Abstract
Harnessing the potential of the immune system to treat cancers has been the goal of many scientific investigations and recent advancements in tumor immunology have allowed for cancer immunotherapy to become a reality. T lymphocytes that express the γδ TCR (γδ T cells) do not require antigen presentation by target cells. Instead, they recognize phospho-antigens that accumulate in tumors with increased activity of the mevalonate metabolic pathway. Additionally, the Natural Killer Group 2D (NKG2D) on γδ T cells recognizes stress-induced self-antigens widely expressed on cancer cells, such as the MHC Class I-like stress-associated molecules MIC-A and MIC-B or the UL-16 binding proteins ULBP-1, 2, and 3. This recognition can mediate direct cytotoxicity against tumor cells without prior antigen exposure or priming. Moreover, γδ T cells can be expanded when stimulated with IL-2 and Zoledronate. Collectively, these biological qualities of γδ T cells make them a promising option for cancer immunotherapy.
Keywords
- Gamma Delta T cell
- cancer immunotherapy
- immuno-oncology
- adoptive cell therapy
- CAR-T cell therapy
1. Introduction
Cancer has posed a formidable challenge to researchers for decades. Since the earliest days of cancer research, scientists have been striving to develop treatments that specifically target cancer cells while sparing the surrounding healthy tissues. Recent advancements in cancer biology and emerging technologies suggest that this goal is on the cusp of becoming a reality. At the forefront of these advancements is immunotherapy, which harnesses the body’s immune system to seek out and destroy cancer cells, leaving healthy tissues unharmed.
The underlying principle of immunotherapy is to leverage the body’s natural defense mechanisms to treat cancer more effectively. However, like many other treatments, immunotherapy has been met with variable success across different cancer types. When exposed to treatment pressures, many cancers evolve mechanisms to evade immune detection. Traditional immunotherapies often rely on Antigen-Presenting Cells (APCs) to prime αβ T lymphocytes to attack cancer cells specifically. This process involves recognizing Major Histocompatibility Complex (MHC) Class I molecules on the surface of cancerous cells. However, many cancers can downregulate the expression of MHC Class I molecules, evading αβ T lymphocytes and preventing these immune cells from exerting their cytotoxic effects [1]. Additionally, cancer cells can escape immune surveillance through various strategies, such as secreting the immunosuppressive cytokines Interleukin-10 (IL-10) and Transforming Growth Factor β (TGF-β) into the tumor microenvironment, dampening the immune response and leading to unchecked cancer growth [2, 3, 4]. This complex interplay between cancer cells and the immune system poses significant challenges to conventional immunotherapy strategies.
2. The biology of Gamma Delta T cell
To overcome challenges faced by the traditional cancer immunotherapy approaches, researchers have been exploring other creative solutions, including a subset of immune cells known as γδ T lymphocytes. These cells offer a promising approach to cancer immunotherapy as they do not rely on the traditional MHC Class I pathway for antigen recognition. γδ T lymphocytes are a unique subset of T cells characterized by their distinct γδ T-cell receptors (TCRs), which distinguish them from the more common αβ T cells. Instead of requiring antigen presentation from MHC Class I molecules, they are activated by conserved stress-induced self-antigens on cancer cells. The C-type lectin Natural Killer Group 2D (NKG2D) receptor on γδ T-cell recognizes these stress-induced ligands, such as MHC Class I-chain (MIC) molecules, MIC-A and MIC-B, along with UL-16 binding proteins ULBP-1, ULBP-2, and ULBP-3 on cancer cells directly and without the need for antigen presentation or priming [5]. Thus, granting γδ T lymphocytes the ability to elicit an immunological response akin to that of the innate immune system. Consequently, γδ T cells can identify and react to a wider variety of cancer cells, including those that have evolved defenses against other immune cell types. Moreover, as a subset of T cells, γδ T cells can also establish memory responses, enabling them to attack cancer cells upon subsequent exposures.
Human γδ T cells are divided into two subsets: Vδ1 and Vδ2 [6, 7]. The Vδ1 phenotype is primarily found in epithelial tissues, such as the skin, gut, and respiratory tract, where they play a critical role in immune surveillance, serving as first responders to cellular stress and transformation. Vδ1 T cells possess the C-type Natural Killer Group 2D (NKG2D) receptor, which recognizes stress-induced self-antigens, including MIC-A and MIC-B. These antigens are typically overexpressed on cancer cells, marking them for attack [8, 9, 10]. Stimulation of the NKG2D receptor, on γδ T cells, upon binding to these stress ligands, on cancer cells, can mediate direct cytotoxicity and lysis of tumor cells. Additionally, this interaction activates the release of anti-tumor cytokines, such as Interferon gamma (IFN-γ) and Tumor Necrosis Factor-α (TNF-α) [11, 12, 13].
Conversely, the second phenotype, Vδ2, is predominantly found in the peripheral blood. This subset typically binds with a second receptor, Vγ9, forming the Vγ9Vδ2 phenotype that circulates throughout the body. These cells can recognize non-peptidic phospho-antigens, such as isopentenyl pyrophosphate (IPP), which is a metabolite of the mevalonate metabolic pathway, crucial for cellular metabolism and proliferation. In normal cells, IPP concentrations are low, and these cells remain undetected by Vγ9Vδ2 cells. However, in cancer cells, the mevalonate metabolic pathway is often upregulated to meet increased energy demands, leading to IPP accumulation and its recognition as a tumor antigen [14]. This metabolic adaptation presents a distinct opportunity for Vγ9Vδ2 T cells, which can identify and eradicate tumor cells via both TCR-dependent and TCR-independent mechanisms. This unique ability makes Vγ9Vδ2 T cells an attractive option for immunotherapy, especially when tumors become resistant to other therapeutic modalities.
Despite the promising capabilities of γδ T cells, one of the current challenges in using them for immunotherapy is their relatively low abundance in the human body. Normally, Vγ9Vδ2 cells constitute about 1–5% of peripheral blood T cells, which can be even lower in cancer patients. To address this pitfall, protocols were developed to expand these cells ex vivo. One successful strategy involves using agonist molecules to stimulate their proliferation. Zoledronate, which is already an FDA-approved bisphosphonate used to prevent bone fractures [15, 16, 17] can be used to expand Vγ9Vδ2 T cells from cancer patients with hepatocellular [18], colorectal [18], prostate [19], lymphoid [20], and breast cancers [21, 22]. Zoledronate’s mechanism of action is to inhibit the enzyme, farnesyl diphosphate synthase (FDPS), which is part of the mevalonate metabolic pathway leading to the accumulation of IPP in cancer cells and subsequent TCR-dependent activation of Vγ9Vδ2 T cells. Similarly, Bromohydrin pyrophosphate (BrHPP) also interferes with the mevalonate metabolic pathway (Figure 1) [23, 24, 25].
In cancer patients, levels of γδ T cells are often low due to exposure to rigorous radiation and chemotherapy regimens, making ex vivo expansion a necessary solution. Studies have also shown that Vγ9Vδ2 T cells can be expanded from healthy donors and transferred to cancer patients with minimal adverse effects [22, 26, 27, 28]. This is largely possible because these cells do not rely on MHC recognition for antigen detection. Consequently, donor-derived allogeneic γδ T cells are unlikely to trigger graft-versus-host disease (GVHD), a major concern in other forms of cell-based therapy.
3. Cancer immunotherapies based on γδ T cells
Research on agonist-expanded Vγ9Vδ2 T cells has been conducted across various cancer types. In colorectal cancer (CRC), Vγ9Vδ2 T cells have been identified as tumor-infiltrating lymphocytes (TILs), and their presence seems to correlate with favorable outcomes. Ex vivo expanded Vγ9Vδ2 T cells exhibit strong lytic activity against colorectal carcinoma cell lines, relying on both the TCR and the NKG2D receptor for costimulatory signaling [18, 29, 30, 31]. Interestingly, expanding Vγ9Vδ2 T cells with BrHPP or Zoledronate generates effector memory cells capable of continuously eliminating CRC cell lines upon re-exposure. Similarly, BrHPP- or Zoledronate-stimulated γδ T cells effectively lyse hepatocellular carcinoma cells while sparing healthy surrounding tissues [18]. Further research indicates that this cytotoxicity occurs through both TCR-dependent and independent mechanisms, involving interactions between the NKG2D receptor and MIC A/B, as well as between the DNAX Accessory Molecule-1 (DNAM-1) receptor and the Nectin-like molecule-5 (Necl-5) on hepatocellular carcinoma cells [32].
While expanded γδ T cells show great promise, they tend to undergo mitogen-induced apoptosis more readily than traditional αβ T lymphocytes. To address this challenge, researchers attempted expanding Vγ9Vδ2 T cells in the presence of Interleukin-12 (IL-12), which selectively protects a subset of cells from apoptosis, leading to the proliferation of these apoptosis-resistant cells. These resistant Vγ9Vδ2 T cells demonstrated effectiveness against prostate cancer cell lines, such as the DU145 and PC-3 cell lines [33, 34]. These cells appear to kill cancer cell lines via TCR-dependent interactions, as well as through interactions between Integrin Beta Chain-2 (DC18) and Intercellular Adhesion Molecule-1 (ICAM-1), facilitated by the perforin/granzyme pathway [34]. Additionally, a modified protocol for ex vivo expansion using pulsed Zoledronate stimulation was developed to mitigate the toxicity produced by inhibiting farnesyl diphosphate synthase using a continuous exposure protocol [35]. Compared to continuous exposure, the pulsed γδ T cells showed increased purity and quantities. Moreover, pulse-expanded Vγ9Vδ2 T cells produced higher levels of perforin and degranulated their granzymes in greater numbers when co-cultured with PC-3 prostate cancer cells, resulting in a 2.5-fold increase in their anti-tumor cytolytic activity [36]. In immunocompromised mice with PC-3 tumor xenotransplants, the pulsed Vγ9Vδ2 T cells reduced tumor size by 50% compared to those receiving continuously expanded Vγ9Vδ2 T cells [37].
Furthermore, the Vδ2 subset of γδ T cells has been found in mammary ductal epithelial organoids, indicating their role in immunosurveillance within these tissues. These cells produce the anti-tumor cytokine IFN-γ while efficiently killing human breast cancer cells that are triple-negative for estrogen and progesterone receptors, as well as HER2/neu [38, 39]. Additional studies have also shown that Vγ9Vδ2 T cells exhibit cytotoxic activity against various breast cancer cell lines both in vitro and in murine models. This anti-tumor activity appears to depend on breast cancer subtype, TCR engagement, and the expression of MIC A/B and ICAM-1 [40, 41, 42]. While these results are promising, it is important to note that the predominant γδ T cell subset, Vδ1, found in TILs of breast cancer, exerts immunosuppressive effects, including the suppression of naïve T cell proliferation, dendritic cell maturation, and the secretion of immunosuppressive cytokines. The pro-tumor effects seem to be mediated by interferon gamma-induced protein 10 (IP-10), secreted by tumor cells. IP-10 recruits Vδ1 T cells to the tumor microenvironment, where they suppress anti-tumor immune responses and promote tumor growth [43].
Since ex vivo expanded Vγ9Vδ2 T cells are well-tolerated, several clinical studies have used highly enriched autologous Vγ9Vδ2 T cells prepared with Zoledronate, which were then re-infused into cancer patients to evaluate safety and potential therapeutic effects. In two early-phase clinical trials for Non-Small Cell Lung Cancer (NSCLC), stable disease was observed in three out of ten patients in one study and six out of fifteen patients in another [44, 45]. In advanced renal cell carcinoma (RCC), the transfer of γδ T cells into eleven patients resulted in prolonged tumor doubling time (DT), leading to one complete remission and stable disease in five patients [46]. Similar outcomes were achieved in metastatic RCC, where six out of ten patients showed stable disease [23]. In a clinical trial involving gastric cancer, intraperitoneal injection of ex vivo expanded Vγ9Vδ2 T cells led to a significant reduction in the volume of malignant ascites due to peritoneal dissemination in two of the seven patients enrolled [47]. In another trial, four patients with advanced refractory hematological malignancies received γδ T cells from haploidentical family donors, resulting in three complete responses [48].
Alternatively, Vγ9Vδ2 T cells can also be expanded in vivo rather than strictly ex vivo. This is typically done through the administration of the same FDA-approved amino-bisphosphonates used in ex vivo expansion, along with a low dose of IL-2. Phase I/II clinical trials demonstrated that this approach is both safe and feasible. Indeed, in vivo stimulation of Vγ9Vδ2 T cells in three breast cancer patients resulted in one case of partial remission and two cases of stable disease. This treatment was administered for over twelve months and correlated with declining levels of Cancer Antigen 15–3 (CA 15–3), a surrogate breast cancer biomarker [49]. Additionally, in vivo expansion with Zoledronate and IL-2 in nine hormone-refractory prostate cancer patients resulted in three instances of partial remission and five instances of stable disease [18]. Finally, in a trial focusing on nine patients with relapsed/refractory Non-Hodgkin’s Lymphoma or multiple myeloma, partial remissions were achieved in three patients [20].
4. Conclusion
Although several clinical trials have demonstrated the safety and feasibility of γδ T cell immunotherapies, the overall response in patients has been variable and inconsistent. There might be several potential contributing factors. For instance, the agonists used in in vivo expansion may have off-target effects, such as systemic toxicity in a subset of patients. Additionally, in several clinical trials, γδ T cell infiltration and activation have been limited, possibly due to the immunosuppressive effects of the tumor microenvironment on γδ T cells [50]. There is also limited knowledge about γδ T cell checkpoint inhibition and their exact mechanisms for suppressing tumor growth. Currently, efforts are invested in developing Chimeric Antigen Receptor (CAR)-engineered γδ T cells. These genetically engineered immune cells, whether autologous or allogeneic, have the potential to be off-the-shelf cell therapy products due to their ability to recognize antigens in an MHC-independent manner. One study successfully transduced γδ T cells with a second-generation CAR targeting GD2, demonstrating precise antitumor cytotoxicity against cancer cells expressing this specific antigen [51]. Despite the challenges and limitations observed in early clinical trials, these CAR-transduced γδ T cells could offer significant advantages as personalized cancer treatments. As the understanding of cancer biology expands and new technologies continue to develop, cancer treatments are becoming increasingly personalized, and γδ T cell therapy may be on the brink of a significant breakthrough.
References
- 1.
Read SB, Kulprathipanja NV, Gomez GG, Paul DB, Winston KR, Robbins JM, et al. Human alloreactive CTL inter-actions with gliomas and with those having upregulated Hla expression from exogenous IFN-gamma or IFN-gamma gene modification. Journal of Interferon & Cytokine Research. 2003; 23 (7):379-393 - 2.
Ksendzovsky A, Feinstein D, Zengou R, Sharp A, Polak P, Lichtor T, et al. Investigation of immunosuppressive mechanisms in a mouse glioma model. Journal of Neuro-Oncology. 2009; 93 (1):107-114 - 3.
Ashley DM, Kong FM, Bigner DD, Hale LP. Endogenous expression of transforming growth factor Beta1 inhibits growth and tumorigenicity and enhances Fas-mediated apoptosis in a murine high-grade glioma model. Cancer Research. 1998; 58 (2):302-309 - 4.
Jachimczak P, Bogdahn U, Schneider J, Beha C, Meixensberger J, Apfel R, et al. The effect of transforming growth factor-beta 2-specific phosphorothioate-anti-sense oligodeoxynucleotides in reversing cellular immunosuppression in malignant glioma. Journal of Neurosurgery. 1993; 78 (6):944-951 - 5.
Groh V, Rhinehart R, Secrist H, Bauer S, Grabstein KH, Spies T. Broad tumor-associated expression and recognition by tumor-derived Gamma Delta T cells of MICA and MICB. Proceedings of the National Academy of Sciences of the United States of America. 1999; 96 (12):6879-6884 - 6.
Hayday AC. Gamma Delta cells: A right time and a right place for a conserved third way of protection. Annual Review of Immunology. 2000; 18 :975-1026 - 7.
Deniger DC, Moyes JS, Cooper LJ. Clinical applications of Gamma Delta T cells with multivalent immunity. Frontiers in Immunology. 2014; 5 :636 - 8.
LeFranc MP, Forster A, Baer R, Stinson MA, Rabbitts TH. Diversity and rearrangement of the human T cell rearranging gamma genes: Nine germ-line variable genes belonging to two subgroups. Cell. 1986; 45 (2):237-246 - 9.
Forster A, Huck S, Ghanem N, Lefranc MP, Rabbitts TH. New subgroups in the human T cell rearranging V gamma gene locus. The EMBO Journal. 1987; 6 (7):1945-1950 - 10.
Strauss WM, Quertermous T, Seidman JG. Measuring the human T cell receptor gamma-chain locus. Science. 1987; 237 (4819):1217-1219 - 11.
Groh V, Steinle A, Bauer S, Spies T. Recognition of stress-induced MHC molecules by intestinal epithelial gamma delta T cells. Science. 1998; 279 (5357):1737-1740 - 12.
Bauer S, Groh V, Wu J, Steinle A, Phillips JH, Lanier LL, et al. Activation of NK cells and T cells by NKG2D, a receptor for stress-inducible MICA. Science. 1999; 285 (5428):727-729 - 13.
Saura-Esteller J, de Jong M, King LA, Ensing E, Winograd B, de Gruijl TD, et al. Gamma Delta T-cell based cancer immunotherapy: Past-present-future. Frontiers in Immunology. 2022; 13 :915837. DOI: 10.3389/fimmu.2022.915837 - 14.
Lafont V, Sanchez F, Laprevotte E, Michaud HA, Gros L, Eliaou JF, et al. Plasticity of γδ T cells: Impact on the anti-tumor response. Frontiers in Immunology. 2014; 5 :622 - 15.
Kunzmann V, Bauer E, Wilhelm M. Gamma/Delta T-cell stimulation by pamidronate. The New England Journal of Medicine. 1999; 340 (9):737-738 - 16.
Kondo M, Sakuta K, Noguchi A, Ariyoshi N, Sato K, Sato S, et al. Zoledronate facilitates large-scale ex vivo expansion of functional Gammadelta T cells from cancer patients for use in adoptive immunotherapy. Cytotherapy. 2008; 10 (8):842-856 - 17.
Sato K, Kimura S, Segawa H, Yokota A, Matsumoto S, Kuroda J, et al. Cytotoxic effects of gamma delta T cells expanded ex vivo by a third generation bisphosphonate for cancer immunotherapy. International Journal of Cancer. 2005; 116 (1):94-99 - 18.
Bouet-Toussaint F, Cabillic F, Toutirais O, Le Gallo M, Thomas de la Pintiere C, Daniel P, et al. Vgamma9vdelta2 T cell-mediated recognition of human solid tumors. Potential for immunotherapy of hepatocellular and colorectal carcinomas. Cancer Immunology, Immunotherapy. 2008; 57 (4):531-539 - 19.
Dieli F, Vermijlen D, Fulfaro F, Caccamo N, Meraviglia S, Cicero G, et al. Targeting human Gamma Delta T cells with zoledronate and Interleukin-2 for immunotherapy of hormone-refractory prostate cancer. Cancer Research. 2007; 67 (15):7450-7457 - 20.
Wilhelm M, Kunzmann V, Eckstein S, Reimer P, Weissinger F, Ruediger T, et al. Gamma delta T cells for immune therapy of patients with lymphoid malignancies. Blood. 2003; 102 (1):200-206 - 21.
Santini D, Martini F, Fratto ME, Galluzzo S, Vincenzi B, Agrati C, et al. In vivo effects of zoledronic acid on peripheral Gammadelta T lymphocytes in early breast cancer patients. Cancer Immunology, Immunotherapy. 2008; 58 (1):31-38 - 22.
Gaafar A, Aljurf MD, Al-Sulaiman A, Iqniebi A, Manogaran PS, Mohamed GE, et al. Defective gammadelta T-cell function and granzyme B gene polymorphism in a cohort of newly diagnosed breast cancer patients. Experimental Hematology. 2009; 37 (7):838-848 - 23.
Bennouna J, Bompas E, Neidhardt EM, Rolland F, Philip I, Galéa C, 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 Immunology, Immunotherapy. 2008; 57 (11):1599-1609 - 24.
Gober HJ, Kistowska M, Angman L, Jeno P, Mori L, De Libero G. Human T cell receptor gammadelta cells recognize endogenous mevalonate metabolites in tumor cells. The Journal of Experimental Medicine. 2003; 197 (2):163-168 - 25.
Kunzmann V, Bauer E, Feurle J, Weissinger F, Tony HP, Wilhelm M. Stimulation of gamma delta T cells by amino bisphosphonates and induction of anti plasma cell activity in multiple myeloma. Blood. 2000; 96 (2):384-392 - 26.
Bryant NL, Gillespie GY, Lopez RD, Markert JM, Cloud GA, Langford CP, et al. Preclinical evaluation of ex vivo expanded/activated γδ T cells for immunotherapy of glioblastoma multiforme. Journal of Neuro-Oncology. 2011; 101 (2):179-188 - 27.
Bryant NL, Suarez-Cuervo C, Gillespie GY, Markert JM, Nabors LB, Meleth S, et al. Characterization and immunotherapeutic potential of gammadelta T-cells in patients with glioblastoma. Neuro-Oncology. 2009; 11 (4):357-367 - 28.
Knight A, Arnouk H, Britt W, Gillespie GY, Cloud GA, Harkins L, et al. CMV-independent lysis of glioblastoma by ex vivo expanded/activated Vδ1+ γδ T cells. PLoS One. 2013; 8 (8):e68729 - 29.
Corvaisier M, Moreau-Aubry A, Diez E, Bennouna J, Mosnier JF, Scotet E, et al. V Gamma 9v Delta 2 T cell response to colon carcinoma cells. Journal of Immunology. 2005; 175 (8):5481-5488 - 30.
Gentles AJ, Newman AM, Liu CL, Bratman SV, Feng W, Kim D, et al. The prognostic landscape of genes and infiltrating immune cells across human cancers. Nature Medicine. 2015;(8):938-945 - 31.
Tosolini M, Pont F, Poupot M, Vergez F, Nicolau-Travers VML, Sarry D, et al. Assessment of tumor-infiltrating TCRVγ9Vδ2 lymphocyte abundance by deconvolution of human cancers microarrays. Oncoimmunology. 2017; 6 :1-10 - 32.
Toutirais O, Cabillic F, Le Friec G, et al. DNAX accessory molecule-1 (CD226) promotes human hepatocellular carcinoma celllysis by γ9Vδ2 T cells. European Journal of Immunology. 2009; 39 (5):1361-1368 - 33.
Lopez RD, Xu S, Guo B, Negrin RS, Waller EK. CD2-mediated Il-12-dependent signals render human Gamma Delta-T cells resistant to mitogen-induced apoptosis, permitting the large-scale ex vivo expansion of functionally distinct lymphocytes: Implications for the development of adoptive immunotherapy strategies. Blood. 2000; 96 (12):3827-3837 - 34.
Liu Z, Guo BL, Gehrs BC, Nan L, Lopez RD. Ex vivo expanded human Vgamma9vdelta2+ gamma delta-T cells mediate innate antitumor activity against human prostate cancer cells in vitro. The Journal of Urology. 2005; 173 (5):1552-1556 - 35.
Wang H, Sarikonda G, Puan KJ, Tanaka Y, Feng J, Giner JL, et al. Indirect stimulation of human Vγ2vδ2 T cells through alterations in isoprenoid metabolism. Journal of Immunology. 2011; 187 (10):5099-5113 - 36.
Alter G, Malenfant JM, Altfeld M. Cd107a as a functional marker for the identification of natural killer cell activity. Journal of Immunological Methods. 2004; 294 (1-2):15-22 - 37.
Nada MH, Wang H, Workalemahu G, Tanaka Y, Morita CT. Enhancing adoptive cancer immunotherapy with Vγ2vδ2 T cells through pulse zoledronate stimulation. Journal for Immunotherapy of Cancer. 2017; 5 :9 - 38.
Smyth MJ, Dunn GP, Schreiber RD. Cancer immunosurveillance and immunoediting: The roles of immunity in suppressing tumor development and shaping tumor immunogenicity. Advances in Immunology. 2006; 90 :1-50 - 39.
Zumwalde NA, Haag JD, Sharma D, Mirrielees JA, Wilke LG, Gould MN, et al. Analysis of immune cells from human mammary ductal epithelial organoids reveals Vδ2+ T cells that efficiently target breast carcinoma cells in the presence of bisphosphonate. Cancer Prevention Research (Philadelphia, Pa.). 2016; 9 (4):305-316 - 40.
Bank I, Book M, Huszar M, Baram Y, Schnirer I, Brenner H. V Delta 2+ Gamma Delta T lymphocytes are cytotoxic to the MCF 7 breast carcinoma cell line and can Be detected among the T cells that infiltrate breast tumors. Clinical Immunology and Immunopathology. 1993; 67 (1):17-24 - 41.
Guo BL, Liu Z, Aldrich WA, Lopez RD. Innate anti-breast cancer immunity of apoptosis-resistant human gamma delta-T cells. Breast Cancer Research and Treatment. 2005; 93 (2):169-175 - 42.
Aggarwal R, Lu J, Kanji S, Das M, Joseph M, Lustberg B, et al. Human Vγ2vδ2 T cells limit breast cancer growth by modulating cell survival-, apoptosis-related molecules and microenvironment in tumors. International Journal of Cancer. 2013; 133 (9):2133-2144 - 43.
Ye J, Ma C, Wang F, Hsueh EC, Toth K, Huang Y, et al. Specific recruitment of γδ regulatory T cells in human breast cancer. Cancer Research. 2013; 73 (20):6137-6148 - 44.
Nakajima J, Murakawa T, Fukami T, Goto S, Kaneko T, Yoshida Y, et al. A phase I study of adoptive immunotherapy for recurrent non-small-cell lung cancer patients with autologous gammadelta T cells. European Journal of Cardiothoracic Surgery; 37 (5):1191-1197 - 45.
Miki S, Nakajima J, Murakawa T, Fukami T, Yoshida Y, Murayama T, et al. Adoptive immunotherapy for advanced non-small cell lung cancer using zoledronate-expanded γδ T cells: A phase I clinical study. Journal of Immunotherapy. 2011; 34 (2):202-211 - 46.
Kobayashi H, Tanaka Y, Yagi J, Minato N, Tanabe K. Phase I/II study of adoptive transfer of γδ T cells in combination with zoledronic acid and IL-2 to patients with advanced renal cell carcinoma. Cancer Immunology, Immunotherapy. 2011; 60 (8):1075-1084 - 47.
Wada I, Matsushita H, Noji S, Mori K, Yamashita H, Nomura S, et al. Intraperitoneal injection of in vitro expanded Vγ9vδ2 T cells together with zoledronate for the treatment of malignant ascites due to gastric cancer. Cancer Medicine. 2014; 3 (2):362-375 - 48.
Martin W, Smetak M, Schaefer-Eckart K, Kimmel B, Birkmann J, Einsele H, et al. Successful adoptive transfer and in vivo expansion of haploidentical γδ T cells. Journal of Translational Medicine. 2014; 12 (1):45 - 49.
Meraviglia S, Eberl M, Vermijlen D, Todaro M, Buccheri S, Cicero G, et al. In vivo manipulation of Vgamma-9vdelta2 T cells with zoledronate and low-dose Interleukin-2 for immunotherapy of advanced breast cancer patients. Clinical and Experimental Immunology. 2010; 161 (2):290-297 - 50.
Revesz IA, Joyce P, Ebert LM, Prestidge CA. Effective γδ T-cell clinical therapies: Current limitations and future perspectives for cancer immunotherapy. Clinical and Translational Immunology. 2024; 13 (2):e1492 - 51.
Capsomidis A, Benthall G, Van Acker HH, Fisher J, Kramer AM, Abeln Z, et al. Chimeric antigen receptor-engineered human Gamma Delta T cells: Enhanced cytotoxicity with retention of cross presentation. Molecular Therapy. 2018; 26 :354-365