scholarly journals Adoptively transferred ex vivo expanded γδ-T cells mediate in vivo antitumor activity in preclinical mouse models of breast cancer

2009 ◽  
Vol 122 (1) ◽  
pp. 135-144 ◽  
Author(s):  
Benjamin H. Beck ◽  
Hyung-Gyoon Kim ◽  
Hyunki Kim ◽  
Sharon Samuel ◽  
Zhiyong Liu ◽  
...  
Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 1347-1347
Author(s):  
Kiyoshi Sato ◽  
Shinya Kimura ◽  
Takeshi Yuasa ◽  
Hiromi Wada ◽  
Taira Maekawa

Abstract Bisphosphonates (BPs), widely used to treat bone diseases, have recently been attracted much interest for their antitumor activity and have been reported to exert direct antitumor effects on several cancer cell lines via the inactivation of Ras proteins. BPs inhibit farnesyl pyrophosphate (FPP) synthetase in the mevalonate pathway and deplete cellular pools of PPs such as farnesyl PP and geranylgeranyl PP which are indispensable for the activation of Ras proteins. In addition to their direct antitumor activity, BPs expand γδ T-cells which are potent effector cells and also induce the accumulation of isopentenyl PP as a tumor antigen in target cancer cells. The purpose of this study was to clarify the cytotoxic activity of γδ T-cells expanded ex vivo by the potent third generation BP zoledronate (ZOL). Peripheral blood mononuclear cells of five healthy donors were incubated with different concentrations of ZOL and interleukin-2. After 14 days incubation, 1 μM ZOL increased the absolute number of γδ T-cells 500–800 fold. Expanded γδ T-cells were of the Vγ9Vδ2 subset and cytokine levels of IL-2, -4, -5, -10, TNF-α and IFN-γ were not elevated at resting i.e. before contact to target cancer cells. In vitro cytotoxic activities of γδ T-cells against the luciferase-labeled small cell lung cancer (SCLC) cell line SBC-5 were examined by a newly developed cytotoxic assay using an in vivo imaging system (Xenogen, Alameda, CA) and video microscopy, Leica AS MDW (Leica Microsystems Inc., Bannockburn, IL). γδ T-cells killed SBC-5 cells pre-treated with 5 μM ZOL for 12 h after 1.5–3.0 h contact with the target cells whereas untreated SBC-5 were rarely killed. SBC-5 cells pretreated with 5 μM ZOL showed a marked increase in their sensitivity to lysis by γδ T-cells, percentages of specific lysis were 42% and 55% at effector/target (E/T) ratios of 5:1 and 10:1, respectively, while those of untreated SBC-5 cells were 8% and 13% at E/T ratios of 5:1 and 10:1, respectively. In vivo efficacy of γδ T-cells was investigated in mice xenografted subcutaneously with SBC-5 cells. Pretreatment with 80 μg/kg ZOL enhanced significantly antitumor activity of γδ T-cells also in vivo. These findings showed that ZOL stimulated the proliferation of γδ T-cells significantly and that the cytotoxic activity of γδ T-cells required pre-treatment of target cells with ZOL, indicating the potential use of autologous ex vivo expanded γδ T-cells for cancer immunotherapy.


2010 ◽  
Vol 33 (5) ◽  
pp. 280 ◽  
Author(s):  
Hua Li ◽  
Yubin Wang ◽  
FuXiang Zhou

Objective: To investigate the antitumor activity of ex vivo-expanded γδ-T cells derived from tumor-infiltrating lymphocytes(γδTILs) from cervical cancer patients when combined with galectin-1 antibody and studied both in vitro and in vivo. Methods: The presence of γδTILs in cervical cancer specimens was detected by immunohistochemistry and γδTILs were expanded using the solid-phase antibody method. The expression of galectin-1 by the human cervical cancer cell line, SiHa, was measured by Western blot and ELISA. In vitro cytotoxic activities of expanded γδTILs, with or without galectin-1 inhibitor, were determined using the LDH-release test. In vivo antitumor activity of γδTILs, combined with galectin-1 antibody, was evaluated using the SCID mouse model. Results: γδTILs existed in the cervical cancer and the percentage of TCRγδ+ cells in γδTILs after ex vivo expansion was 91.2±1.2% detected by flow cytometry. SiHa cell expressed and secreted galectin-1 as measured by Western blot and ELISA. Expanded γδTILs from human cervical cancer demonstrated marked cytotoxicity to SiHa or Hela cells. In comparison with non-treated group, the cytotoxicity of γδ TILs towards SiHa or Hela cell was significantly increased when effector and target cells were incubated with either lactose or galectin-1 antibody at E/T ratio of 1:1 (p < 0.05). γδTILs, in combination with galectin-1 antibody treatment, significantly suppressed the growth of xenografts in SCID mice, in comparison with all other groups (p < 0.05). γδTILs alone also showed the ability to inhibit tumour growth in vivo, but were more efficient when combined with specific antibody (p < 0.05). Conclusion: Taken together, our results suggest that γδ-T cells, combined with galectin-1 antibody treatment, could be a more effective adoptive immunotherapy for patients with cervical cancer than traditional adoptive immunotherapy methods.


Blood ◽  
2003 ◽  
Vol 102 (1) ◽  
pp. 200-206 ◽  
Author(s):  
Martin Wilhelm ◽  
Volker Kunzmann ◽  
Susanne Eckstein ◽  
Peter Reimer ◽  
Florian Weissinger ◽  
...  

Abstract There is increasing evidence that γδ T cells have potent innate antitumor activity. We described previously that synthetic aminobisphosphonates are potent γδ T cell stimulatory compounds that induce cytokine secretion (ie, interferon γ [IFN-γ]) and cell-mediated cytotoxicity against lymphoma and myeloma cell lines in vitro. To evaluate the antitumor activity of γδ T cells in vivo, we initiated a pilot study of low-dose interleukin 2 (IL-2) in combination with pamidronate in 19 patients with relapsed/refractory low-grade non-Hodgkin lymphoma (NHL) or multiple myeloma (MM). The objectives of this trial were to determine toxicity, the most effective dose for in vivo activation/proliferation of γδ T cells, and antilymphoma efficacy of the combination of pamidronate and IL-2. The first 10 patients (cohort A) who entered the study received 90 mg pamidronate intravenously on day 1 followed by increasing dose levels of continuous 24-hour intravenous (IV) infusions of IL-2 (0.25 to 3 × 106 IU/m2) from day 3 to day 8. Even at the highest IL-2 dose level in vivo, γδ T-cell activation/proliferation and response to treatment were disappointing with only 1 patient achieving stable disease. Therefore, the next 9 patients were selected by positive in vitro proliferation of γδ T cells in response to pamidronate/IL-2 and received a modified treatment schedule (6-hour bolus IV IL-2 infusions from day 1-6). In this patient group (cohort B), significant in vivo activation/proliferation of γδ T cells was observed in 5 patients (55%), and objective responses (PR) were achieved in 3 patients (33%). Only patients with significant in vivo proliferation of γδ T cells responded to treatment, indicating that γδ T cells might contribute to this antilymphoma effect. Overall, administration of pamidronate and low-dose IL-2 was well tolerated. In conclusion, this clinical trial demonstrates, for the first time, that γδ T-cell–mediated immunotherapy is feasible and can induce objective tumor responses. (Blood. 2003;102:200-206)


2011 ◽  
Vol 4 (4) ◽  
pp. 211
Author(s):  
Serena Meraviglia ◽  
Carmela La Mendola ◽  
Valentina Orlando ◽  
Francesco Scarpa ◽  
Giuseppe Cicero ◽  
...  

The potent anti-tumor activities of γδ T cells, their ability to produce pro-inflammatory cytokines, and their strong cytolytic activity have prompted the development of protocols in which γδ agonists or ex vivo-expanded γδ cells are administered to tumor patients. γδ T cells can be selectively activated by either synthetic phosphoantigens or by drugs that enhance their accumulation into stressed cells as aminobisphosphonates, thus offering new avenues for the development of γδ T cell-based immunotherapies. The recent development of small drugs selectively activating Vγ9Vδ2 T lymphocytes, which upregulate the endogenous phosphoantigens, has enabled the investigators to design the experimental approaches of cancer immunotherapies; several ongoing phase I and II clinical trials are focused on the role of the direct bioactivity of drugs and of adoptive cell therapies involving phosphoantigen- or aminobisphosphonate-activated Vγ9Vδ2 T lymphocytes in humans. In this review, we focus on the recent advances in the activation/expansion of γδ T cells in vitro and in vivo that may represent a promising target for the design of novel and highly innovative immunotherapy in patients with hematologic malignancies.<br />


2010 ◽  
Vol 5 (5) ◽  
pp. 1934578X1000500 ◽  
Author(s):  
Vidya Menon ◽  
Sujata Bhat

Andrographolide 1, a diterpene lactone of Andrographis paniculata, displays in vitro and in vivo antitumor activity against breast cancer models and mouse myeloid leukemia (M1) cells. In the present study, we report the semi-synthesis of andrographolide derivatives and their in vitro activity against A549 (ATCC) (NSCL cancer) cell line. Amongst the derivatives tested, compounds 3- 5 displayed maximum activity, with IC50 values of 22-31 μg/mL.


2012 ◽  
Vol 30 (15_suppl) ◽  
pp. e21004-e21004
Author(s):  
Tomoharu Sugie ◽  
Kaoru Murata-Hirai ◽  
Masashi Iwasaki ◽  
Craig T Morita ◽  
Wen Li ◽  
...  

e21004 Background: Human γδ T cells display potent cytotoxicity against various tumor cells pretreated with zoledronic acid (Zol). Zol has shown benefits when added to adjuvant endocrine therapy for patients with early-stage breast cancer or to standard chemotherapy for patients with multiple myeloma. Although γδ T cells may contribute to this additive effect, the responsiveness of γδ T cells from early-stage breast cancer patients has not been fully investigated. In this study, we determined the number, frequency, and responsiveness of Vγ2Vδ2 T cells from early- and late-stage breast cancer patients and examined the effect of IL-18 on their ex vivo expansion. Methods: Breast cancer patients (n=80) were enrolled after institutional review board approval and with written informed consent. Peripheral blood mononuclear cells (PBMC) were purified and stimulated with Zol/IL-2 or Zol/IL-2/IL-18 for 2 to 10 days. The expanded cells were assessed on flow cytometry and the production of IFN-γ and TNF-α measured through ELISA. Results: The responsiveness of Vγ2Vδ2 T cells from patients with low frequencies of Vγ2Vδ2 T cells was significantly diminished. IL-18, however, enhanced ex vivo proliferative responses of Vγ2Vδ2T cells and helper NK cells (CD3-CD56brightCD11c+CD14-CD16+NKGD2+NKp44low) from patients with either low or high frequencies of Vγ2Vδ2 T cells. Cell-to-cell contact between γδ T and helper NK cells appeared to promote expansion of γδ T cells. Exogenous IL-18 markedly enhanced IFN-γ and TNF-α production from PBMC stimulated by Zol/IL-2, whereas the addition of an anti-IL-18Rα mAb reduced cytokine production. Conclusions: These results demonstrate that Zol elicits immunological responses by γδ T cells from early-stage breast cancer patients and IL-18 enhances proliferative responses and effector functions of γδ T cells in the context of helper NK cells.


2020 ◽  
Vol 38 (15_suppl) ◽  
pp. e15637-e15637
Author(s):  
Chundao Yang ◽  
Zhengwei Li ◽  
Qi Li ◽  
Yuanfeng Xia ◽  
Chi-Chung Chan ◽  
...  

e15637 Background: TP53 mutation is common in cancer cells, especially in small cell lung cancer (SCLC) and pancreatic cancer (PC). The G1 checkpoint function is lost in these cancer cells, which more rely on G2 checkpoint to repair damaged DNA. Wee1 is a serine/threonine kinase, which phosphorylates the Tyr15 locus of CDC2, inhibits the activity of CDC2, and stays in the G2 phase. Wee1 inhibition can promote the cells with DNA damage into the M phase, thus driving the TP53 mutant cancer cells to apoptosis. The purpose of this study was to investigate the in vitro and in vivo antitumor activity of SC0191, a small molecule inhibitor of Wee1 kinase, in preclinical models of SCLC and PC with TP53 mutation. Methods: The kinase inhibiting activity of SC0191 was determined using the Wee1 kinase assays. The cellular anti-proliferative activity was evaluated with TP53 mutant NCI-H446 SCLC and BxPC-3 PC cell lines. The in vivo antitumor activity of SC0191 was evaluated in the NCI-H446 and BxPC-3 cells-derived xenograft (CDX) mouse models, respectively. Results: SC0191 displayed potent kinase inhibiting activity for Wee1 with IC50 22.3 nM. SC0191 significantly inhibited tumor cell proliferation in TP53 mutant NCI-H446 cells with IC50 231.1 nM and BxPC-3 cells with IC50 223.8 nM. Moreover, SC0191 oral administration showed significant antitumor efficacy which was better than AZD1775, in the NCI-H446 and BxPC-3 CDX mouse models, respectively. Conclusions: A novel Wee1 inhibitor, SC0191, has demonstrated excellent antitumor efficacy in TP53 mutant solid cancer preclinical studies, and represents a promising clinical candidate for treating solid tumors, such as SCLC and PC.


2021 ◽  
Vol 9 (5) ◽  
pp. e002222
Author(s):  
Jeong A Park ◽  
Brian H Santich ◽  
Hong Xu ◽  
Lawrence G Lum ◽  
Nai-Kong V Cheung

BackgroundT cell-based immunotherapies using chimeric antigen receptors (CAR) or bispecific antibodies (BsAb) have produced impressive responses in hematological malignancies. However, major hurdles remained, including cytokine release syndrome, neurotoxicity, on-target off-tumor effects, reliance on autologous T cells, and failure in most solid tumors. BsAb armed T cells offer a safe alternative.MethodsWe generated ex vivo armed T cells (EATs) using IgG-[L]-scFv-platformed BsAb, where the anti-CD3 (huOKT3) scFv was attached to the light chain of a tumor-binding IgG. BsAb density on EAT, in vitro cytotoxicity, cytokine release, in vivo trafficking into tumors, and their antitumor activities were evaluated in multiple cancer cell lines and patient-derived xenograft mouse models. The efficacy of EATs after cryopreservation was studied, and gamma delta (γδ) T cells were investigated as unrelated alternative effector T cells.ResultsThe antitumor potency of BsAb armed T cells was substantially improved using the IgG-[L]-scFv BsAb platform. When compared with separate BsAb and T cell injection, EATs released less TNF-α, and infiltrated tumors faster, while achieving robust antitumor responses. The in vivo potency of EAT therapy depended on BsAb dose for arming, EAT cell number per injection, total number of EAT doses, and treatment schedule intensity. The antitumor efficacy of EATs was preserved following cryopreservation, and EATs using γδ T cells were safe and as effective as αβ T cell-EATs.ConclusionsEATs exerted potent antitumor activities against a broad spectrum of human cancer targets with remarkable safety. The antitumor potency of EATs depended on BsAb dose, cell number and total dose, and schedule. EATs were equally effective after cryopreservation, and the feasibility of third-party γδ-EATs offered an alternative for autologous T cell sources.


2021 ◽  
Vol 11 (1) ◽  
Author(s):  
Yun-Hsiang Chen ◽  
Yun Wang ◽  
Cheng-Hao Liao ◽  
Shu-Ching Hsu

AbstractBlinatumomab, a bispecific T cell engager (BiTE) antibody targeting CD19 and CD3ε, can redirect T cells toward CD19-positive tumor cells and has been approved to treat relapsed/refractory B-cell acute lymphoblastic leukemia (R/R B-ALL). However, chemotherapeutic regimens can severely reduce T cells’ number and cytotoxic function, leading to an inadequate response to blinatumomab treatment in patients. In addition, it was reported that a substantial portion of R/R B-ALL patients failing blinatumomab treatment had the extramedullary disease, indicating the poor ability of blinatumomab in treating extramedullary disease. In this study, we investigated whether the adoptive transfer of ex vivo expanded γ9δ2 T cells could act as the effector of blinatumomab to enhance blinatumomab’s antitumor activity against B-cell malignancies in vivo. Repeated infusion of blinatumomab and human γ9δ2 T cells led to more prolonged survival than that of blinatumomab or human γ9δ2 T cells alone in the mice xenografted with Raji cells. Furthermore, adoptive transfer of γ9δ2 T cells reduced tumor mass outside the bone marrow, indicating the potential of γ9δ2 T cells to eradicate the extramedullary disease. Our results suggest that the addition of γ9δ2 T cells to the blinatumomab treatment regimens could be an effective approach to enhancing blinatumomab’s therapeutic efficacy. The concept of this strategy may also be applied to other antigen-specific BiTE therapies for other malignancies.


Sign in / Sign up

Export Citation Format

Share Document