CBSS2-6 Interferon-alpha boosts anti-tumor immunity through effects on T cells and dendritic cells and augments the clinical efficacy of regulatory T cell depletion

Cytokine ◽  
2009 ◽  
Vol 48 (1-2) ◽  
pp. 8
Blood ◽  
2004 ◽  
Vol 104 (9) ◽  
pp. 2794-2800 ◽  
Author(s):  
Terry J. Fry ◽  
Manoj Sinha ◽  
Matthew Milliron ◽  
Yu-Waye Chu ◽  
Veena Kapoor ◽  
...  

Abstract Despite recent progress in our understanding of the biology of T-cell homeostasis, clinically available therapies to substantially improve immune reconstitution in patients sustaining T-cell depletion are lacking. T cells are regenerated via a dynamic interplay between thymopoiesis and thymic-independent homeostatic peripheral expansion (HPE). Using athymic mice subjected to T-cell depletion, we observed that HPE is critically dependent on dendritic cells (DCs) for presentation of antigen, raising the possibility that the availability of DCs might be limiting in vivo for HPE to occur efficiently. Indeed, flt3 ligand (flt3L) treatment of athymic mice subjected to T-cell depletion (without DC depletion) substantially enhanced HPE and improved immune competence. Following bone marrow transplantation (BMT) in athymic hosts, both dendritic cells and T cells were profoundly depleted and flt3L therapy restored DC numbers and enhanced HPE. In addition, thymus-bearing BMT recipients treated with flt3L regenerated increased numbers of thymic-dependent progeny with increased numbers of T-cell receptor excision circle (TREC)-positive T cells, indicating increased thymopoiesis. Therefore, flt3L is a potent immunorestorative agent that enhances both thymic-dependent and thymic-independent pathways of T-cell regeneration. (Blood. 2004;104:2794-2800)


2021 ◽  
pp. clincanres.CCR-20-4594-A.2020
Author(s):  
Suzanne Thibodeaux ◽  
Brian Barnett ◽  
Srilakshmi Pandeswara ◽  
Shawna Wall ◽  
Vincent Hurez ◽  
...  

Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 5209-5209
Author(s):  
Baldev Vasir ◽  
Corrine Lenahan ◽  
Jacalyn Rosenblatt ◽  
Adam Bissonnette ◽  
Zekui Wu ◽  
...  

Abstract In pre-clinical models, vaccination with attenuated vaccinia and fowl pox virus expressing tumor antigens and costimulatory molecules (CD54/ICAM-1; CD58/LFA-1 and CD80/B7.1-TRICOM) potently stimulates anti-tumor immune responses. However, vaccine efficacy may be limited by intrinsic deficiencies of native dendritic cells (DC) populations in patients with malignancy that are required to process and present the virally introduced antigens. An alternative strategy involves the transduction of ex vivo generated activated DCs. We have examined the capacity of DCs transduced with a fowl pox vector expressing MUC-1, CEA, and TriCOM (PANVAC-F) to elicit antigen specific responses and expand activated as compared to regulatory T cell populations. Partially mature DCs were generated from leukopak preparations obtained from normal volunteers by culturing adherent peripheral blood mononuclear cells for 5 days with GM-CSF and IL-4. DCs were transduced with PANVAC-F vector and matured with either TNFa or the combination of PG-E2, TNFa, IL-6 and IL-1b. In 5 serial studies, transduction with PANVAC-F resulted in mean MUC1 expression in 64.6% (SEM: ±1.6) of cells with a mean fluorescent intensity (MFI) of 249.8 (SEM: ±45.7). In addition, transduced DCs also demonstrated high levels of expression of class II (99%; MFI:250), CD54 (98.4%;MFI:516), CD58 (98.8%;MFI:155) and CD80 (78.2%; MFI:115). Of note, transduced DCs demonstrated higher levels of the maturation marker CD83 (40.3%; SEM: ±1.6, n=3) as compared to untransduced DCs 19.7% (SEM: ±2.9) (p=0.02) suggesting that transduction enhanced DC maturation and activation. Transduced DCs matured with PGE2, TNFa, IL-6 and IL-1b as compared to TNFa alone demonstrated higher levels of CD83 and CCR7 and a more stable phenotype following withdrawal of cytokine support. To assess the ability of PANVAC-F to stimulate tumor antigen specific responses, the presence of T cells binding the MUC-1 specific tetramer was quantified following stimulation of autologous T cells derived from HLA*0201 healthy donors. An increase in CD8+ and MUC1+ cells were observed (8.3%) with stimulation with PANVAC-F transduced DCs as compared to untransduced DCs (2.5%). The capacity of DCs transduced with PANVAC-F to stimulate interferon gamma producing activated T cells, as compared to CD4+/CD25+/Foxp3+ regulatory T cell populations is being assessed. In summary, PANVAC-F transduced DCs stimulate expansion of antigen specific T cell populations suggesting their potential role as tumor vaccines for MUC-1/CEA expressing tumors. We are initiating a trial for patients with ovarian carcinoma in early (marker only) relapse in which patients will be randomized to undergo serial vaccination with PANVAC-V/PANVAC-F or DCs transduced with PANVAC-F. The capacity to generate anti-tumor immunity in vivo will be assessed as a primary endpoint.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 4166-4166 ◽  
Author(s):  
Hailing Lu ◽  
Eric Ford ◽  
Jeffrey L Schwartz ◽  
Jessica Hewitt ◽  
Frank J Hsu ◽  
...  

Abstract Background: Radiation therapy is a standard established treatment modality for many malignancies, including lymphoma. There is growing evidence that the combination of radiation and immunotherapies can have synergistic immune and clinical effects; however, the mechanisms underlying these responses are poorly understood. Hypothesis: We hypothesized that the potential beneficial effect of adding immunotherapy to radiation might be due to alteration of the tumor microenvironment (TME) to an inflammatory phenotype and the facilitation of T-cell infiltration. Methods: Mice received two bilateral subcutaneous implant of 5 million A20 lymphoma cells each. When tumors are established (average tumor size = 45mm2, around Day 7~11), mice received intratumoral injection of G100, a synthetic TLR4 agonist, GLA-SE, tumor irradiation, or the combination of G100 and irradiation. X-ray radiation was delivered by using a Small Animal Radiation Research Platform (SARRP) with CT guided imaging that can accurately target lesions. Mice received a single dose of 10 Gy at one of the tumor implants. Intratumoral injection of G100 (10ug) was administered 3 times a week for up to 3 weeks, starting on the day post radiation. Results: The combination of G100 and radiation had synergistic antitumor effects and demonstrated an abscopal effect with tumor growth delay in the non-treated contralateral tumor implant. Long-term tumor immunity was also demonstrated as the surviving mice rejected secondary tumor challenge. Study of the TME at gene expression and cellular levels showed that G100 induces a pro-inflammatory cytokine and chemokine milieu that induced T cell trafficking into the tumors. G100-treated tumors showed increased infiltration of T cells, including both CD4 and CD8 T cells. In contrast, tumors from animals that received only irradiation but no G100 had significantly decreased levels of T lymphocytes as compared to untreated tumors (9.5±2.0% T cells in irradiated tumors vs. 32.0±5.0% in untreated tumors, p=0.0076), due to the cytotoxic effect of radiation on lymphocytes. CD4 tumor infiltrating lymphocytes (TILs) were depleted by 72% (6.8±2.2% vs. 24.4±4.6%, p=0.02) and CD8 TIL were depleted by 77% (1.5±0.3% vs. 6.6±0.7%, p=0.0004) in irradiated mice as compared to untreated mice. In the irradiation and G100 combination treatment group, the tumors had levels of T cells that were close to non-irradiated animals. Conclusion: The anti-tumor effect of irradiation is synergistically enhanced by concomitant intratumoral treatment by G100, which also results in abscopal responses and durable anti-tumor immunity. The modulation of the TME by G100 after irradiation also rescues the T-cell depletion caused by irradiation allowing for an influx of T cells. These preclinical results support the rationale for an on-going Phase 1/2 clinical trial in low-grade follicular non-Hodgkin's lymphoma (NHL) patients, who are randomized to receive G100 following local radiation with or without pembrolizumab (NCT02501473). Disclosures Lu: Immune Design: Employment. Hewitt:Immune Design: Employment. Hsu:Immune Design: Employment. ter Meulen:Immune Design: Employment.


2016 ◽  
Vol 69 ◽  
pp. S100
Author(s):  
A. Beebe ◽  
A. Mahne ◽  
S. Mauze ◽  
B. Joyce-Shaikh ◽  
D. Cua ◽  
...  

2020 ◽  
Vol 8 (Suppl 3) ◽  
pp. A574-A574
Author(s):  
Ellen Duong ◽  
Timothy Fessenden ◽  
Arjun Bhutkar ◽  
Stefani Spranger

BackgroundCytotoxic (CD8+) T-cells are required for tumor eradication and durable anti-tumor immunity.1 The induction of tumor-reactive CD8+ T-cells is predominately attributed to a subset of dendritic cells (DC) called Batf3-driven DC1, given their robust ability to cross-present antigens for T-cell priming and their role in effector T-cell recruitment.2–4 Presence of the DC1 signature in tumors correlates with improved survival and response to immunotherapies.5–7 Yet, most tumors with a DC1 infiltrate still progress, suggesting that while DC1 can initiate tumor-reactive CD8+ T-cell responses, they are unable to sustain them. Therefore, there is a critical need to identify and engage additional stimulatory DC subsets to strengthen anti-tumor immunity and boost immunotherapy responses.MethodsTo identify DC subsets that drive poly-functional CD8+ T-cell responses, we compared the DC infiltrate of a spontaneously regressing tumor with a progressing tumor. Multicolor flow immunophenotyping and single-cell RNA-sequencing were used to profile the DC compartment of both tumors. IFNγ-ELISpot was performed on splenocytes to assess for systemic tumor-reactive T-cell responses. Sorted DC subsets from tumors were co-cultured with TCR-transgenic T-cells ex vivo to evaluate their stimulatory capacity. Cross-dressing (in vivo/ex vivo) was assayed by staining for transfer of tumor-derived H-2b MHC complexes to Balb/c DC, which express the H-2d haplotype. Protective systemic immunity was assayed via contralateral flank tumor outgrowth experiments.ResultsRegressor tumors were infiltrated with more cross-presenting DC1 than progressor tumors. However, tumor-reactive CD8+ T-cell responses and tumor control were preserved in Batf3-/- mice lacking DC1, indicating that anti-tumor immune responses could be induced independent of DC1. Through functional assays, we established that anti-tumor immunity against regressor tumors required CD11c+ DC and cGAS/STING-independent type-I-interferon-sensing. Single-cell RNA-sequencing of the immune infiltrate of regressor tumors revealed a novel CD11b+ DC subset expressing an interferon-stimulated gene signature (ISG+ DC). Flow studies demonstrated that ISG+ DC were more enriched in regressor tumors than progressor tumors. We showed that ISG+ DC could activate CD8+ T-cells by cross-dressing with tumor-derived peptide-MHC complexes, thereby bypassing the requirement for cross-presentation to initiate CD8+ T-cell-driven immunity. ISG+ DC highly expressed cytosolic dsRNA sensors (RIG-I/MDA5) and could be therapeutically harnessed by exogenous addition of a dsRNA analog to drive protective CD8+ T-cell responses in DC1-deficient mice.ConclusionsThe DC infiltrate in tumors can dictate the strength of anti-tumor immunity. Harnessing multiple stimulatory DC subsets, such as cross-presenting DC1 and cross-dressing ISG+ DC, provides a therapeutic opportunity to enhance anti-tumor immunity and increase immunotherapy responses.ReferencesFridman WH, et al. The immune contexture in human tumours: impact on clinical outcome. Nature Reviews Cancer 2012;12(4): p. 298–306.Hildner K, et al. Batf3 deficiency reveals a critical role for CD8alpha+ dendritic cells in cytotoxic T cell immunity. Science 2008;322(5904):p. 1097–100.Spranger S, et al. Tumor-Residing Batf3 dendritic cells are required for effector T cell trafficking and adoptive T cell therapy. Cancer Cell 2017;31(5):p. 711–723.e4.Roberts, EW, et al., Critical role for CD103(+)/CD141(+) dendritic cells bearing CCR7 for tumor antigen trafficking and priming of T cell immunity in melanoma. Cancer Cell 2016;30(2): p. 324–336.Broz ML, et al. Dissecting the tumor myeloid compartment reveals rare activating antigen-presenting cells critical for T cell immunity. Cancer Cell 2014;26(5): p. 638–52.Salmon H., et al., Expansion and activation of CD103(+) dendritic cell progenitors at the tumor site enhances tumor responses to therapeutic PD-L1 and BRAF inhibition. Immunity, 2016. 44(4): p. 924–38.Sánchez-Paulete AR, et al., Cancer immunotherapy with immunomodulatory anti-CD137 and Anti-PD-1 monoclonal antibodies requires BATF3-dependent dendritic cells. Cancer Discov, 2016;6(1):p. 71–9.


Blood ◽  
1985 ◽  
Vol 65 (3) ◽  
pp. 663-679
Author(s):  
L Levitt ◽  
TJ Kipps ◽  
EG Engleman ◽  
PL Greenberg

The efficacy of four separate methods of human bone marrow T lymphocyte depletion was assessed, and the effect of T cells and monocytes on in vitro growth of marrow (CFU-GEMM, BFU-E, and CFU-GM) and peripheral blood (BFU-E) hematopoietic progenitors was determined. Extent of T cell depletion was assessed by multiparameter fluorescent cell sorter (FACS) analysis and by functional studies. Cells staining positively by FACS analysis for one or more of three separate fluorescent pan-T cell monoclonal antibodies (MCAbs) comprised 8.4% to 9.5% of control marrow mononuclear cells (MNCs). T cells constituted 3.2% to 5.1% of marrow following single, sequential, or combination treatment with two different pan-T cell MCAbs (Leu 1 and TM1) plus complement, 1.5% to 2.2% of marrow following solid-phase immunoabsorption (“panning”), 0.2% of marrow after sheep cell rosetting, and only 0.05% of marrow after FACS selective cell sorting and gated separation. T cells made up 59% to 73% of control peripheral blood MNCs and 0.8% to 2.8% of peripheral MNCs following sheep cell rosetting plus treatment with Leu 1 MCAb and complement. Mitogen (PHA, Con A) and allogeneic MLC-induced blastogenic responses (stimulation indices, experimental/control or E/C) revealed a concordant decrement in marrow T cell function after MCAb plus complement (E/C of 3.9 to 9.0), after panning (E/C of 1.6 to 3.5) and after sheep cell rosetting (E/C of 0.7 to 1.3), compared with control marrow (E/C of 5.3 to 15.7). After T cell depletion, marrow BFU-E growth was 95% to 120% of control, CFU-GM growth was 90% to 108% of control, and CFU-GEMM growth was 89% to 111% of control. Marrow T cell and/or monocyte depletion did not alter erythropoietin-dependent BFU-E growth in the absence of Mo-conditioned medium (81% to 95% of control), and the addition of as many as 50 to 100 X 10(3) purified marrow monocytes or T cells to 10(5) autologous nonadherent T cell-depleted marrow target cells had a negligible (P greater than .1) effect on marrow BFU-E growth in vitro. Peripheral blood (PB) BFU-E/10(5) T- depleted target cells were 106% +/- 19% of expected; PB BFU-E growth was significantly diminished after monocyte depletion alone (7% +/- 6% of expected) or after monocyte plus T cell depletion (8% +/- 4% of expected).(ABSTRACT TRUNCATED AT 400 WORDS)


Sign in / Sign up

Export Citation Format

Share Document