigen levels in bisphosphonate-treated human breast tumors promote Vgamma9Vdelta2 T-cell chemotaxis and cytotoxicity in vivo. Cancer Res. 2011, 71, 4562–4572. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Keller, R.K.; Fliesler, S.J. Mechanism of aminobisphosphonate action: Characterization of alendronate inhibition of the isoprenoid pathway. Biochem. Biophys. Res. Commun. 1999, 266, 560–563. [Google Scholar] [CrossRef] [PubMed]
  44. Göbel, A.; Thiele, S.; Browne, A.J.; Rauner, M.; Zinna, V.M.; Hofbauer, L.C.; Rachner, T.D. Combined inhibition of the mevalonate pathway with statins and zoledronic acid potentiates their anti-tumor effects in human breast cancer cells. Cancer Lett. 2016, 375, 162–171. [Google Scholar] [CrossRef] [PubMed]
  45. Nakazawa, T.; Nakamura, M.; Matsuda, R.; Nishimura, F.; Park, Y.S.; Motoyama, Y.; Hironaka, Y.; Nakagawa, I.; Yokota, H.; Yamada, S.; et al. Antitumor effects of minodronate, a third-generation nitrogen-containing bisphosphonate, in synergy with γδT cells in human glioblastoma in vitro and in vivo. J. Neurooncol. 2016, 129, 231–241. [Google Scholar] [CrossRef]
  46. Göbel, A.; Zinna, V.M.; Dell’Endice, S.; Jaschke, N.; Kuhlmann, J.D.; Wimberger, P.; Rachner, T.D. Anti-tumor effects of mevalonate pathway inhibition in ovarian cancer. BMC Cancer 2020, 20, 703. [Google Scholar] [CrossRef]
  47. Harly, C.; Guillaume, Y.; Nedellec, S.; Peigne, C.M.; Monkkonen, H.; Monkkonen, J.; Li, J.; Kuball, J.; Adams, E.J.; Netzer, S.; et al. Key implication of CD277/butyrophilin-3 (BTN3A) in cellular stress sensing by a major human gammadelta T-cell subset. Blood 2012, 120, 2269–2279. [Google Scholar] [CrossRef] [Green Version]
  48. Rigau, M.; Ostrouska, S.; Fulford, T.S.; Johnson, D.N.; Woods, K.; Ruan, Z.; McWilliam, H.E.G.; Hudson, C.; Tutuka, C.; Wheatley, A.K.; et al. Butyrophilin 2A1 is essential for phosphoantigen reactivity by γδ T cells. Science 2020, 367, eaay5516. [Google Scholar] [CrossRef]
  49. Sandstrom, A.; Peigné, C.M.; Léger, A.; Crooks, J.E.; Konczak, F.; Gesnel, M.C.; Breathnach, R.; Bonneville, M.; Scotet, E.; Adams, E.J. The intracellular B30.2 domain of butyrophilin 3A1 binds phosphoantigens to mediate activation of human Vγ9Vδ2 T cells. Immunity 2014, 40, 490–500. [Google Scholar] [CrossRef] [Green Version]
  50. Vavassori, S.; Kumar, A.; Wan, G.S.; Ramanjaneyulu, G.S.; Cavallari, M.; El Daker, S.; Beddoe, T.; Theodossis, A.; Williams, N.K.; Gostick, E.; et al. Butyrophilin 3A1 binds phosphorylated antigens and stimulates human gammadelta T cells. Nat. Immunol. 2013, 14, 908–916. [Google Scholar] [CrossRef]
  51. Gu, S.; Borowska, M.T.; Boughter, C.T.; Adams, E.J. Butyrophilin3A proteins and Vγ9Vδ2 T cell activation. Semin. Cell. Dev. Biol. 2018, 84, 65–74. [Google Scholar] [CrossRef]
  52. Arnett, H.A.; Viney, J.L. Immune modulation by butyrophilins. Nat. Rev. Immunol. 2014, 14, 559–569. [Google Scholar] [CrossRef]
  53. Jeong, J.; Rao, A.U.; Xu, J.; Ogg, S.L.; Hathout, Y.; Fenselau, C.; Mather, I.H. The PRY/SPRY/B30.2 domain of butyrophilin 1A1 (BTN1A1) binds to xanthine oxidoreductase: Implications for the function of BTN1A1 in the mammary gland and other tissues. J. Biol. Chem. 2009, 284, 22444–22456. [Google Scholar] [CrossRef] [Green Version]
  54. Gu, S.; Sachleben, J.R.; Boughter, C.T.; Nawrocka, W.I.; Borowska, M.T.; Tarrasch, J.T.; Skiniotis, G.; Roux, B.; Adams, E.J. Phosphoantigen-induced conformational change of butyrophilin 3A1 (BTN3A1) and its implication on Vγ9Vδ2 T cell activation. Proc. Natl. Acad. Sci. USA 2017, 114, E7311–E7320. [Google Scholar] [CrossRef] [Green Version]
  55. Yang, Y.; Li, L.; Yuan, L.; Zhou, X.; Duan, J.; Xiao, H.; Cai, N.; Han, S.; Ma, X.; Liu, W.; et al. A Structural Change in Butyrophilin upon Phosphoantigen Binding Underlies Phosphoantigen-Mediated Vγ9Vδ2 T Cell Activation. Immunity 2019, 50, 1043–1053.e1045. [Google Scholar] [CrossRef] [Green Version]
  56. Palakodeti, A.; Sandstrom, A.; Sundaresan, L.; Harly, C.; Nedellec, S.; Olive, D.; Scotet, E.; Bonneville, M.; Adams, E.J. The molecular basis for modulation of human Vγ9Vδ2 T cell responses by CD277/butyrophilin-3 (BTN3A)-specific antibodies. J. Biol. Chem. 2012, 287, 32780–32790. [Google Scholar] [CrossRef] [Green Version]
  57. Wang, H.; Henry, O.; Distefano, M.D.; Wang, Y.C.; Räikkönen, J.; Mönkkönen, J.; Tanaka, Y.; Morita, C.T. Butyrophilin 3A1 plays an essential role in prenyl pyrophosphate stimulation of human Vγ2Vδ2 T cells. J. Immunol. 2013, 191, 1029–1042. [Google Scholar] [CrossRef] [Green Version]
  58. De Gassart, A.; Le, K.S.; Brune, P.; Agaugué, 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. 2021, 13, eabj0835. [Google Scholar] [CrossRef]
  59. Soriano-Sarabia, N.; Sandvold, H.; Jomaa, H.; Kubin, T.; Bein, G.; Hackstein, H. Primary MHC-class II+ cells are necessary to promote resting Vδ2 cell expansion in response to (E)-4-hydroxy-3-methyl-but-2-enyl-pyrophosphate and isopentenyl pyrophosphate. J. Immunol. 2012, 189, 5212–5222. [Google Scholar] [CrossRef] [Green Version]
  60. Landin, A.M.; Cox, C.; Yu, B.; Bejanyan, N.; Davila, M.; Kelley, L. Expansion and Enrichment of Gamma-Delta (γδ) T Cells from Apheresed Human Product. J. Vis. Exp. 2021, e62622. [Google Scholar] [CrossRef]
  61. Caccamo, N.; Dieli, F.; Wesch, D.; Jomaa, H.; Eberl, M. Sex-specific phenotypical and functional differences in peripheral human Vgamma9/Vdelta2 T cells. J. Leukoc. Biol. 2006, 79, 663–666. [Google Scholar] [CrossRef]
  62. Cairo, C.; Armstrong, C.L.; Cummings, J.S.; Deetz, C.O.; Tan, M.; Lu, C.; Davis, C.E.; Pauza, C.D. Impact of age, gender, and race on circulating γδ T cells. Hum. Immunol. 2010, 71, 968–975. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Baker, F.L.; Bigley, A.B.; Agha, N.H.; Pedlar, C.R.; O’Connor, D.P.; Bond, R.A.; Bollard, C.M.; Katsanis, E.; Simpson, R.J. Systemic β-Adrenergic Receptor Activation Augments the ex vivo Expansion and Anti-Tumor Activity of Vγ9Vδ2 T-Cells. Front. Immunol. 2019, 10, 3082. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Clohosey, M.L.; Mann, B.T.; Ryan, P.L.; Apanasovich, T.V.; Maggirwar, S.B.; Pennington, D.J.; Soriano-Sarabia, N. Comparable Vδ2 Cell Functional Characteristics in Virally Suppressed People Living with HIV and Uninfected Individuals. Cells 2020, 9, 2568. [Google Scholar] [CrossRef] [PubMed]
  65. Burnham, R.E.; Zoine, J.T.; Story, J.Y.; Garimalla, S.N.; Gibson, G.; Rae, A.; Williams, E.; Bixby, L.; Archer, D.; Doering, C.B.; et al. Characterization of Donor Variability for γδ T Cell ex vivo Expansion and Development of an Allogeneic γδ T Cell Immunotherapy. Front. Med. 2020, 7, 588453. [Google Scholar] [CrossRef] [PubMed]
  66. Poccia, F.; Gioia, C.; Martini, F.; Sacchi, A.; Piacentini, P.; Tempestilli, M.; Agrati, C.; Amendola, A.; Abdeddaim, A.; Vlassi, C.; et al. Zoledronic acid and interleukin-2 treatment improves immunocompetence in HIV-infected persons by activating Vgamma9Vdelta2 T cells. Aids 2009, 23, 555–565. [Google Scholar] [CrossRef]
  67. Poonia, B. Adoptive transfer of aminobisphonate-expanded Vgamma9Vdelta2+ T cells does not control HIV replication in a humanized mouse model. Immunotherapy 2016, 8, 521–526. [Google Scholar] [CrossRef] [Green Version]
  68. Poonia, B.; Pauza, C.D. Gamma delta T cells from HIV+ donors can be expanded in vitro by zoledronate/interleukin-2 to become cytotoxic effectors for antibody-dependent cellular cytotoxicity. Cytotherapy 2012, 14, 173–181. [Google Scholar] [CrossRef]
  69. Wallace, M.; Bartz, S.R.; Chang, W.L.; Mackenzie, D.A.; Pauza, C.D.; Malkovsky, M. Gamma delta T lymphocyte responses to HIV. Clin. Exp. Immunol. 1996, 103, 177–184. [Google Scholar] [CrossRef]
  70. Ali, Z.; Yan, L.; Plagman, N.; Reichenberg, A.; Hintz, M.; Jomaa, H.; Villinger, F.; Chen, Z.W. Gammadelta T cell immune manipulation during chronic phase of simian-human immunodeficiency virus infection [corrected] confers immunological benefits. J. Immunol. 2009, 183, 5407–5417. [Google Scholar] [CrossRef]
  71. Garrido, C.; Clohosey, M.L.; Whitworth, C.P.; Hudgens, M.; Margolis, D.M.; Soriano-Sarabia, N. γδ T cells: An immunotherapeutic approach for HIV cure strategies. JCI Insight 2018, 3, e120121. [Google Scholar] [CrossRef]
  72. James, K.S.; Trumble, I.; Clohosey, M.L.; Moeser, M.; Roan, N.R.; Adimora, A.A.; Joseph, S.B.; Archin, N.M.; Hudgens, M.; Soriano-Sarabia, N. Measuring the contribution of gammadelta T cells to the persistent HIV reservoir. AIDS 2020, 34, 363–371. [Google Scholar] [CrossRef]
  73. Archin, N.M.; Sung, J.M.; Garrido, C.; Soriano-Sarabia, N.; Margolis, D.M. Eradicating HIV-1 infection: Seeking to clear a persistent pathogen. Nat. Rev. Microbiol. 2014, 12, 750–764. [Google Scholar] [CrossRef] [Green Version]
  74. Board, N.L.; Moskovljevic, M.; Wu, F.; Siliciano, R.F.; Siliciano, J.D. Engaging innate immunity in HIV-1 cure strategies. Nat. Rev. Immunol. 2021, 22, 499–512. [Google Scholar] [CrossRef]
  75. Fletcher, C.V.; Dyavar, S.R.; Acharya, A.; Byrareddy, S.N. The Contributions of Clinical Pharmacology to HIV Cure Research. Clin. Pharmacol. Ther. 2021, 110, 334–345. [Google Scholar] [CrossRef]
  76. Mann, B.T.; Sambrano, E., 3rd; Maggirwar, S.B.; Soriano-Sarabia, N. Boosting the Immune System for HIV Cure: A γδ T Cell Perspective. Front. Cell. Infect. Microbiol. 2020, 10, 221. [Google Scholar] [CrossRef]
  77. Bricker, K.M.; Chahroudi, A.; Mavigner, M. New Latency Reversing Agents for HIV-1 Cure: Insights from Nonhuman Primate Models. Viruses 2021, 13, 1560. [Google Scholar] [CrossRef]
  78. Chen, M.; Li, M.; Budai, M.M.; Rice, A.P.; Kimata, J.T.; Mohan, M.; Wang, J. Clearance of HIV-1 or SIV reservoirs by promotion of apoptosis and inhibition of autophagy: Targeting intracellular molecules in cure-directed strategies. J. Leukoc. Biol. 2022, 112, 1245–1259. [Google Scholar] [CrossRef]
  79. Caron, J.; Ridgley, L.A.; Bodman-Smith, M. How to Train Your Dragon: Harnessing Gamma Delta T Cells Antiviral Functions and Trained Immunity in a Pandemic Era. Front. Immunol. 2021, 12, 666983. [Google Scholar] [CrossRef]
  80. Wang, T.; Gao, Y.; Scully, E.; Davis, C.T.; Anderson, J.F.; Welte, T.; Ledizet, M.; Koski, R.; Madri, J.A.; Barrett, A.; et al. Gamma delta T cells facilitate adaptive immunity against West Nile virus infection in mice. J. Immunol. 2006, 177, 1825–1832. [Google Scholar] [CrossRef]
  81. Agrati, C.; Castilletti, C.; Cimini, E.; Romanelli, A.; Lapa, D.; Quartu, S.; Martini, F.; Capobianchi, M.R. Antiviral activity of human Vδ2 T-cells against WNV includes both cytolytic and non-cytolytic mechanisms. New Microbiol. 2016, 39, 139–142. [Google Scholar] [PubMed]
  82. Agrati, C.; Alonzi, T.; De Santis, R.; Castilletti, C.; Abbate, I.; Capobianchi, M.R.; D’Offizi, G.; Siepi, F.; Fimia, G.M.; Tripodi, M.; et al. Activation of Vgamma9Vdelta2 T cells by non-peptidic antigens induces the inhibition of subgenomic HCV replication. Int. Immunol. 2006, 18, 11–18. [Google Scholar] [CrossRef] [PubMed]
  83. Cimini, E.; Bonnafous, C.; Bordoni, V.; Lalle, E.; Sicard, H.; Sacchi, A.; Berno, G.; Gioia, C.; D’Offizi, G.; Visco Comandini, U.; et al. Interferon-α improves phosphoantigen-induced Vγ9Vδ2 T-cells interferon-γ production during chronic HCV infection. PLoS ONE 2012, 7, e37014. [Google Scholar] [CrossRef] [PubMed]
  84. Chen, C.Y.; Yao, S.; Huang, D.; Wei, H.; Sicard, H.; Zeng, G.; Jomaa, H.; Larsen, M.H.; Jacobs, W.R., Jr.; Wang, R.; et al. Phosphoantigen/IL2 expansion and differentiation of Vγ2Vδ2 T cells increase resistance to tuberculosis in nonhuman primates. PLoS Pathog. 2013, 9, e1003501. [Google Scholar] [CrossRef] [PubMed]
  85. Qaqish, A.; Huang, D.; Chen, C.Y.; Zhang, Z.; Wang, R.; Li, S.; Yang, E.; Lu, Y.; Larsen, M.H.; Jacobs, W.R., Jr.; et al. Adoptive Transfer of Phosphoantigen-Specific γδ T Cell Subset Attenuates Mycobacterium tuberculosis Infection in Nonhuman Primates. J. Immunol. 2017, 198, 4753–4763. [Google Scholar] [CrossRef] [Green Version]
  86. Mazzola, T.N.; Da Silva, M.T.; Moreno, Y.M.; Lima, S.C.; Carniel, E.F.; Morcillo, A.M.; Antonio, M.A.; Zanolli, M.L.; Netto, A.A.; Blotta, M.H.; et al. Robust gammadelta+ T cell expansion in infants immunized at birth with BCG vaccine. Vaccine 2007, 25, 6313–6320. [Google Scholar] [CrossRef]
  87. Zufferey, C.; Germano, S.; Dutta, B.; Ritz, N.; Curtis, N. The contribution of non-conventional T cells and NK cells in the mycobacterial-specific IFNγ response in Bacille Calmette-Guérin (BCG)-immunized infants. PLoS ONE 2013, 8, e77334. [Google Scholar] [CrossRef]
  88. Shen, L.; Frencher, J.; Huang, D.; Wang, W.; Yang, E.; Chen, C.Y.; Zhang, Z.; Wang, R.; Qaqish, A.; Larsen, M.H.; et al. Immunization of Vγ2Vδ2 T cells programs sustained effector memory responses that control tuberculosis in nonhuman primates. Proc. Natl. Acad. Sci. USA 2019, 116, 6371–6378. [Google Scholar] [CrossRef] [Green Version]
  89. Hoft, D.F.; Brown, R.M.; Roodman, S.T. Bacille Calmette-Guérin vaccination enhances human gamma delta T cell responsiveness to mycobacteria suggestive of a memory-like phenotype. J. Immunol. 1998, 161, 1045–1054. [Google Scholar]
  90. Girardi, M.; Oppenheim, D.E.; Steele, C.R.; Lewis, J.M.; Glusac, E.; Filler, R.; Hobby, P.; Sutton, B.; Tigelaar, R.E.; Hayday, A.C. Regulation of cutaneous malignancy by gammadelta T cells. Science 2001, 294, 605–609. [Google Scholar] [CrossRef]
  91. Tosolini, M.; Pont, F.; Poupot, M.; Vergez, F.; Nicolau-Travers, M.L.; Vermijlen, D.; Sarry, J.E.; Dieli, F.; Fournie, J.J. Assessment of tumor-infiltrating TCRVgamma9Vdelta2 gammadelta lymphocyte abundance by deconvolution of human cancers microarrays. Oncoimmunology 2017, 6, e1284723. [Google Scholar] [CrossRef]
  92. Poccia, F.; Agrati, C.; Martini, F.; Capobianchi, M.R.; Wallace, M.; Malkovsky, M. Antiviral reactivities of gammadelta T cells. Microbes Infect. 2005, 7, 518–528. [Google Scholar] [CrossRef]
  93. Lafont, V.; Sanchez, F.; Laprevotte, E.; Michaud, H.A.; Gros, L.; Eliaou, J.F.; Bonnefoy, N. Plasticity of gammadelta T Cells: Impact on the Anti-Tumor Response. Front. Immunol. 2014, 5, 622. [Google Scholar] [CrossRef] [Green Version]
  94. Zou, C.; Zhao, P.; Xiao, Z.; Han, X.; Fu, F.; Fu, L. gammadelta T cells in cancer immunotherapy. Oncotarget 2017, 8, 8900–8909. [Google Scholar] [CrossRef] [Green Version]
  95. Zhao, Y.; Niu, C.; Cui, J. Gamma-delta (γδ) T cells: Friend or foe in cancer development? J. Transl. Med. 2018, 16, 3. [Google Scholar] [CrossRef] [Green Version]
  96. Silva-Santos, B.; Mensurado, S.; Coffelt, S.B. γδ T cells: Pleiotropic immune effectors with therapeutic potential in cancer. Nat. Rev. Cancer 2019, 19, 392–404. [Google Scholar] [CrossRef] [Green Version]
  97. Park, J.H.; Lee, H.K. Function of γδ T cells in tumor immunology and their application to cancer therapy. Exp. Mol. Med. 2021, 53, 318–327. [Google Scholar] [CrossRef]
  98. Shafi, S.; Vantourout, P.; Wallace, G.; Antoun, A.; Vaughan, R.; Stanford, M.; Hayday, A. An NKG2D-mediated human lymphoid stress surveillance response with high interindividual variation. Sci. Transl. Med. 2011, 3, 113ra124. [Google Scholar] [CrossRef] [Green Version]
  99. Correia, D.V.; Lopes, A.; Silva-Santos, B. Tumor cell recognition by γδ T lymphocytes: T-cell receptor vs. NK-cell receptors. Oncoimmunology 2013, 2, e22892. [Google Scholar] [CrossRef] [Green Version]
  100. Ochoa, M.C.; Minute, L.; Rodriguez, I.; Garasa, S.; Perez-Ruiz, E.; Inogés, S.; Melero, I.; Berraondo, P. Antibody-dependent cell cytotoxicity: Immunotherapy strategies enhancing effector NK cells. Immunol. Cell. Biol. 2017, 95, 347–355. [Google Scholar] [CrossRef]
  101. Tokuyama, H.; Hagi, T.; Mattarollo, S.R.; Morley, J.; Wang, Q.; So, H.F.; Moriyasu, F.; Nieda, M.; Nicol, A.J. V gamma 9 V delta 2 T cell cytotoxicity against tumor cells is enhanced by monoclonal antibody drugs--rituximab and trastuzumab. Int. J. Cancer 2008, 122, 2526–2534. [Google Scholar] [CrossRef] [PubMed]
  102. Kim, M.; Kim, H.; Han, M.; Hwang, H.J.; Kim, H.; Im, H.J.; Kim, N.; Koh, K.N. Characteristics of Human Peripheral Blood γδ T Cells Expanded With Zoledronate. Anticancer Res. 2021, 41, 6031–6038. [Google Scholar] [CrossRef] [PubMed]
  103. Maniar, A.; Zhang, X.; Lin, W.; Gastman, B.R.; Pauza, C.D.; Strome, S.E.; Chapoval, A.I. Human gammadelta T lymphocytes induce robust NK cell-mediated antitumor cytotoxicity through CD137 engagement. Blood 2010, 116, 1726–1733. [Google Scholar] [CrossRef] [PubMed]
  104. Brandes, M.; Willimann, K.; Bioley, G.; Lévy, N.; Eberl, M.; Luo, M.; Tampé, R.; Lévy, F.; Romero, P.; Moser, B. Cross-presenting human gammadelta T cells induce robust CD8+ alphabeta T cell responses. Proc. Natl. Acad. Sci. USA 2009, 106, 2307–2312. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Kunzmann, V.; Bauer, E.; Feurle, J.; Weissinger, F.; Tony, H.P.; Wilhelm, M. Stimulation of gammadelta T cells by aminobisphosphonates and induction of antiplasma cell activity in multiple myeloma. Blood 2000, 96, 384–392. [Google Scholar] [CrossRef]
  106. Wilhelm, M.; Kunzmann, V.; Eckstein, S.; Reimer, P.; Weissinger, F.; Ruediger, T.; Tony, H.P. Gammadelta T cells for immune therapy of patients with lymphoid malignancies. Blood 2003, 102, 200–206. [Google Scholar] [CrossRef] [Green Version]
  107. Dieli, F.; Vermijlen, D.; Fulfaro, F.; Caccamo, N.; Meraviglia, S.; Cicero, G.; Roberts, A.; Buccheri, S.; D’Asaro, M.; Gebbia, N.; et al. Targeting human γδ T cells with zoledronate and interleukin-2 for immunotherapy of hormone-refractory prostate cancer. Cancer Res. 2007, 67, 7450–7457. [Google Scholar] [CrossRef] [Green Version]
  108. Meraviglia, S.; Eberl, M.; Vermijlen, D.; Todaro, M.; Buccheri, S.; Cicero, G.; La Mendola, C.; Guggino, G.; D’Asaro, M.; Orlando, V.; et al. In vivo manipulation of Vgamma9Vdelta2 T cells with zoledronate and low-dose interleukin-2 for immunotherapy of advanced breast cancer patients. Clin. Exp. Immunol. 2010, 161, 290–297. [Google Scholar] [CrossRef]
  109. Lang, J.M.; Kaikobad, M.R.; Wallace, M.; Staab, M.J.; Horvath, D.L.; Wilding, G.; Liu, G.; Eickhoff, J.C.; McNeel, D.G.; Malkovsky, M. Pilot trial of interleukin-2 and zoledronic acid to augment gammadelta T cells as treatment for patients with refractory renal cell carcinoma. Cancer Immunol. Immunother. 2011, 60, 1447–1460. [Google Scholar] [CrossRef] [Green Version]
  110. Kunzmann, V.; Smetak, M.; Kimmel, B.; Weigang-Koehler, K.; Goebeler, M.; Birkmann, J.; Becker, J.; Schmidt-Wolf, I.G.; Einsele, H.; Wilhelm, M. Tumor-promoting versus tumor-antagonizing roles of gammadelta T cells in cancer immunotherapy: Results from a prospective phase I/II trial. J. Immunother. 2012, 35, 205–213. [Google Scholar] [CrossRef]
  111. Pressey, J.G.; Adams, J.; Harkins, L.; Kelly, D.; You, Z.; Lamb, L.S., Jr. In vivo expansion and activation of gammadelta T cells as immunotherapy for refractory neuroblastoma: A phase 1 study. Medicine 2016, 95, e4909. [Google Scholar] [CrossRef] [PubMed]
  112. Kobayashi, H.; Tanaka, Y.; Yagi, J.; Osaka, Y.; Nakazawa, H.; Uchiyama, T.; Minato, N.; Toma, H. Safety profile and anti-tumor effects of adoptive immunotherapy using gamma-delta T cells against advanced renal cell carcinoma: A pilot study. Cancer Immunol. Immunother. 2007, 56, 469–476. [Google Scholar] [CrossRef] [PubMed]
  113. Bennouna, J.; Levy, V.; Sicard, H.; Senellart, H.; Audrain, M.; Hiret, S.; Rolland, F.; Bruzzoni-Giovanelli, H.; Rimbert, M.; Galea, C.; et al. Phase I study of bromohydrin pyrophosphate (BrHPP, IPH 1101), a Vgamma9Vdelta2 T lymphocyte agonist in patients with solid tumors. Cancer Immunol. Immunother. 2010, 59, 1521–1530. [Google Scholar] [CrossRef] [PubMed]
  114. Bennouna, J.; Bompas, E.; Neidhardt, E.M.; Rolland, F.; Philip, I.; Galéa, C.; Salot, S.; Saiagh, S.; Audrain, M.; Rimbert, M.; et al. Phase-I study of Innacell gammadelta, an autologous cell-therapy product highly enriched in gamma9delta2 T lymphocytes, in combination with IL-2, in patients with metastatic renal cell carcinoma. Cancer Immunol. Immunother. 2008, 57, 1599–1609. [Google Scholar] [CrossRef] [PubMed]
  115. Benzaïd, I.; Mönkkönen, H.; Bonnelye, E.; Mönkkönen, J.; Clézardin, P. In vivo phosphoantigen levels in bisphosphonate-treated human breast tumors trigger Vγ9Vδ2 T-cell antitumor cytotoxicity through ICAM-1 engagement. Clin. Cancer Res. 2012, 18, 6249–6259. [Google Scholar] [CrossRef] [Green Version]
  116. Abe, Y.; Muto, M.; Nieda, M.; Nakagawa, Y.; Nicol, A.; Kaneko, T.; Goto, S.; Yokokawa, K.; Suzuki, K. Clinical and immunological evaluation of zoledronate-activated Vgamma9gammadelta T-cell-based immunotherapy for patients with multiple myeloma. Exp. Hematol. 2009, 37, 956–968. [Google Scholar] [CrossRef]
  117. Kobayashi, H.; Tanaka, Y.; Shimmura, H.; Minato, N.; Tanabe, K. Complete remission of lung metastasis following adoptive immunotherapy using activated autologous gammadelta T-cells in a patient with renal cell carcinoma. Anticancer Res. 2010, 30, 575–579. [Google Scholar]
  118. Nakajima, J.; Murakawa, T.; Fukami, T.; Goto, S.; Kaneko, T.; Yoshida, Y.; Takamoto, S.; Kakimi, K. A phase I study of adoptive immunotherapy for recurrent non-small-cell lung cancer patients with autologous gammadelta T cells. Eur. J. Cardiothorac. Surg. 2010, 37, 1191–1197. [Google Scholar] [CrossRef] [Green Version]
  119. Kobayashi, H.; Tanaka, Y.; Nakazawa, H.; Yagi, J.; Minato, N.; Tanabe, K. A new indicator of favorable prognosis in locally advanced renal cell carcinomas: Gamma delta T-cells in peripheral blood. Anticancer Res. 2011, 31, 1027–1031. [Google Scholar]
  120. Sakamoto, M.; Nakajima, J.; Murakawa, T.; Fukami, T.; Yoshida, Y.; Murayama, T.; Takamoto, S.; Matsushita, H.; Kakimi, K. Adoptive immunotherapy for advanced non-small cell lung cancer using zoledronate-expanded γδTcells: A phase I clinical study. J. Immunother. 2011, 34, 202–211. [Google Scholar] [CrossRef]
  121. Nicol, A.J.; Tokuyama, H.; Mattarollo, S.R.; Hagi, T.; Suzuki, K.; Yokokawa, K.; Nieda, M. Clinical evaluation of autologous gamma delta T cell-based immunotherapy for metastatic solid tumours. Br. J. Cancer 2011, 105, 778–786. [Google Scholar] [CrossRef] [Green Version]
  122. Fujimi, A.; Kamihara, Y.; Kanisawa, Y.; Hashimoto, A.; Nakajima, C.; Hayasaka, N.; Uemura, N.; Okuda, T.; Minami, S.; Iyama, S.; et al. Anti-erythropoietin receptor antibody-associated pure red cell aplasia accompanied by Coombs-negative autoimmune hemolytic anemia in a patient with T cell/histiocyte-rich large B cell lymphoma. Int. J. Hematol. 2014, 100, 490–493. [Google Scholar] [CrossRef]
  123. Aoki, T.; Matsushita, H.; Hoshikawa, M.; Hasegawa, K.; Kokudo, N.; Kakimi, K. Adjuvant combination therapy with gemcitabine and autologous γδ T-cell transfer in patients with curatively resected pancreatic cancer. Cytotherapy 2017, 19, 473–485. [Google Scholar] [CrossRef]
  124. Kakimi, K.; Matsushita, H.; Masuzawa, K.; Karasaki, T.; Kobayashi, Y.; Nagaoka, K.; Hosoi, A.; Ikemura, S.; Kitano, K.; Kawada, I.; et al. Adoptive transfer of zoledronate-expanded autologous Vγ9Vδ2 T-cells in patients with treatment-refractory non-small-cell lung cancer: A multicenter, open-label, single-arm, phase 2 study. J. Immunother. Cancer 2020, 8, e001185. [Google Scholar] [CrossRef]
  125. Lin, M.; Zhang, X.; Liang, S.; Luo, H.; Alnaggar, M.; Liu, A.; Yin, Z.; Chen, J.; Niu, L.; Jiang, Y. Irreversible electroporation plus allogenic Vγ9Vδ2 T cells enhances antitumor effect for locally advanced pancreatic cancer patients. Signal Transduct. Target. Ther. 2020, 5, 215. [Google Scholar] [CrossRef]
  126. Wilhelm, M.; Smetak, M.; Schaefer-Eckart, K.; Kimmel, B.; Birkmann, J.; Einsele, H.; Kunzmann, V. Successful adoptive transfer and in vivo expansion of haploidentical γδ T cells. J. Transl. Med. 2014, 12, 45. [Google Scholar] [CrossRef]
  127. Alnaggar, M.; Xu, Y.; Li, J.; He, J.; Chen, J.; Li, M.; Wu, Q.; Lin, L.; Liang, Y.; Wang, X.; et al. Allogenic Vγ9Vδ2 T cell as new potential immunotherapy drug for solid tumor: A case study for cholangiocarcinoma. J. Immunother. Cancer 2019, 7, 36. [Google Scholar] [CrossRef]
  128. Zhang, T.; Chen, J.; Niu, L.; Liu, Y.; Ye, G.; Jiang, M.; Qi, Z. Clinical Safety and Efficacy of Locoregional Therapy Combined with Adoptive Transfer of Allogeneic γδ T Cells for Advanced Hepatocellular Carcinoma and Intrahepatic Cholangiocarcinoma. J. Vasc. Interv. Radiol. 2022, 33, 19–27.e13. [Google Scholar] [CrossRef]
  129. Xu, Y.; Xiang, Z.; Alnaggar, M.; Kouakanou, L.; Li, J.; He, J.; Yang, J.; Hu, Y.; Chen, Y.; Lin, L.; et al. Allogeneic Vgamma9Vdelta2 T-cell immunotherapy exhibits promising clinical safety and prolongs the survival of patients with late-stage lung or liver cancer. Cell. Mol. Immunol. 2021, 18, 427–439. [Google Scholar] [CrossRef]
  130. Jhita, N.; Raikar, S.S. Allogeneic gamma delta T cells as adoptive cellular therapy for hematologic malignancies. Explor. Immunol. 2022, 2, 334–350. [Google Scholar] [CrossRef]
  131. Kang, N.; Zhou, J.; Zhang, T.; Wang, L.; Lu, F.; Cui, Y.; Cui, L.; He, W. Adoptive immunotherapy of lung cancer with immobilized anti-TCRgammadelta antibody-expanded human gammadelta T-cells in peripheral blood. Cancer Biol. Ther. 2009, 8, 1540–1549. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  132. Meidenbauer, N.; Marienhagen, J.; Laumer, M.; Vogl, S.; Heymann, J.; Andreesen, R.; Mackensen, A. Survival and tumor localization of adoptively transferred Melan-A-specific T cells in melanoma patients. J. Immunol. 2003, 170, 2161–2169. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  133. Ferrara, J.L.; Levine, J.E.; Reddy, P.; Holler, E. Graft-versus-host disease. Lancet 2009, 373, 1550–1561. [Google Scholar] [CrossRef]
  134. Lamb, L.S., Jr.; Henslee-Downey, P.J.; Parrish, R.S.; Godder, K.; Thompson, J.; Lee, C.; Gee, A.P. Increased frequency of TCR gamma delta + T cells in disease-free survivors following T cell-depleted, partially mismatched, related donor bone marrow transplantation for leukemia. J. Hematother. 1996, 5, 503–509. [Google Scholar] [CrossRef] [PubMed]
  135. Godder, K.T.; Henslee-Downey, P.J.; Mehta, J.; Park, B.S.; Chiang, K.Y.; Abhyankar, S.; Lamb, L.S. Long term disease-free survival in acute leukemia patients recovering with increased gammadelta T cells after partially mismatched related donor bone marrow transplantation. Bone Marrow Transplant. 2007, 39, 751–757. [Google Scholar] [CrossRef] [Green Version]
  136. Chauvin, C.; Joalland, N.; Perroteau, J.; Jarry, U.; Lafrance, L.; Willem, C.; Retière, C.; Oliver, L.; Gratas, C.; Gautreau-Rolland, L.; et al. NKG2D Controls Natural Reactivity of Vγ9Vδ2 T Lymphocytes against Mesenchymal Glioblastoma Cells. Clin. Cancer Res. 2019, 25, 7218–7228. [Google Scholar] [CrossRef] [Green Version]
  137. Ravens, S.; Fichtner, A.S.; Willers, M.; Torkornoo, D.; Pirr, S.; Schoning, J.; Deseke, M.; Sandrock, I.; Bubke, A.; Wilharm, A.; et al. Microbial exposure drives polyclonal expansion of innate gammadelta T cells immediately after birth. Proc. Natl. Acad. Sci. USA 2020, 117, 18649–18660. [Google Scholar] [CrossRef]
  138. Dimova, T.; Brouwer, M.; Gosselin, F.; Tassignon, J.; Leo, O.; Donner, C.; Marchant, A.; Vermijlen, D. Effector Vγ9Vδ2 T cells dominate the human fetal γδ T-cell repertoire. Proc. Natl. Acad. Sci. USA 2015, 112, E556–E565. [Google Scholar] [CrossRef] [Green Version]
  139. Davey, M.S.; Willcox, C.R.; Hunter, S.; Kasatskaya, S.A.; Remmerswaal, E.B.M.; Salim, M.; Mohammed, F.; Bemelman, F.J.; Chudakov, D.M.; Oo, Y.H.; et al. The human Vδ2+ T-cell compartment comprises distinct innate-like Vγ9+ and adaptive Vγ9 subsets. Nat. Commun. 2018, 9, 1760. [Google Scholar] [CrossRef]
  140. Van Acker, H.H.; Anguille, S.; Willemen, Y.; Van den Bergh, J.M.; Berneman, Z.N.; Lion, E.; Smits, E.L.; Van Tendeloo, V.F. Interleukin-15 enhances the proliferation, stimulatory phenotype, and antitumor effector functions of human gamma delta T cells. J. Hematol. Oncol. 2016, 9, 101. [Google Scholar] [CrossRef] [Green Version]
  141. Capsomidis, A.; Benthall, G.; Van Acker, H.H.; Fisher, J.; Kramer, A.M.; Abeln, Z.; Majani, Y.; Gileadi, T.; Wallace, R.; Gustafsson, K.; et al. Chimeric Antigen Receptor-Engineered Human Gamma Delta T Cells: Enhanced Cytotoxicity with Retention of Cross Presentation. Mol. Ther. 2018, 26, 354–365. [Google Scholar] [CrossRef] [Green Version]
  142. Mariani, S.; Muraro, M.; Pantaleoni, F.; Fiore, F.; Nuschak, B.; Peola, S.; Foglietta, M.; Palumbo, A.; Coscia, M.; Castella, B.; et al. Effector gammadelta T cells and tumor cells as immune targets of zoledronic acid in multiple myeloma. Leukemia 2005, 19, 664–670. [Google Scholar] [CrossRef]
  143. Fisher, J.P.; Yan, M.; Heuijerjans, J.; Carter, L.; Abolhassani, A.; Frosch, J.; Wallace, R.; Flutter, B.; Capsomidis, A.; Hubank, M.; et al. Neuroblastoma killing properties of Vδ2 and Vδ2-negative γδT cells following expansion by artificial antigen-presenting cells. Clin. Cancer Res. 2014, 20, 5720–5732. [Google Scholar] [CrossRef]
  144. Kondo, M.; Izumi, T.; Fujieda, N.; Kondo, A.; Morishita, T.; Matsushita, H.; Kakimi, K. Expansion of human peripheral blood γδ T cells using zoledronate. J. Vis. Exp. 2011, e3182. [Google Scholar] [CrossRef] [Green Version]
  145. Khan, M.W.A.; Otaibi, A.A.; Sherwani, S.; Alshammari, E.M.; Al-Zahrani, S.A.; Khan, W.A.; Alsukaibi, A.K.D.; Alouffi, S.; Khan, S.N. Optimization of methods for peripheral blood mononuclear cells isolation and expansion of human gamma delta T cells. Bioinformation 2021, 17, 460–469. [Google Scholar] [CrossRef]
  146. Schilbach, K.; Krickeberg, N.; Kaißer, C.; Mingram, S.; Kind, J.; Siegers, G.M.; Hashimoto, H. Suppressive activity of Vδ2+ γδ T cells on αβ T cells is licensed by TCR signaling and correlates with signal strength. Cancer Immunol. Immunother. 2020, 69, 593–610. [Google Scholar] [CrossRef] [Green Version]
  147. Duault, C.; Franchini, D.M.; Familliades, J.; Cayrol, C.; Roga, S.; Girard, J.P.; Fournié, J.J.; Poupot, M. TCRVγ9 γδ T Cell Response to IL-33: A CD4 T Cell-Dependent Mechanism. J. Immunol. 2016, 196, 493–502. [Google Scholar] [CrossRef] [Green Version]
  148. Jonus, H.C.; Burnham, R.E.; Ho, A.; Pilgrim, A.A.; Shim, J.; Doering, C.B.; Spencer, H.T.; Goldsmith, K.C. Dissecting the cellular components of ex vivo γδ T cell expansions to optimize selection of potent cell therapy donors for neuroblastoma immunotherapy trials. Oncoimmunology 2022, 11, 2057012. [Google Scholar] [CrossRef]
  149. Kouakanou, L.; Peters, C.; Sun, Q.; Floess, S.; Bhat, J.; Huehn, J.; Kabelitz, D. Vitamin C supports conversion of human gammadelta T cells into FOXP3-expressing regulatory cells by epigenetic regulation. Sci. Rep. 2020, 10, 6550. [Google Scholar] [CrossRef] [Green Version]
  150. Schilbach, K.; Welker, C.; Krickeberg, N.; Kaisser, C.; Schleicher, S.; Hashimoto, H. In the Absence of a TCR Signal IL-2/IL-12/18-Stimulated gammadelta T Cells Demonstrate Potent Anti-Tumoral Function Through Direct Killing and Senescence Induction in Cancer Cells. Cancers 2020, 12, 130. [Google Scholar] [CrossRef] [Green Version]
  151. Domae, E.; Hirai, Y.; Ikeo, T.; Goda, S.; Shimizu, Y. Cytokine-mediated activation of human ex vivo-expanded Vgamma9Vdelta2 T cells. Oncotarget 2017, 8, 45928–45942. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  152. García, V.E.; Jullien, D.; Song, M.; Uyemura, K.; Shuai, K.; Morita, C.T.; Modlin, R.L. IL-15 enhances the response of human gamma delta T cells to nonpeptide [correction of nonpetide] microbial antigens. J. Immunol. 1998, 160, 4322–4329. [Google Scholar] [PubMed]
  153. Li, W.; Kubo, S.; Okuda, A.; Yamamoto, H.; Ueda, H.; Tanaka, T.; Nakamura, H.; Yamanishi, H.; Terada, N.; Okamura, H. Effect of IL-18 on expansion of gammadelta T cells stimulated by zoledronate and IL-2. J. Immunother. 2010, 33, 287–296. [Google Scholar] [CrossRef] [PubMed]
  154. Nussbaumer, O.; Gruenbacher, G.; Gander, H.; Komuczki, J.; Rahm, A.; Thurnher, M. Essential requirements of zoledronate-induced cytokine and γδ T cell proliferative responses. J. Immunol. 2013, 191, 1346–1355. [Google Scholar] [CrossRef] [Green Version]
  155. Hudspeth, K.; Fogli, M.; Correia, D.V.; Mikulak, J.; Roberto, A.; Della Bella, S.; Silva-Santos, B.; Mavilio, D. Engagement of NKp30 on Vδ1 T cells induces the production of CCL3, CCL4, and CCL5 and suppresses HIV-1 replication. Blood 2012, 119, 4013–4016. [Google Scholar] [CrossRef] [Green Version]
  156. Correia, D.V.; Fogli, M.; Hudspeth, K.; da Silva, M.G.; Mavilio, D.; Silva-Santos, B. Differentiation of human peripheral blood Vδ1+ T cells expressing the natural cytotoxicity receptor NKp30 for recognition of lymphoid leukemia cells. Blood 2011, 118, 992–1001. [Google Scholar] [CrossRef] [Green Version]
  157. Mikulak, J.; Oriolo, F.; Bruni, E.; Roberto, A.; Colombo, F.S.; 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 2019, 4, e125884. [Google Scholar] [CrossRef] [Green Version]
  158. von Lilienfeld-Toal, M.; Nattermann, J.; Feldmann, G.; Sievers, E.; Frank, S.; Strehl, J.; Schmidt-Wolf, I.G. Activated gammadelta T cells express the natural cytotoxicity receptor natural killer p 44 and show cytotoxic activity against myeloma cells. Clin. Exp. Immunol. 2006, 144, 528–533. [Google Scholar] [CrossRef]
  159. Groh, V.; Steinle, A.; Bauer, S.; Spies, T. Recognition of stress-induced MHC molecules by intestinal epithelial gammadelta T cells. Science 1998, 279, 1737–1740. [Google Scholar] [CrossRef]
  160. Chalupny, N.J.; Sutherland, C.L.; Lawrence, W.A.; Rein-Weston, A.; Cosman, D. ULBP4 is a novel ligand for human NKG2D. Biochem. Biophys. Res. Commun. 2003, 305, 129–135. [Google Scholar] [CrossRef]
  161. Bauer, S.; Groh, V.; Wu, J.; Steinle, A.; Phillips, J.H.; Lanier, L.L.; Spies, T. Activation of NK cells and T cells by NKG2D, a receptor for stress-inducible MICA. Science 1999, 285, 727–729. [Google Scholar] [CrossRef]
  162. Lamb, L.S., Jr.; Bowersock, J.; Dasgupta, A.; Gillespie, G.Y.; Su, Y.; Johnson, A.; Spencer, H.T. Engineered drug resistant γδ T cells kill glioblastoma cell lines during a chemotherapy challenge: A strategy for combining chemo- and immunotherapy. PLoS ONE 2013, 8, e51805. [Google Scholar] [CrossRef]
  163. Shimizu, T.; Tomogane, M.; Miyashita, M.; Ukimura, O.; Ashihara, E. Low dose gemcitabine increases the cytotoxicity of human Vγ9Vδ2 T cells in bladder cancer cells in vitro and in an orthotopic xenograft model. Oncoimmunology 2018, 7, e1424671. [Google Scholar] [CrossRef]
  164. Bhat, J.; Dubin, S.; Dananberg, A.; Quabius, E.S.; Fritsch, J.; Dowds, C.M.; Saxena, A.; Chitadze, G.; Lettau, M.; Kabelitz, D. Histone Deacetylase Inhibitor Modulates NKG2D Receptor Expression and Memory Phenotype of Human Gamma/Delta T Cells Upon Interaction With Tumor Cells. Front. Immunol. 2019, 10, 569. [Google Scholar] [CrossRef] [Green Version]
  165. Joncker, N.T.; Fernandez, N.C.; Treiner, E.; Vivier, E.; Raulet, D.H. NK cell responsiveness is tuned commensurate with the number of inhibitory receptors for self-MHC class I: The rheostat model. J. Immunol. 2009, 182, 4572–4580. [Google Scholar] [CrossRef] [Green Version]
  166. Long, E.O.; Kim, H.S.; Liu, D.; Peterson, M.E.; Rajagopalan, S. Controlling natural killer cell responses: Integration of signals for activation and inhibition. Annu. Rev. Immunol. 2013, 31, 227–258. [Google Scholar] [CrossRef] [Green Version]
  167. Abd Hamid, M.; Wang, R.Z.; Yao, X.; Fan, P.; Li, X.; Chang, X.M.; Feng, Y.; Jones, S.; Maldonado-Perez, D.; Waugh, C.; et al. Enriched HLA-E and CD94/NKG2A Interaction Limits Antitumor CD8+ Tumor-Infiltrating T Lymphocyte Responses. Cancer Immunol. Res. 2019, 7, 1293–1306. [Google Scholar] [CrossRef] [Green Version]
  168. Chen, G.; Wu, D.; Guo, W.; Cao, Y.; Huang, D.; Wang, H.; Wang, T.; Zhang, X.; Chen, H.; Yu, H.; et al. Clinical and immunologic features in severe and moderate Coronavirus Disease 2019. J. Clin. Investig. 2020, 130, 2620–2629. [Google Scholar] [CrossRef] [Green Version]
  169. Gooden, M.; Lampen, M.; Jordanova, E.S.; Leffers, N.; Trimbos, J.B.; van der Burg, S.H.; Nijman, H.; van Hall, T. HLA-E expression by gynecological cancers restrains tumor-infiltrating CD8⁺ T lymphocytes. Proc. Natl. Acad. Sci. USA 2011, 108, 10656–10661. [Google Scholar] [CrossRef] [Green Version]
  170. Bossard, C.; Bézieau, S.; Matysiak-Budnik, T.; Volteau, C.; Laboisse, C.L.; Jotereau, F.; Mosnier, J.F. HLA-E/β2 microglobulin overexpression in colorectal cancer is associated with recruitment of inhibitory immune cells and tumor progression. Int. J. Cancer 2012, 131, 855–863. [Google Scholar] [CrossRef]
  171. Iwasaki, M.; Tanaka, Y.; Kobayashi, H.; Murata-Hirai, K.; Miyabe, H.; Sugie, T.; Toi, M.; Minato, N. Expression and function of PD-1 in human γδ T cells that recognize phosphoantigens. Eur. J. Immunol. 2011, 41, 345–355. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  172. Gogoi, D.; Biswas, D.; Borkakoty, B.; Mahanta, J. Exposure to Plasmodium vivax is associated with the increased expression of exhaustion markers on γδ T lymphocytes. Parasite Immunol. 2018, 40, e12594. [Google Scholar] [CrossRef] [PubMed]
  173. Girard, P.; Charles, J.; Cluzel, C.; Degeorges, E.; Manches, O.; Plumas, J.; De Fraipont, F.; Leccia, M.T.; Mouret, S.; Chaperot, L.; et al. The features of circulating and tumor-infiltrating γδ T cells in melanoma patients display critical perturbations with prognostic impact on clinical outcome. Oncoimmunology 2019, 8, 1601483. [Google Scholar] [CrossRef] [PubMed]
  174. Cimini, E.; Grassi, G.; Beccacece, A.; Casetti, R.; Castilletti, C.; Capobianchi, M.R.; Nicastri, E.; Agrati, C. In Acute Dengue Infection, High TIM-3 Expression May Contribute to the Impairment of IFNγ Production by Circulating Vδ2 T Cells. Viruses 2022, 14, 130. [Google Scholar] [CrossRef] [PubMed]
  175. Tawbi, H.A.; Schadendorf, D.; Lipson, E.J.; Ascierto, P.A.; Matamala, L.; Castillo Gutiérrez, E.; Rutkowski, P.; Gogas, H.J.; Lao, C.D.; De Menezes, J.J.; et al. Relatlimab and Nivolumab versus Nivolumab in Untreated Advanced Melanoma. N. Engl. J. Med. 2022, 386, 24–34. [Google Scholar] [CrossRef]
  176. Wei, S.C.; Duffy, C.R.; Allison, J.P. Fundamental Mechanisms of Immune Checkpoint Blockade Therapy. Cancer Discov 2018, 8, 1069–1086. [Google Scholar] [CrossRef] [Green Version]
  177. Hoeres, T.; Holzmann, E.; Smetak, M.; Birkmann, J.; Wilhelm, M. PD-1 signaling modulates interferon-γ production by Gamma Delta (γδ) T-Cells in response to leukemia. Oncoimmunology 2019, 8, 1550618. [Google Scholar] [CrossRef] [Green Version]
  178. Nada, M.H.; Wang, H.; Hussein, A.J.; Tanaka, Y.; Morita, C.T. PD-1 checkpoint blockade enhances adoptive immunotherapy by human Vγ2Vδ2 T cells against human prostate cancer. Oncoimmunology 2021, 10, 1989789. [Google Scholar] [CrossRef]
  179. Han, M.G.; Jang, B.S.; Kang, M.H.; Na, D.; Kim, I.A. PI3Kγδ inhibitor plus radiation enhances the antitumour immune effect of PD-1 blockade in syngenic murine breast cancer and humanised patient-derived xenograft model. Eur. J. Cancer 2021, 157, 450–463. [Google Scholar] [CrossRef]
  180. Li, X.; Lu, H.; Gu, Y.; Zhang, X.; Zhang, G.; Shi, T.; 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. 2020, 386, 111719. [Google Scholar] [CrossRef]
  181. Wistuba-Hamprecht, K.; Martens, A.; Haehnel, K.; Geukes Foppen, M.; Yuan, J.; Postow, M.A.; Wong, P.; Romano, E.; Khammari, A.; Dreno, B.; et al. Proportions of blood-borne Vδ1+ and Vδ2+ T-cells are associated with overall survival of melanoma patients treated with ipilimumab. Eur. J. Cancer 2016, 64, 116–126. [Google Scholar] [CrossRef] [Green Version]
  182. Zumwalde, N.A.; Sharma, A.; Xu, X.; Ma, S.; Schneider, C.L.; Romero-Masters, J.C.; Hudson, A.W.; Gendron-Fitzpatrick, A.; Kenney, S.C.; Gumperz, J.E. Adoptively transferred Vγ9Vδ2 T cells show potent antitumor effects in a preclinical B cell lymphomagenesis model. JCI Insight 2017, 2, e93179. [Google Scholar] [CrossRef] [Green Version]
  183. Guo, Q.; Zhao, P.; Zhang, Z.; Zhang, J.; Zhang, Z.; Hua, Y.; Han, B.; Li, N.; Zhao, X.; Hou, L. TIM-3 blockade combined with bispecific antibody MT110 enhances the anti-tumor effect of γδ T cells. Cancer Immunol. Immunother. 2020, 69, 2571–2587. [Google Scholar] [CrossRef]
  184. Akdis, M.; Burgler, S.; Crameri, R.; Eiwegger, T.; Fujita, H.; Gomez, E.; Klunker, S.; Meyer, N.; O’Mahony, L.; Palomares, O.; et al. Interleukins, from 1 to 37, and interferon-γ: Receptors, functions, and roles in diseases. J. Allergy Clin. Immunol. 2011, 127, 701–721.e70. [Google Scholar] [CrossRef]
  185. Taniguchi, T.; Matsui, H.; Fujita, T.; Takaoka, C.; Kashima, N.; Yoshimoto, R.; Hamuro, J. Structure and expression of a cloned cDNA for human interleukin-2. Nature 1983, 302, 305–310. [Google Scholar] [CrossRef]
  186. Rosenberg, S.A. IL-2: The first effective immunotherapy for human cancer. J. Immunol. 2014, 192, 5451–5458. [Google Scholar] [CrossRef] [Green Version]
  187. Pilipow, K.; Roberto, A.; Roederer, M.; Waldmann, T.A.; Mavilio, D.; Lugli, E. IL15 and T-cell Stemness in T-cell-Based Cancer Immunotherapy. Cancer Res. 2015, 75, 5187–5193. [Google Scholar] [CrossRef] [Green Version]
  188. Steel, J.C.; Waldmann, T.A.; Morris, J.C. Interleukin-15 biology and its therapeutic implications in cancer. Trends Pharmacol. Sci. 2012, 33, 35–41. [Google Scholar] [CrossRef] [Green Version]
  189. Viey, E.; Lucas, C.; Romagne, F.; Escudier, B.; Chouaib, S.; Caignard, A. Chemokine receptors expression and migration potential of tumor-infiltrating and peripheral-expanded Vgamma9Vdelta2 T cells from renal cell carcinoma patients. J. Immunother. 2008, 31, 313–323. [Google Scholar] [CrossRef]
  190. Wu, K.; Feng, J.; Xiu, Y.; Li, Z.; Lin, Z.; Zhao, H.; Zeng, H.; Xia, W.; Yu, L.; 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. 2020, 80, 106122. [Google Scholar] [CrossRef]
  191. Wu, K.; Zhao, H.; Xiu, Y.; Li, Z.; Zhao, J.; Xie, S.; Zeng, H.; Zhang, H.; Yu, L.; Xu, B. IL-21-mediated expansion of Vγ9Vδ2 T cells is limited by the Tim-3 pathway. Int. Immunopharmacol. 2019, 69, 136–142. [Google Scholar] [CrossRef] [PubMed]
  192. Okamura, H.; Tsutsi, H.; Komatsu, T.; Yutsudo, M.; Hakura, A.; Tanimoto, T.; Torigoe, K.; Okura, T.; Nukada, Y.; Hattori, K.; et al. Cloning of a new cytokine that induces IFN-gamma production by T cells. Nature 1995, 378, 88–91. [Google Scholar] [CrossRef] [PubMed]
  193. Dinarello, C.A. Interleukin-18 and the pathogenesis of inflammatory diseases. Semin. Nephrol. 2007, 27, 98–114. [Google Scholar] [CrossRef] [PubMed]
  194. Nakanishi, K.; Yoshimoto, T.; Tsutsui, H.; Okamura, H. Interleukin-18 regulates both Th1 and Th2 responses. Annu. Rev. Immunol. 2001, 19, 423–474. [Google Scholar] [CrossRef] [PubMed]
  195. Clark, D.A.; Coker, R. Transforming growth factor-beta (TGF-beta). Int. J. Biochem. Cell. Biol. 1998, 30, 293–298. [Google Scholar] [CrossRef]
  196. Peters, C.; Häsler, R.; Wesch, D.; Kabelitz, D. Human Vδ2 T cells are a major source of interleukin-9. Proc. Natl. Acad. Sci. USA 2016, 113, 12520–12525. [Google Scholar] [CrossRef] [Green Version]
  197. Siegers, G.M. Integral Roles for Integrins in γδ T Cell Function. Front. Immunol. 2018, 9, 521. [Google Scholar] [CrossRef]
  198. Beatson, R.E.; Parente-Pereira, A.C.; Halim, L.; Cozzetto, D.; Hull, C.; Whilding, L.M.; Martinez, O.; Taylor, C.A.; Obajdin, J.; Luu Hoang, K.N.; et al. TGF-beta1 potentiates Vgamma9Vdelta2 T cell adoptive immunotherapy of cancer. Cell Rep. Med. 2021, 2, 100473. [Google Scholar] [CrossRef]
  199. D’Aniello, C.; Cermola, F.; Patriarca, E.J.; Minchiotti, G. Vitamin C in Stem Cell Biology: Impact on Extracellular Matrix Homeostasis and Epigenetics. Stem Cells Int. 2017, 2017, 8936156. [Google Scholar] [CrossRef] [Green Version]
  200. Mastrangelo, D.; Pelosi, E.; Castelli, G.; Lo-Coco, F.; Testa, U. Mechanisms of anti-cancer effects of ascorbate: Cytotoxic activity and epigenetic modulation. Blood Cells Mol. Dis. 2018, 69, 57–64. [Google Scholar] [CrossRef]
  201. Tu, W.; Zheng, J.; Liu, Y.; Sia, S.F.; Liu, M.; Qin, G.; Ng, I.H.; Xiang, Z.; Lam, K.T.; Peiris, J.S.; et al. The aminobisphosphonate pamidronate controls influenza pathogenesis by expanding a gammadelta T cell population in humanized mice. J. Exp. Med. 2011, 208, 1511–1522. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Mevalonate pathway and Vδ2 T cell-mediated activation. (A) Statins inhibit IPP production whereas N-BPs induce IPP accumulation that is (B) specifically recognized by the Vδ2 TCR in the context of butyrophilin molecules. (Created with Biorender, accessed on 10 December 2022).
Figure 1. Mevalonate pathway and Vδ2 T cell-mediated activation. (A) Statins inhibit IPP production whereas N-BPs induce IPP accumulation that is (B) specifically recognized by the Vδ2 TCR in the context of butyrophilin molecules. (Created with Biorender, accessed on 10 December 2022).
Cells 11 03572 g001
Figure 2. Modulation of Vδ2 T cell pleiotropic function. (Created with Biorender, accessed on 10 December 2022).
Figure 2. Modulation of Vδ2 T cell pleiotropic function. (Created with Biorender, accessed on 10 December 2022).
Cells 11 03572 g002
Table 1. Summary of the clinical trials that used adoptive transfer of expanded Vδ2 T cells.
Table 1. Summary of the clinical trials that used adoptive transfer of expanded Vδ2 T cells.
Cancer TypeApproachNExpansion MethodRef.
AutologousAdvanced RCCAdoptive therapy7100 mM 2M3B1-PP + 100 U/mL IL-2, 14 days[112]
Advanced RCCAdoptive therapy + low dose IL-2103 mM BrHPP + 20–60 ng/mL IL-2, 14 days[114]
MMAdoptive therapy65 mM Zol + 1000 U/mL IL-2, 14 days[116]
RCC and multiple lung metastasisAdoptive therapy + Zol + low dose IL-214 mg Zol + 1.4 million unit of IL-2[117]
NSCLC*Autologous gd infusion105 mM Zol + 1000 U/mL IL-2, 14 days[118]
Advanced RCC and lung metastasisAdoptive therapy + Zol + low dose IL-241100 mM 2M3B1-PP + 100 U/mL IL-2, 11 days[119]
NSCLCAdoptive therapy155 mM Zol + 1000 U/mL IL-2, 14 days[120]
Advanced solid tumorsAdoptive therapy + Zol181 mM Zol + 700 U/mL IL-2[121]
GastricAdoptive therapy + Zol75 mM Zol + 1000 U/mL IL-2, 14 days[122]
PancreaticAdoptive therapy + Gemcitabine565 mM Zol + 1000 U/mL IL-2, 14 days[123]
NSCLCAdoptive therapy255 mM Zol + 1000 U/mL IL-2, 14 days[124]
Advanced pancreaticElectroporation + adoptive therapy6250 mM Zol + 10 ng/mL IL-2[125]
AllogeneicCancer typeApproachNExpansion methodRef.
Hematological malignanciesHaploidentical adoptive therapy4CD4 and CD8 depletion[126]
Cholangiocarcinoma, liver transplantedAdoptive therapy1patent pending: not shown in paper[127]
Advanced HCC (N = 30)/ICC (N = 29)Locoregional + adoptive therapy5950 mM Zol + 100 U/mL IL-2 + 100 U/mL IL-15 + 70 mM Vit C, 12–14 days[128]
Late-stage lung or liverAdoptive therapy1850 mM Zol + 100 U/mL IL-2 + 100 U/mL IL-15 + 70 mM Vit C, 12–14 days[129]
RCC: Renal cell cancer, NSCLC: non-small cell lung cancer, HCC: hepatocellular carcinoma, ICC: intrahepatic cholangiocarcinoma, MM: Multiple myeloma.
Table 2. Summary of basic and clinical studies using phosphoantigens or bisphosphonates and different cytokines for Vδ2 T cell expansion.
Table 2. Summary of basic and clinical studies using phosphoantigens or bisphosphonates and different cytokines for Vδ2 T cell expansion.
CKConcentrationpAg or BPAdditional StimuliConditionEffect in γδ T cellsTargetRef.
IL-250 U/mLZol (2.5 µM)TGF-ßPBMCsIFN, GrzA, GrzB, Perforin, TNF, adhesion moleculesPancreas and colon tumor cells[18]
100 U/mLSynthetic HMBPP-PBMCsIFN, TNF, IL5, IL-13n/a[28]
20 ng/mLHDMAPP-PBMCsProliferation, CD56, NKG2D/A, GrzA/B, PerforinRCC and Burkitt’s lymphoma[29]
100 U/mLIPP or ZOL-Isolated γδHLA-DR, CD69, CD56, CD16, IFNBurkitt’s lymphoma and MM[140]
100 U/mLZOL (40µg/mL)-PBMCsCD25, CD69, CD94, CD152, ICAM NKG2D, IFN, TNFHuman Neuroblastoma LAN 1[141]
10 U/mLZOL (1µM)-PBMCsIFN, TNFMM[142]
100 U/mLIPPaAPC, Anti-γδ TCR mAbPBMCs/Isolated γδNKG2D, cytotoxicityNeuroblastoma[143]
1000 U/mLZol (5 µM)-PBMCsNKG2D, CD69, IFNn/a[144]
100 U/mLZol (1µM)IL-15PBMCsCD69, HLA-DR, CD80, CD86n/a[145]
6.5 U/MLIPP or TGF-BAnti-γδ TCR mAbIsolated γδGrzB, perforin, CD107aGlioblastoma and melanoma[146]
400 U/mLBrHPP (100 nM)IL-33PBMCsCD16, CD28, NKG2A, NKG2D, Perforin, IFN, TNF, GrzBBurkitt´s lymphoma[147]
CXCR3, CD28, CCR5, Trail
15.2 ng/mLZol (5 µM)IL-15Isolated γδNKG2D, NKG2ANeuroblastoma cell lines[148]
50 U/mLBrHPP (300 nM)Vitamin C, TGF-ßIsolated γδProliferationn/a[149]
IL-1210 ng/mLIMMU510IL2,IL-15,IL-18Isolated γδIFN, TNF, GrzB, Perforinsarcoma, rhabdomyosarcoma, neuroblastoma[150]
10 ng/mL IL-18PBMCsIFNn/a[151]
IL-1510 ng/mLZol (2.5 µM)TGF-ßPBMCsIFN, GrzA, GrzB, Perforin, TNF, adhesion moleculesPancreas and colon tumor cells[18]
10 ng/mLHMBPP-PBMCsProliferation, TNF, IFN, CD16, CD94n/a[27]
12.5 ng/mLIPP or ZOL-Isolated γδCytotoxicity, HLA-DR, CD69, CD56, CD16, IFNBurkitt´s lymphoma and MM[140]
50 ng/mLIPP or TGF-BAnti-γδ TCR mAbIsolated γδGrzB, perforin, CD107aGlioblastoma and melanoma[146]
10 ng/mLIPP (30µM)IL-12PBMCsProliferation, IFN,n/a[152]
IL-1850 ng/mLZOL (1µM)IL-2PBMCsIFN, TNF, cytotoxicityMesothelioma[153]
200 ng/mLZol (10 or 30 µM)IL-1β, IL-2PBMCs/Isolated γδCytokine responsen/a[154]
IL-2110 ng/mLHMBPP-PBMCsProliferation, CD16, CD94n/a[27]
10 ng/mLSynthetic HMBPP-PBMCsCD25, CD27, CD69, NKG2Dn/a[28]
10 ng/mLHDMAPP-PBMCsProliferation, CD56, NKG2D, GrzA, GrzB, PerforinRCC and Burkitt´s lymphoma[29]
RCC: Renal cell carcinoma, MM: Multiple myeloma, n/a: not applicable.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Sanz, M.; Mann, B.T.; Chitrakar, A.; Soriano-Sarabia, N. Human Vδ2 T Cells and Their Versatility for Immunotherapeutic Approaches. Cells 2022, 11, 3572. https://doi.org/10.3390/cells11223572

AMA Style

Sanz M, Mann BT, Chitrakar A, Soriano-Sarabia N. Human Vδ2 T Cells and Their Versatility for Immunotherapeutic Approaches. Cells. 2022; 11(22):3572. https://doi.org/10.3390/cells11223572

Chicago/Turabian Style

Sanz, Marta, Brendan T. Mann, Alisha Chitrakar, and Natalia Soriano-Sarabia. 2022. "Human Vδ2 T Cells and Their Versatility for Immunotherapeutic Approaches" Cells 11, no. 22: 3572. https://doi.org/10.3390/cells11223572

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

Article Metrics

Back to TopTop