Synergistic Tumor Immunity Induced by Chemotherapy and Agonist Anti-GITR Antibody.

Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 1788-1788
Author(s):  
Adam D. Cohen ◽  
Daniel Hirschhorn-Cymmerman ◽  
Adi Diab ◽  
Miguel A. Perales ◽  
Taha Merghoub ◽  
...  

Abstract Ligation of GITR (glucocorticoid-induced tumor necrosis factor receptor) can both co-stimulate effector CD4 and CD8 T cells (Teff) and abrogate suppression by CD4+foxp3+ regulatory T cells (Tregs). This may be beneficial for the purposes of tumor immunotherapy, and we and others have demonstrated that the agonist anti-GITR antibody DTA1 can enhance both vaccine-induced and naturally-arising tumor immunity in murine models (Turk et al, JEM 2004; Cohen et al, Cancer Res 2006). In this study, we assessed the efficacy of combining GITR ligation with cyclophosphamide (CTX), a cytotoxic chemotherapeutic with immunomodulatory properties, to treat established, poorly immunogenic tumors. C57BL/6 mice received 50,000 B16 melanoma intradermally and were treated on day 6 with CTX 250 mg/kg intraperitoneally (ip), followed 1 day later by 1mg DTA1 or control rat IgG ip. In repeated experiments, 0–20% of mice treated with DTA1 alone or CTX + IgG had long-term tumor-free survival, compared with 60–80% long-term tumor-free survival in mice treated with CTX + DTA1. Starting CTX + DTA1 treatment on day 10 led to 40% tumor-free survival, with no survivors seen with either treatment alone. Synergy was lost with lower doses of CTX, or when CTX was given prior to tumor inoculation, indicating a likely requirement of tumor cell death and cross-presentation of tumor antigens to T cells. Consistent with this, the proliferation and activation of naïve pmel-1 CD8+ T cells (specific for the melanoma antigen gp100) was significantly enhanced when transferred into B16-bearing mice treated 1 day earlier with CTX, compared with untreated mice. This was not simply due to homeostatic proliferation in a lymphopenic state, as no differences were seen when cells were transferred into mice without tumors. In addition, increased GITR expression on both Teff and Tregs was observed for up to 4 days after in vivo CTX treatment, particularly on the proliferating (Ki67+) fraction, providing a greater target for GITR ligation by DTA1. Analysis of T cell populations in spleen, tumor-draining lymph node (DLN), and tumor following CTX + IgG treatment showed a relative decrement in Treg frequency in the first week, followed by a strong rebound, such that by day 14 after CTX roughly 50–60% of the tumor-infiltrating CD4+ cells were foxp3+. This rebound was largely abrogated, however, in the CTX + DTA1-treated mice, with only 5–15% of tumor-infiltrating CD4+ cells expressing foxp3. This led to a dramatic increase in the ratio of CD8+ to CD4+foxp3+ in the tumor (40:1 vs. 5:1 for CTX + IgG-treated mice), with lesser increases seen in the spleen and DLN as well, without significant changes in overall cellularity. Granzyme B expression was also increased in CD8+ and, to a lesser extent, CD4+foxp3- T cells from CTX + DTA1-treated mice, demonstrating increased cytolytic potential by Teff. In sum, CTX + agonist anti-GITR antibody can induce rejection of an aggressive syngeneic tumor, at a stage when either agent alone is ineffective. This synergy likely involves enhanced cross-priming and co-stimulation of Teff, with concomitant decrease in tumor-infiltrating Tregs, leading to a more effective anti-tumor immune response. This combination warrants further evaluation as an immunotherapeutic strategy for cancer.

Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 1911-1911
Author(s):  
Barry Flutter ◽  
Noha Edwards ◽  
Lei Zhang ◽  
Shivajanani Sivakumaran ◽  
Michael Croft ◽  
...  

Abstract Abstract 1911 A major limitation of adoptive T cell therapies for cancer is the failure to maintain durable anti-tumor immunity. Graft-versus-tumor responses following bone marrow transplantation (BMT) may only be short-lived due to 1) defects in memory precursor generation and 2) exhaustion of surviving CTL that results from direct recognition of alloantigen upon non-hematopoietic cells {Flutter et al. JCI 2010}. In this study, we have explored the potential for enhancing co-stimulatory signals either alone, or in combination with co-inhibitory PD-1-PD-L1 blockade to improve the long term CTL response. Signalling through OX40, a TNF-receptor family member, has been shown to have an important role in long-term immunity, including an enhancement in the generation of CD8 T cell memory precursors. The mechanisms of action are complex and may include both direct effects on CD8 cells and indirect effects on CD4 helper cells or via inhibition of Treg. In initial experiments, we evaluated the effects of early enforced OX40 co-stimulation following delayed transfer of donor T cells to haplo MHC-mismatched chimeras, 10 weeks following nonmyeloablative BMT. OX40 expression peaked on transferred CD4 and CD8 T cells in the first 1–2 weeks following transfer and was sustained thereafter, especially in the CD4 subset. 48 hours after T cell transfer, recipient mice were treated with agonistic anti-OX40 antibody (OX86) or isotype control. OX86 treatment led to a 9-fold increase in the expansion of CTL in comparison to isotype control treated mice, enhanced production of Granzyme B and IFNγ and led to more rapid eradication of host hematopoietic targets or host tumor cells. Moreover, OX86 antibody acted directly on CD8 T cells and bypassed the requirement for help from donor CD4 cells. However, although enforced OX40 co-stimulation boosted the primary effector response, it did not increase numbers of memory precursor cells, as assessed by survival and recall responses following transfer to antigen free hosts, and was unable to prevent eventual exhaustion of surviving donor CTL as tested at 60 days following transfer. Similarly, OX86 was unable to prevent exhaustion of CD8 cells transgenic for the male antigen-specific Matahari (Mh) TCR following adoptive transfer to male BMT recipients reconstituted with female BM. We have shown previously that the functions of exhausted donor CD8 cells are partially restored by blockade of the co-inhibitory PD-1 pathway in both haplo mismatched and MHC-matched mHAg mismatch models. We hypothesized that provision of co-stimulatory signals when exhaustion had become established would increase the effectiveness of co-inhibitory blockade. Therefore, 6 weeks after Mh CD8 T cell transfer to male BMT recipients, we examined the effect of OX86, with or without additional blockade of the PD-1 pathway. Only a minority of Mh CD8 cells from animals receiving isotype control antibody were proliferating in vivo as measured by BrdU incorporation over a 7 day pulse (20 +/−3% BrdU+) and few cells were able to produce IFNγ following antigen stimulation in vitro (3.5+/−1.4 x104 IFNγ+ cells/spleen). OX86 alone offered no restoration of function (15 +/− 2% BrdU+; 3.3+/−0.4 x104 IFNγ+ cells; p=ns). Blockade of PD-L1 modestly increased turnover of cells (37 +/− 6 % BrdU+; p<0.01 vs isotype), but in the absence of CD4 cells, did not significantly increase production of IFNγ (4.4+/−0.9 x104 IFNγ+ cells; p=ns). However, in vivo administration of OX86 combined with anti-PD-L1 blockade dramatically increased turnover of Mh CD8s (77 +/− 8% BrdU+; p<0.001 vs anti-PD-L1 alone, OX86 alone or Isotype) and enhanced their effector function ∼ 9-fold (27.4 +/− 6.8 x104 IFNγ+ cells/spleen; p<0.01 vs all others). In conclusion, forced co-stimulation via OX40 alone is unable either to prevent CTL exhaustion or restore CD8 T cell function when exhaustion has become established. In contrast, the marked synergy observed when agonistic OX40 signals are combined with co-inhibitory blockade, is consistent with a model in which the PD-1 pathway acts at a critical checkpoint that regulates the response to co-stimulation. Thus, these data suggest a novel approach to restoring the functions of exhausted anti-tumor CTL by modulating co-stimulatory and co-inhibitory pathways simultaneously. Disclosures: No relevant conflicts of interest to declare.


2004 ◽  
Vol 78 (21) ◽  
pp. 11641-11647 ◽  
Author(s):  
Hong He ◽  
Ronald J. Messer ◽  
Shimon Sakaguchi ◽  
Guojun Yang ◽  
Shelly J. Robertson ◽  
...  

ABSTRACT Chronic infection with Friend retrovirus is associated with suppressed antitumor immune responses. In the present study we investigated whether modulation of T-cell responses during acute infection would restore antitumor immunity in persistently infected mice. T-cell modulation was done by treatments with DTA-1 anti- glucocorticoid-induced tumor necrosis factor receptor monoclonal antibodies. The DTA-1 monoclonal antibody is nondepleting and delivers costimulatory signals that both enhance the activation of effector T cells and inhibit suppression by regulatory T cells. DTA-1 therapy produced faster Th1 immune responses, significant reductions in both acute virus loads and pathology and, most importantly, long-term improvement of CD8+ T-cell-mediated antitumor responses.


2000 ◽  
Vol 192 (11) ◽  
pp. 1637-1644 ◽  
Author(s):  
Cassian Yee ◽  
John A. Thompson ◽  
Patrick Roche ◽  
David R. Byrd ◽  
Peter P. Lee ◽  
...  

Current strategies for the immunotherapy of melanoma include augmentation of the immune response to tumor antigens represented by melanosomal proteins such as tyrosinase, gp100, and MART-1. The possibility that intentional targeting of tumor antigens representing normal proteins can result in autoimmune toxicity has been postulated but never demonstrated previously in humans. In this study, we describe a patient with metastatic melanoma who developed inflammatory lesions circumscribing pigmented areas of skin after an infusion of MART-1–specific CD8+ T cell clones. Analysis of the infiltrating lymphocytes in skin and tumor biopsies using T cell–specific peptide–major histocompatibility complex tetramers demonstrated a localized predominance of MART-1–specific CD8+ T cells (&gt;28% of all CD8 T cells) that was identical to the infused clones (as confirmed by sequencing of the complementarity-determining region 3). In contrast to skin biopsies obtained from the patient before T cell infusion, postinfusion biopsies demonstrated loss of MART-1 expression, evidence of melanocyte damage, and the complete absence of melanocytes in affected regions of the skin. This study provides, for the first time, direct evidence in humans that antigen-specific immunotherapy can target not only antigen-positive tumor cells in vivo but also normal tissues expressing the shared tumor antigen.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 188-188
Author(s):  
Sharon Lam ◽  
Julia Marsh Sung ◽  
Conrad RY Cruz ◽  
Paul Castillo-Caro ◽  
Minh T. Ngo ◽  
...  

Abstract Antiretroviral therapy (ART) does not eliminate HIV from latently infected reservoirs, has long-term toxicities and fails to fully prevent immune attenuation. Therefore there is a need for alternative therapies that will decrease dependency on ART. Previous studies have demonstrated the safety and feasibility of infusing single-epitope specific CD8 T cells or artificial T cell receptor- transduced T cells to HIV+ patients. However, these T cells were restricted to a single HLA restricted epitope and had limited persistence in vivo. Hence, we hypothesized that broadly HIV-specific T cells could be expanded from patients on ART and HIV negative individuals to effectively target HIV infection using a non-HLA restricted, GMP-compliant approach. We developed a method by which PBMCs from patients on ART were stimulated with autologous dendritic cells (DCs) pulsed with gag, pol, and nef peptide libraries (pepmixes) in the presence of IL-7, IL-12, and IL-15, followed by a second stimulation with pepmix-pulsed PHA blasts and IL-15 and a third stimulation with pepmix-pulsed PHA blasts, co-stimulatory K562 cells, and IL-2. Starting from 60 to 100 mL of blood, T cells expanded to clinically relevant numbers (Mean=1.62e8 cells, Range (3.72e7, 2.87e8 cells), n=7) using co-stimulatory K-562 cells and gas-permeable tissue culture devices in the presence of antiretrovirals to prevent possible viral spread during expansion. The majority of the expanded T cells had an effector memory phenotype (CD3+CD45RO+CD62L-) with approximately 10% suggestive of a central memory phenotype (CD3+CD45RO+CD62L+) which is important for long-term persistence of T cells in vivo. After 3 stimulations, 5 of 7 patient sample lines showed specific activity to all 3 HIV antigens in IFNY ELISPOT assays, with the remaining 2 showing specificity to 1 of 3 antigens. The T cell lines were broadly specific to gag (mean=99.33 SFC/10e5 cells), pol (mean=131.11 SFC/10e5 cells) and nef (mean=337.26 SFC/10e5 cells), and polyclonal as shown by flow-based Vbeta usage analysis (mean usage= 14.67 of the 24 Vbetas analyzed). In addition, due to the association of gag-specific T cell responses with viral control in long-term nonprogressors we also determined whether gag-specific T cells could be derived from seronegative donors for potential third party use. Using the same methodology developed for HIV+ patients, but using only the gag pepmix, gag-specific T cells were expanded from HIV seronegative individuals. These T cells released IFNγ in response to gag pepmix (163.79 SFC/1e5 cells, n=9) but not an irrelevant antigen (mean=7.0 SFC/1e5 cells). Importantly, T cells expanded from both ART patients and HIV seronegative individuals were cytotoxic, as expanded T cells lysed antigen loaded autologous PHA blasts (mean=67.55% specific lysis at 10:1 effector:target ratio) but not PHA blasts alone (mean=0.46% specific lysis at 10:1 effector target ratio) in chromium release assays. Expanded T cells from ART patients also showed a greater ability to suppress HIV outgrowth in vitro compared to unexpanded CD8 T cells when co-cultured with reactivated resting CD4+ T cells from ART-suppressed HIV+ patients, the authentic latently infected cells that define viral reservoirs in treated patients. In 5 patients on ART a statistically lower recovery of virus from resting CD4+ cells was seen in the presence of CTLs as compared to no effectors (p<0.006 by Mann Whitney), while the unexpanded autologous CD8 cells showed only a modest trend towards decreased recovery that was not statistically significant (p>0.9). Similarly, HIV-specific T cells derived from HIV seronegative individuals were able to suppress HIV replication more than unexpanded CD8 T cells when co-cultured with autologous CD4 T cells infected with HIVSF162 (HIV only condition p24=681.95 pg/mL, nonspecific CD8 T cells=448.80 pg/mL, expanded CTL=145.82 pg/mL). We have developed robust GMP-compliant methodologies for expanding functional HIV-specific T cells from both HIV+ patients and HIV negative donors for autologous and third-party use, respectively. We now plan to translate our approach to the clinical setting where we will test HIV-polyspecific T cell products as a part of a strategy to fully eradicate HIV infection. Disclosures: No relevant conflicts of interest to declare.


1990 ◽  
Vol 171 (3) ◽  
pp. 953-958 ◽  
Author(s):  
S R Webb ◽  
J Sprent
Keyword(s):  
T Cells ◽  

Contrary to existing dogma, evidence is presented that proliferative responses of mature unprimed T cells to Mlsa antigens involve CD8+ cells as well as CD4+ cells. The response of CD8+ cells to Mlsa antigens proved to be heavily dependent on help from CD4+ cells, and responses were stronger in three I-E+ strain combinations than in an I-E- combination. In I-E+ combinations, CD8+ blast cells accounted for 20-25% of the blasts generated from unseparated T cells responding to Mlsa-bearing stimulator cells in vitro; similar findings applied to blast cells generated in vivo. The observation that the majority (greater than or equal to 50%) of Mlsa-stimulated CD8+ cells (and CD4+ cells) were V beta 6+ indicated that CD8+ cells respond to Mlsa antigens, per se, rather than to nonspecific stimuli. Whether CD4+ and CD8+ cells use the same or different H-2-restricting elements to respond to Mlsa antigens has yet to be resolved.


Blood ◽  
2005 ◽  
Vol 105 (1) ◽  
pp. 241-250 ◽  
Author(s):  
Daniel J. Powell ◽  
Mark E. Dudley ◽  
Paul F. Robbins ◽  
Steven A. Rosenberg

Abstract In humans, the pathways of memory T-cell differentiation remain poorly defined. Recently, adoptive cell transfer (ACT) of tumor-reactive T lymphocytes to metastatic melanoma patients after nonmyeloablative chemotherapy has resulted in persistence of functional, tumor-reactive lymphocytes, regression of disease, and induction of melanocyte-directed autoimmunity in some responding patients. In the current study, longitudinal phenotypic analysis was performed on melanoma antigen-specific CD8+ T cells during their transition from in vitro cultured effector cells to long-term persistent memory cells following ACT to 6 responding patients. Tumor-reactive T cells used for therapy were generally late-stage effector cells with a CD27Lo CD28Lo CD45RA- CD62 ligand- (CD62L-) CC chemokine receptor 7- (CCR7-) interleukin-7 receptor αLo (IL-7RαLo) phenotype. After transfer, rapid up-regulation and continued expression of IL-7Rα in vivo suggested an important role for IL-7R in immediate and long-term T-cell survival. Although the tumor antigen-specific T-cell population contracted between 1 and 4 weeks after transfer, stable numbers of CD27+ CD28+ tumor-reactive T cells were maintained, demonstrating their contribution to the development of long-term, melanoma-reactive memory CD8+ T cells in vivo. At 2 months after transfer, melanoma-reactive T cells persisted at high levels and displayed an effector memory phenotype, including a CD27+ CD28+ CD62L- CCR7- profile, which may explain in part their ability to mediate tumor destruction. (Blood. 2005;105:241-250)


2021 ◽  
Vol 9 (Suppl 3) ◽  
pp. A276-A276
Author(s):  
Cassandra Gilmour ◽  
Li Wang ◽  
Juan Dong ◽  
Sarah Stone ◽  
Keman Zhang ◽  
...  

BackgroundCancer immunotherapies, specifically checkpoint blockade therapies, have demonstrated clinical importance for long term patient survival. One of the major limitations to checkpoint blockade therapies, is the low response rate: ~30% with anti-CTLA4 and anti-PD1 treatment. This may be due to heterogeneity of the patients‘ immune system and the tumor microenvironment including T cell inhibitions. There is a clear need to study this phenomenon and develop additional therapies for long term survival to include a broad range of patients. V-domain Immunoglobulin Suppressor of T-cell Activation (VISTA) is a suppressive protein expressed on many cell types in the tumor microenvironment including cytotoxic T cells. VISTA’s role on T cells has been described as maintaining quiescence and peripheral tolerance in a graft vs host disease model, but is not fully understood in context of the tumor microenvironment.MethodsWe use a series of invivo experiments, including T cell specific VISTA knock outs, to understand the role of VISTA on T cells in the tumor microenvironment.ResultsHere we show a series of in vivo experiments that suggest VISTA has a potent intrinsic role on T cells and therefore anti-tumor immunity. Using a T cell specific VISTA knock out, our results suggest that the absence of VISTA on T cells in combination with anti-CTLA4 and vaccine is a very powerful tumor suppressor compared to vaccine and anti-CTLA4 treatment alone. These results also indicate that the absence of VISTA alters the phenotype of cytotoxic T cells in several ways including the production of inflammatory cytokines.ConclusionsOur preliminary data provides foundation to study VISTA’s role intrinsic to T cells in the tumor microenvironment and how disrupting VISTA’s influence intrinsic to T cells may be advantageous for anti-tumor immunity and long term patient survival.Ethics ApprovalAll in vivo studies were reviewed and approved by Institutional Animal Care and Use Committee (Approval number 2019–2142).


2007 ◽  
Vol 25 (18_suppl) ◽  
pp. 3058-3058
Author(s):  
A. D. Cohen ◽  
A. Diab ◽  
M. A. Perales ◽  
F. Duan ◽  
R. Jenq ◽  
...  

3058 Background: Signaling through GITR (glucocorticoid-induced tumor necrosis factor receptor) can abrogate the suppressive effects of CD4+foxp3+ regulatory T cells and co-stimulate activated effector CD4+ and CD8+ T cells. We have previously shown that in vivo GITR ligation using the agonist anti-GITR mAb DTA-1 augments concomitant immunity and immunity generated by active immunization with self- tumor antigens. In the present study, we assessed the activity of anti-GITR mAb used alone, focusing on the effects of GITR ligation on CD8+ T cells during tumor growth. Methods: C57BL/6 mice were injected intradermally with B16 melanoma and received 1mg of DTA-1 or control rat IgG intraperitoneally on various days after tumor injection. In some experiments, naïve, CFSE-labeled pmel-1 CD8+ transgenic T cells (specific for the melanoma antigen gp10025–33 epitope) were transferred into naïve recipients 1 day prior to B16 inoculation. Results: DTA-1 treatment on days 0 and 4 led to tumor rejection in 20–30% and 50–60% of mice, respectively, compared with rejection in 0–5% of mice treated with control IgG (p<0.05 for both). Treatment at day 7 or later had no significant impact on tumor-free survival. The importance of CD8+ T cells in mediating DTA-1-induced tumor immunity was demonstrated by 4 findings: 1) in untreated mice, tumor-infiltrating CD8+ lymphocytes significantly upregulated GITR expression during tumor growth; 2) DTA-1-treated mice had greater CD8+ T cell infiltration into tumors than IgG-treated mice; 3) depletion of CD8+ cells completely abrogated the tumor protection provided by DTA-1; and 4) tumor-specific CD8+ cells proliferated more extensively, became more activated, and exhibited greater effector function following DTA-1 administration compared with control IgG. This was most dramatically seen within the tumor (compared with spleen or draining lymph node), suggesting that a major mechanism of tumor immunity induced by anti-GITR mAb may be overcoming impaired CD8+ T cell function within the tumor microenvironment. Conclusions: Ligating GITR using an agonist mAb can by itself augment tumor-specific CD8+ T cell responses and induce rejection of an aggressive, poorly immunogenic tumor. This strategy merits further consideration as an immune-modulating therapy for cancer. No significant financial relationships to disclose.


2021 ◽  
Vol 11 ◽  
Author(s):  
Siqi Guo ◽  
Ronald B. Smeltz ◽  
Anthony Nanajian ◽  
Richard Heller

Interleukin 15 (IL-15) has been evaluated as a potential treatment for solid tumors in clinical trials, but the effectiveness of systemic IL-15 administration as a monotherapy has not been realized. IL-15 receptor alpha (IL-15Rα) can stabilize IL-15 and enhance its bioactivity. The goal of this study was to examine the activity of IL-15/IL-15Rα complex (IL-15cx) to CD8+ T cells and evaluate its potential efficacy in murine breast cancer models. The antitumor efficacy was studied in mouse mammary carcinoma models (Her2/neu transgenic and 4T1-luc mammary cancers) treated with systemic recombinant protein with/without the depletion of myeloid-derived suppressor cells or intra-tumoral gene electrotransfer (GET). IL-15cx shows superior in vivo bioactivity to expand CD8 T cells in comparison to an equimolar single chain IL-15. T-bet is partially involved in CD8 T cell expansion ex vivo and in vivo due to IL-15 or IL-15cx. Intraperitoneal administration of IL-15cx results in a moderate inhibition of breast cancer growth that is associated with an increase in the frequency of cytotoxic CD8 T cells and the improvement of their function. The depletion of myeloid-derived suppressor cells (MDSCs) has no impact on mouse breast cancer growth. IL-15cx treatment diminishes MDSCs in murine tumors. However, it also antagonizes the effects of anti-Gr-1 depleting antibodies. Intratumoral GET with plasmid IL-15/IL-15Rα leads to a long-term survival benefit in 4T1 mammary carcinoma model. An early increase of local cytotoxic cells correlates with GET treatment and an increase of long-term memory T cells results from animals with complete tumor regression. Systemic and local administration of IL-15cx shows two distinct therapeutic responses, a moderate tumor growth inhibition or heterogeneous tumor regressions with survival improvement. Further studies are warranted to improve the efficacy of IL-15cx as an immunotherapy for breast cancer.


Sign in / Sign up

Export Citation Format

Share Document