Abstract 276: Recruitment of effector T cells into the tumor rim and center with neoadjuvant systemic GM-CSF in patients with localized prostate cancer

Author(s):  
Xiao X. Wei ◽  
Stephen Chan ◽  
Jera Lewis ◽  
Serena Kwek ◽  
Vinh Dao ◽  
...  
2016 ◽  
Vol 4 (11) ◽  
pp. 948-958 ◽  
Author(s):  
Xiao X. Wei ◽  
Stephen Chan ◽  
Serena Kwek ◽  
Jera Lewis ◽  
Vinh Dao ◽  
...  

2021 ◽  
Vol 39 (15_suppl) ◽  
pp. 2626-2626
Author(s):  
Chia-Chi Lin ◽  
Aung Naing ◽  
Manish R. Patel ◽  
Howard A. Burris III ◽  
Giuseppe Curigliano ◽  
...  

2626 Background: Inducible T-cell co-stimulator (ICOS) is an important co-stimulatory receptor on effector T cells (Teffs) that also promotes tumor growth due to its high expression on regulatory T cells (Tregs). KY1044 is a fully human IgG1 that targets ICOS, acting via a dual mode of action (MoA) by depleting ICOShigh Tregs and stimulating ICOSLow Teffs. A Phase 1/2 clinical trial (NCT03829501) is currently assessing the safety and preliminary efficacy of KY1044, as a single agent and in combination with atezolizumab, in subjects with advanced relapsed/refractory malignancies. Using longitudinal blood samples and tumor biopsies, we aim to correlate KY1044 target engagement levels with pharmacodynamic (PD) properties (e.g. dual MoA) in the tumor microenvironment (TME) and the circulation. Methods: Phase 1 subjects were enrolled in dose escalation and enrichment cohorts to evaluate the effect of KY1044 as monotherapy (0.8 – 240 mg) Q3W and in combination (0.8 – 80 mg) with atezolizumab (1200 mg) Q3W. PBMCs, plasma and tumor biopsies were collected over the first 3 cycles to confirm target engagement and KY1044 MoA. The sample analysis included: immunohistochemistry (IHC) of tumor samples (ICOS, FOXP3 and CD8); circulating T cell immunoprofiling and receptor occupancy by chip-cytometry; PBMC and tumor sample pre- and post-treatment transcriptomic analysis; and the assessment of circulating cytokines (e.g. GM-CSF). Results: As assessed in PBMCs, full/prolonged ICOS target engagement on T cells was confirmed in subjects receiving a flat dose of 8 to 240 mg, while partial/transient saturation was observed at lower doses (0.8-2.4 mg). The target engagement was not affected by atezolizumab. The immune cell profiling showed changes in some populations, but there was no significant depletion of peripheral ICOS+ cells. In contrast, pre- and post-treatment IHC analysis of ICOS+/FOXP3+ cells in tumor biopsies confirmed a KY1044-dose dependent reduction of ICOS+ Tregs and maintenance of CD8+ T cells in the TME. Together, this resulted in an increased intratumoral CD8+/ICOS+ Treg ratio at all doses, plateauing from subjects receiving a flat KY1044 dose of 8 mg. KY1044-dependent agonism was indirectly assessed by measuring circulating cytokine levels. A post-dosing transient induction of GM-CSF was evident in subjects dosed with KY1044 at the 0.8 and 2.4 mg dose, whereas minimal induction was observed at dose of 8 mg and higher. Conclusions: LongitudinalPDdata confirmed the expected KY1044 MoA, namely ICOS Treg depletion and increased CD8/ICOS Treg ratio in the TME as well as T cell co-stimulation. The observed PD responses are currently being further explored in a more homogenous patient population.


2012 ◽  
Vol 30 (15_suppl) ◽  
pp. 2564-2564 ◽  
Author(s):  
Lawrence Fong ◽  
Vivian K. Weinberg ◽  
Stephen E Chan ◽  
John M Corman ◽  
Christopher L Amling ◽  
...  

2564 Background: Sipuleucel-T is an FDA-approved autologous cellular immunotherapy for patients with asymptomatic or minimally symptomatic metastatic castrate resistant prostate cancer (mCRPC). To date, studies of sipuleucel-T in patients with mCRPC have studied immune response in peripheral blood. The effects of sipuleucel-T on prostate tumors are unknown. Methods: NeoACT (P07-1; NCT00715104) is an open-label, phase 2 study of patients with localized prostate cancer who received sipuleucel-T prior to radical prostatectomy (RP) to examine the immunologic effects of treatment on prostate tissue. Patients received 3 infusions of sipuleucel-T at approximately 2-week intervals, beginning 6-7 weeks prior to RP. The primary endpoint was the change in the frequency of lymphocytes between prostate biopsies (pre-treatment) and RP tissue (post-treatment), as assessed by immunohistochemistry (IHC). Results: The median age of the 42 enrolled patients was 61 years, and all had an ECOG performance status of 0. Thirty-eight patients received all 3 pre-RP sipuleucel-T infusions. To date, tissue IHC analysis has been completed on 32 patients. Treatment-related AEs were manageable and transient. Sipuleucel-T did not appear to impact surgery, as judged by operative complications, procedure time, and estimated blood loss. Frequent events that occurred ≤1 day after infusion (>10% of patients) were fatigue, headache, and myalgia. Significant increases (≥3 fold) in CD3+ and CD4+ T cell populations were observed at the tumor interface (where benign and malignant glands interface), compared with the pre-treatment biopsy, benign RP tissue, and tumor RP tissue (ANOVA post hoc Newman-Keuls test: p<0.0001 for each comparison). FoxP3+ CD4+ T cells were also increased (p=0.0005) at the tumor interface, but represented a small fraction of the observed CD4+ T cells. Conclusions: Neoadjuvant sipuleucel-T treatment is associated with an increased frequency of T cells in prostate cancer tissue at the interface of the benign and malignant glands. These data suggest that sipuleucel-T can modulate the presence of lymphocytes at the prostate tumor site. Work is ongoing to more fully characterize the immune response.


2020 ◽  
Vol 38 (6_suppl) ◽  
pp. 344-344
Author(s):  
Victor Ricardo Adorno Febles ◽  
Dennis Adeegbe ◽  
Aarif Ahsan ◽  
Jiansheng Wu ◽  
Alireza Khodadad-Jamayran ◽  
...  

344 Background: The immune factors that modulate the aggressiveness of localized treatment-naïve prostate cancer remain poorly understood. Methods: Fresh tumor and peripheral blood were collected at the time of radical prostatectomy in patients with localized prostate cancer. We evaluated the immune cell composition of 22 patient samples employing multi-parametric flow cytometry. Samples were grouped by histological grade into intermediate (INTPCA) and high-grade (HIGHPCA) prostate cancers based on standard NCCN criteria and immune cell abundances were quantified by mean +/- SEM. Statistical significance was assessed using the Mann-Whitney test. Results: INTPCA and HIGHPCA tumors harbored a similar increase in CD8+ T cells ( p < 0.0005 and p < 0.05, respectively) and CD11b+CD68-CD16+ myeloid-derived cells (p < 0.05) relative to the peripheral blood. Other cell types were similarly decreased in both INTPCA and HIGHPCA, including CD11b+CD68+CD14+ ( p < 0.005 and p < 0.05, respectively). By contrast, regulatory T cells were the only cell type in our analysis to be uniquely enriched in HIGHPCA rather than INTPCA ( p < 0.05). The most unique feature found in phenotypic profiling of the immune repertoire of HIGHPCA relative to INTPCA was an increase in the immune inhibitory receptor ligand, PD-L1, in the tumor associated macrophages (CD11b+CD68+CD14+) compared to the periphery (p < 0.05). Conclusions: Collectively, our findings reveal that HIGHPCA harbors a distinct immunological landscape. Although effector CD8+ T cells are preferentially expressed in the tumor, these are met with an increased proportion of regulatory T cells as well as PD-L1 expressing macrophages that contribute to the inert tumor microenvironment. These are key features of aggressive prostate cancer that may serve as potential biomarkers and therapeutic targets.


2012 ◽  
Vol 30 (5_suppl) ◽  
pp. 181-181 ◽  
Author(s):  
Lawrence Fong ◽  
Vivian K. Weinberg ◽  
John M Corman ◽  
Christopher L Amling ◽  
Robert A Stephenson ◽  
...  

181 Background: Sipuleucel-T is an FDA-approved autologous cellular therapy that has been demonstrated to prolong overall survival in patients with asymptomatic or minimally symptomatic metastatic castrate resistant prostate cancer (mCRPC). To better understand the immunologic effects of sipuleucel-T, an open-label Phase 2 study (P07‐1; NCT00715104 ) of sipuleucel-T prior to radical prostatectomy (RP) was undertaken in patients with localized prostate cancer. Methods: Patients received 3 infusions of sipuleucel-T at approximately 2-week intervals, beginning 6–7 weeks prior to RP. Prostate biopsies (pre-treatment) and tissue from RP (post-treatment) were assessed for the presence of lymphocytes by immunohistochemistry (IHC). Results: The median age of the 42 enrolled patients was 61 years, and all had an ECOG performance status of 0. Thirty-eight patients received all 3 pre-RP infusions of sipuleucel-T. To date, tissue IHC analysis has been completed in 19 patients. Treatment-related AEs were manageable and reversible. Sipuleucel-T did not appear to impact surgery, as judged by operative complications, procedure time, and estimated blood loss. Frequent events that occurred ≤1 day after infusion (>10% of patients) were fatigue, headache, and myalgia. Significant increases (>2‐fold) in CD3+ and CD4+ T cells populations were observed at the tumor rim (where benign and malignant glands interface), compared with the pre-treatment biopsy (ANOVA post hoc Newman-Keuls test: p=0.0002, 0.0002, respectively). CD8+ T cells or CD56+ cells were not significantly increased at the tumor rim compared with benign biopsy regions. Conclusions: Neoadjuvant sipuleucel-T treatment appears to result in an increased frequency of T cells in prostate cancer tissue at the rim between the benign and malignant glands. These data suggest that sipuleucel-T may modulate the presence of lymphocytes at the prostate tumor site. Work is ongoing to more fully characterize the immune response within the prostate tumor tissue and in the peripheral blood.


2012 ◽  
Vol 30 (5_suppl) ◽  
pp. 178-178
Author(s):  
Nadeem A. Sheikh ◽  
Johnna D. Wesley ◽  
Nikole Perdue ◽  
Frances P. Stewart ◽  
Lawrence Fong

178 Background: Sipuleucel-T is an FDA-approved autologous cellular therapy that has been demonstrated to prolong overall survival in subjects with asymptomatic or minimally symptomatic metastatic castrate resistant prostate cancer (mCRPC). Methods: Subjects with localized prostate cancer were enrolled in an open-label, Phase 2 study (P07-1; NCT00715104 ) in which they received 3 infusions of sipuleucel-T at approximately 2-week intervals, beginning 6–7 weeks prior to radical prostatectomy. All samples were evaluated for cellular composition and antigen presenting cell (APC) activation pre- and post-culture with PA2024, a fusion protein comprising prostatic acid phosphatase and granulocyte macrophage-colony stimulating factor. Additionally, cytokines within the culture supernatant were assessed, and T and B cell activation were evaluated pre- and post-culture using flow cytometry. Results: Of the 42 enrolled subjects (median age: 61 years; 98% Caucasian), 38 received all 3 infusions of sipuleucel-T. Consistent with previous trials in mCRPC, CD54 upregulation (APC activation) was greater at the second and third infusions relative to the first. While the percent of CD4+ and CD8+ T cells was unchanged with treatment, the expression of early T cell activation markers (CD134, CD137, CD278 and CD279) was increased in pre-culture cells obtained after the first infusion, and further increased post-culture. Similarly, while the percent of B cells was unchanged, there was a progressive increase in memory B cells (CD20+CD27+IgD−CD86+; pre- and post-culture) and activated mature B cells (CD20+CD27+IgD+CD86+; post-culture) following the first infusion. Activated T cell-associated cytokines were significantly elevated (TNF-α, P < 0.001; IFN-γ, P < 0.001; and IL-2, P < 0.001) in the second and third products. Conclusions: Neoadjuvant sipuleucel-T resulted in robust immune system activation that included APCs, memory and activated mature B cells, and both CD4+ and CD8+ T cells. The patterns observed at the second and third infusions, relative to the first, are consistent with an immunological prime-boost profile.


2018 ◽  
Vol 109 (7) ◽  
pp. 2109-2118 ◽  
Author(s):  
Lijun Mo ◽  
Xinji Zhang ◽  
Xiaojun Shi ◽  
Lili Wei ◽  
Dianpeng Zheng ◽  
...  

Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 3075-3075
Author(s):  
Jagdeep S. Walia ◽  
Jianhui Cai ◽  
Daniel H. Fowler ◽  
Jeffrey A. Medin4

Abstract Prostate cancer (Pca) is the most frequently diagnosed cancer in American men, with an estimated 230,110 cases expected in 2004. Despite various treatment strategies for patients including androgen ablation, radical prostatectomy, radiotherapy, and chemotherapy, the incidence of recurrence remains high and there is limited impact on survival, specially for metastatic disease. Our strategy involves the use of genetically-modified dendritic cells (DCs) to induce an immune response. We have previously demonstrated in a murine model that mature DCs engineered to express prostate tumor-associated antigens (TAAs) can stimulate immune system to specifically target TAA-expressing tumor cells. In view of the heterogenous nature of Pca, we hypothesized that stimulating the immune system against two antigens simultaneously may augment the anti-tumor activity. We generated murine DCs from whole bone marrow from mice by culturing them in granulocytemonocyte colony stimulating factor (GM-CSF) and IL-4 (20ng/ml each) and later with TNF-α. During the DC development, they were transduced with a concentrated oncoretrovirus that engineers the coexpression of prostate specific antigen (PSA) and CD25 (a cell surface marker for tranduced cells) (DC-PSA) or solely the expression of prostate specific membrane antigen (DC-PSMA). Transductions of DCs resulted in 30–60% expression of the either CD25 or PSMA as checked by flowcytometry. These DCs displayed high expression of DC markers like CD11c, CD80, CD86, CD40 and MHC class II molecules. There was no change in their allostimulatory capacity as checked by mixed lymphocyte reaction. Later, mice were injected either with DC non-transduced(NT), DC-PSA, with DC-PSMA. After two immunizations at different time points, the splenocytes were collected from all the groups one week after the last immunization. These splenocytes were stimulated to become effectors and were subsequently analysed to check for IFN-γ secretion, IL-10 secretion and cytolytic assays, using the targets as syngeneic murine prostate tumor cells, RM1 engineered to express PSA and PSMA. The effectors showed high IFN-γ and high cytolytic activity low IL-10 secretion as compared to controls. Our next step will be to test the increase of the levels of IFN-γ secretion and cytolytic activity in the mice immunized with DC-PSA and DC-PSMA both as compared to DC-PSA alone and DC-PSMA alone. To show clinically feasibility of our approach, we extended our work to human cells. HuDCs were generated using human CD34+ hematopoietic cells by culturing them in GM-CSF, SCF, Flt3L and TNF-α for 12 days. During DC production, they were transduced to express PSA or PSMA using a concentrated oncoretrovirus. They were checked for DC markers and the expression of the respective TAAs i.e PSA (CD25) or PSMA. Later, these cells were co-cultured with autologous T-cells. When these immunized T cells were used as effectors against the HLA-matched prostate cancer cell lines expressing PSA and PSMA, they showed high IFN-γ secretion and Low IL-10 secretion as compared controls. Thus, we have found that human DCs can be used to sensitize T cells to show antitumor responses and we are going to test in murine model the augmentation of such antitumour response by using multiple antigen immunotherapy approach.


2019 ◽  
Vol 37 (36) ◽  
pp. 3507-3517 ◽  
Author(s):  
Douglas G. McNeel ◽  
Jens C. Eickhoff ◽  
Laura E. Johnson ◽  
Alison R. Roth ◽  
Timothy G. Perk ◽  
...  

PURPOSE We previously reported the safety and immunologic effects of a DNA vaccine (pTVG-HP [MVI-816]) encoding prostatic acid phosphatase (PAP) in patients with recurrent, nonmetastatic prostate cancer. The current trial evaluated the effects of this vaccine on metastatic progression. PATIENTS AND METHODS Ninety-nine patients with castration-sensitive prostate cancer and prostate-specific antigen (PSA) doubling time (DT) of less than 12 months were randomly assigned to treatment with either pTVG-HP co-administered intradermally with 200 μg granulocyte-macrophage colony-stimulating factor (GM-CSF) adjuvant or 200 μg GM-CSF alone six times at 14-day intervals and then quarterly for 2 years. The primary end point was 2-year metastasis-free survival (MFS). Secondary and exploratory end points were median MFS, changes in PSA DT, immunologic effects, and changes in quantitative 18F-sodium fluoride (NaF) positron emission tomography/computed tomography (PET/CT) imaging. RESULTS Two-year MFS was not different between study arms (41.8% vaccine v 42.3%; P = .97). Changes in PSA DT and median MFS were not different between study arms (18.9 v 18.3 months; hazard ratio [HR], 1.6; P = .13). Preplanned subset analysis identified longer MFS in vaccine-treated patients with rapid (< 3 months) pretreatment PSA DT (12.0 v 6.1 months; n = 21; HR, 4.4; P = .03). PAP-specific T cells were detected in both cohorts, including multifunctional PAP-specific T-helper 1–biased T cells. Changes in total activity (total standardized uptake value) on 18F-NaF PET/CT from months 3 to 6 increased 50% in patients treated with GM-CSF alone and decreased 23% in patients treated with pTVG-HP (n = 31; P = .07). CONCLUSION pTVG-HP treatment did not demonstrate an overall increase in 2-year MFS in patients with castration-sensitive prostate cancer, with the possible exception of a subgroup with rapidly progressive disease. Prespecified 18F-NaF PET/CT imaging conducted in a subset of patients suggests that vaccination had detectable effects on micrometastatic bone disease. Additional trials using pTVG-HP in combination with PD-1 blockade are under way.


Sign in / Sign up

Export Citation Format

Share Document