A Study of Armed, Activated T-Cells in Patients With Advanced Pancreatic Cancer

Author(s):  
2015 ◽  
Vol 3 (Suppl 2) ◽  
pp. P55 ◽  
Author(s):  
Lawrence G Lum ◽  
Minsig Choi ◽  
Archana Thakur ◽  
Abhinav Deol ◽  
Kristie Fields ◽  
...  

2018 ◽  
Vol 36 (15_suppl) ◽  
pp. 4108-4108 ◽  
Author(s):  
Lawrence G. Lum ◽  
Minsig Choi ◽  
Tri Minh Le ◽  
Archana Thakur ◽  
Abhinav Deol ◽  
...  

2020 ◽  
Vol 5 (1) ◽  
Author(s):  
Mao Lin ◽  
Xiaoyan Zhang ◽  
Shuzhen Liang ◽  
Haihua Luo ◽  
Mohammed Alnaggar ◽  
...  

Abstract Immunotherapy has limited efficacy against locally advanced pancreatic cancer (LAPC) due to the presence of an immunosuppressive microenvironment (ISM). Irreversible electroporation (IRE) can not only induce immunogenic cell death, but also alleviate immunosuppression. This study aimed to investigate the antitumor efficacy of IRE plus allogeneic γδ T cells in LAPC patients. A total of 62 patients who met the eligibility criteria were enrolled in this trial, then randomized into two groups (A: n = 30 and B: n = 32). All patients received IRE therapy and after receiving IRE, the group A patients received at least two cycles of γδ T-cell infusion as one course continuously. Group A patients had better survival than group B patients (median OS: 14.5 months vs. 11 months; median PFS: 11 months vs. 8.5 months). Moreover, the group A patients treated with multiple courses of γδ T-cell infusion had longer OS (17 months) than those who received a single course (13.5 months). IRE combined with allogeneic γδ T-cell infusion is a promising strategy to enhance the antitumor efficacy in LAPC patients, yielding extended survival benefits. ClinicalTrials.gov ID: NCT03180437.


2019 ◽  
Vol 37 (4_suppl) ◽  
pp. 196-196
Author(s):  
Junjie Hang ◽  
Lixia Wu ◽  
Kequn Xu

196 Background: Naive and memory T cells play a pivotal role in solid tumor pathogenesis but their role in pancreatic cancer progression remains elusive. Thus, we aimed to investigate their clinical potential in advanced pancreatic cancer (APC). Methods: Flow cytometry was performed to evaluate the level of peripheral naive and memory T cells from APC patients. Interrelationships between naive, memory T cells and clinicopathological variables were evaluated using pearson’s correlation. The prognostic impact of naive and memory T cells were assessed by Kaplan-Meier analysis and Cox regression. The correlation between naive/memory T cells and tumor progression was investigated by Student’s t test. Results: CD4+ naive/memory ratio showed close correlations with hemoglobin, red blood cell (RBC), absolute neutrophil count (ANC) and platelet while CD8+ naive/memory ratio was correlated with hemoglobin, RBC and CEA. Higher baseline lever of CD4+CD45RO+/CD4+ was correlated with better overall survival (OS) (P = 0.036). Patients with CD4+ naive/memory ratio ≥ 0.36 had a poorer OS than those with CD4+ naive/memory ratio < 0.36 (P = 0.021). In addition, CD4+ naive/memory ratio showed independent prognostic impact (HR 1.427, 95%CI 1.033-1.973, P = 0.031). Furthermore, poorer clinical response was correlated with higher level of CD8+ naive/memory ratio after the third cycle of chemotherapy (P = 0.01). Besides, patients with an low level of CD8+ naive/memory ratio had longer progression-free survival (PFS) (P = 0.028). Conclusions: We propose CD4+ naive/memory ratio as a novel prognostic biomarker for APC. In addition, CD8+ naive/memory ratio can be a candidate marker for predicting PFS and the change of its level may reflect the progression of APC.


2020 ◽  
Vol 9 (1) ◽  
pp. 1773201 ◽  
Author(s):  
Lawrence G. Lum ◽  
Archana Thakur ◽  
Minsig Choi ◽  
Abhinav Deol ◽  
Vidya Kondadasula ◽  
...  

2019 ◽  
Vol 37 (15_suppl) ◽  
pp. 4135-4135 ◽  
Author(s):  
Lawrence G. Lum ◽  
Tri Minh Le ◽  
Minsig Choi ◽  
Archana Thakur ◽  
Matthew Reilley ◽  
...  

4135 Background: Conventional chemotherapy (chemo) for locally advanced pancreatic cancer (LAPC) and metastatic pancreatic cancer (MPC) has dismal responses and poor survival rates. Arming activated T cells (ATC) with anti-CD3 x anti-EGFR bispecific antibody (BATs) makes every ATC into an EGFR-specific cytotoxic T cell that secretes cytokines, proliferates, and kills tumor. Methods: We report on 5 phase I (P1) and 15 phase II (P2) patients. In our phase I study, BATs were used to treat LAPC or MPC patients at Karmanos Cancer Institute (NCT0140874) in a dose escalation involving 3 weekly infusions of 1, 2, and 4 x 1010 BATs/infusion, followed by a booster infusion at 3 months (mos) for a total of up to 8 x 1010 BATs. No dose limiting toxicities were observed in the outpatient infusions. Fifteen patients treated on a phase II (NCT02620865) at KCI and (NCT03269526) at University of Virginia received biweekly infusions of 1010 BATs/infusion over 4 weeks for a total of 8 x 1010 EGFR BATs. Results: Four patients had stable disease (SD) for 6.1, 6.5, 5.3, and 36 mos. Two patients had complete responses (CR) when chemo was restarted after BATs. The median overall survival (OS) for 17 evaluable patients (3 of 4 infusions in the P1 and all 8 infusions in the P2) was 31 mos, and the median OS for all 20 patients (3 in the P2 who did not complete 8 infusions) is 14.5 mos (95% CI, 7.5-45.2 mos). Patient IT20104 had an apparent “pseudoprogression” after 3 BATs infusions, but achieved a CR after restarting capcitabine and is alive off therapy at 54 mos (24 mos after stopping capecitabine). Immune evaluations on the P1 patients show specific cytotoxicity to MiaPaCa-2 by peripheral blood mononuclear cells (PBMC) increased from 21% to 31% 2 weeks after the 3rd infusion, and IFN-γ EliSpots increased from < 20 to 1000 IFN-γ EliSpots/106 PBMC (p < 0.03). Patient IT 20121 (SD for 36 mos) increased IFN-γ EliSpots from 250 to 3200/106 PBMC after 8 infusions. Innate cytotoxicity responses in the P1 patients increased significantly after infusions (p < 0.04). Levels of IP-10 increased significantly (p < 0.04), and levels of IL-8 decreased but not significantly (p < 0.07). Conclusions: Infusions of BATs are safe and induce endogenous adaptive anti-tumor responses. Targeting PC with BATs may stabilize disease, leading to improved OS, as well as evidence that BATs infusions can induce anti-tumor activity and immunosensitize tumors to subsequent chemo. Clinical trial information: NCT014084,NCT03269526,NCT02620865.


2019 ◽  
Vol 37 (15_suppl) ◽  
pp. e15756-e15756
Author(s):  
Qin Liu ◽  
Zhengyun Zou ◽  
Baorui Liu ◽  
Weiwei Kong ◽  
Fangjun Chen ◽  
...  

e15756 Background: Pancreatic cancer (PC) is one of the most aggressive and death-relating malignancy. Gemcitabine (GEM) is the key agent in the first-line standard regimen for advanced PC, which is mostly diagnosed at advanced stage and unsuitable for curative resection. The objective responsive rate (ORR) and medium progression free survival (PFS) of various GEM-based regimens are still unsatisfied. Therefore, development of new therapeutic modalities, including immunotherapy, is needed. This study is to investigate the efficacy, safety and clinical beneficial of combination neoantigen based immunotherapy with GEM and radiotherapy in locally advanced and metastatic PC patients. Methods: Three locally advanced unresectable and seven metastatic PC patients received at least two cycles of GEM (1000mg/m2 on day 1 and day 6), radiotherapy combing with neoantigen induced DC vaccination on day 7 and cytotoxic T lymphocyte transfer from day 12 to 15 (repeated every 21 days). The locally advanced unresectable PC patients received stereotactic body radiotherapy (SBRT) with a total amount of 50-66Gy during the first cycle. For metastatic patients, their partial lesions received a low dose radiation (0.5Gy bid*2days ) on day 10 and 11 in each cycle. Results: Two cases were observed with partial remission (PR), five with stable disease (sd), and three with progressive disease (PD). The disease control rate (DCR) was 70%. Median progression free survival (PFS) was 6.4 months. After the first treatment cycle, the total effectiveness for pain easement and increasing appetite are 100% (8/8)and 66.7%, respectively. Haematotoxicities with a 40% incidence rate were the most common adverse drug reactions. Two patients had grade 1 to 2 neutropenia, two with grade 3 to 4 thrombocytopenia. Three patients suffered grade 1 to 2 gemcitabine-induced skin rash. No treatment-related mortality occurred. Conclusions: Neoantigen reactive T cells combined chemoradiotherapy demonstrated an acceptable response and safety in advanced pancreatic cancer patients.


2020 ◽  
Vol 38 (15_suppl) ◽  
pp. 4632-4632 ◽  
Author(s):  
Joanne Lundy ◽  
Gavin M. Marx ◽  
Jennifer MacDiarmid ◽  
Himanshu Brahmbhatt ◽  
Vinod Ganju

4632 Background: Targeted EDV nanocells loaded with doxorubicin and microRNA16a have shown excellent safety profiles in Phase I trials in recurrent glioma and mesothelioma. This planned safety analysis of an ongoing first-in-human, open label Phase I/IIa study in patients with treatment-refractory metastatic pancreatic cancer, assesses safety, biologic and clinical activity of EGFR-targeted EDV nanocells carrying cytotoxic drug PNU-159682, designed to overcome drug resistance, combined with EDV nanocells carrying immunomodulatory adjuvant α-galactosyl ceramide, designed to stimulate anti-tumour immune response. Methods: 9 patients with advanced pancreatic cancer enrolled in the dose escalation phase to evaluate safety of the EDV combination. Doses gradually escalated from 2 x 109 EDVs/dose to a maximum of 7 x 109 EDVs/dose in Week 7, with subsequent dosing at the maximum dose achieved in Cycle 1. iRECIST criteria was used to assess tumour response after each cycle, and blood was collected each cycle for cytokine and PBMC analysis. Results: Combination EDVs were well tolerated with no DLTs, and no drug related SAEs. A minority of patients experienced G1 infusion reactions, which responded promptly to supportive treatment. PR or SD was achieved at 8 weeks in 8/9 patients (CBR 89%), with responses confirmed at 4 months in 4/5 evaluable patients (80%), with 2 durable responses seen beyond 6 months. Exploratory analyses have revealed elevation of IFN-α and IFN-γ in almost all evaluable patients (6/8). In addition, we observed elevated CD8+ T cells (2/8), iNKT, dendritic and NK cells (3/8), and a reduction in exhausted CD8+ T cells (3/8), suggesting activation of both innate and adaptive immune responses. Conclusions: EDVs carrying the cytotoxic drug and immune adjuvant are safe and well tolerated. Early signals point to durable responses, possibly related to the development of an innate and adaptive immune response along with cytotoxic effects on drug resistant tumour cells. The Phase IIa study plans to enrol an additional 35 patients to further evaluate safety and anti-tumour efficacy. Clinical trial information: ACTRN12619000385145 .


2020 ◽  
Vol 8 (Suppl 3) ◽  
pp. A189-A189
Author(s):  
Shannon Oda ◽  
Kristin Anderson ◽  
Philip Greenberg ◽  
Nicolas Garcia ◽  
Pranali Ravikumar ◽  
...  

BackgroundAdoptive cell therapy (ACT) with genetically-modified T cells has shown impressive results against some hematologic cancers, but limited efficacy against tumors with restrictive tumor microenvironments (TMEs). FasL is a particular obstacle for ACT;1 it is expressed in many tumors and TMEs,1 including AML,2 ovarian3 and pancreatic cancers,4 and upregulated on activated T cells, where it can mediate activation-induced cell death (AICD).5MethodsWe engineered T cells to boost function with novel immunomodulatory fusion proteins (IFPs) that combine an inhibitory ectodomain with a costimulatory endodomain. Like current checkpoint-blocking therapies, IFPs can abrogate an inhibitory signal, but also provide an often absent costimulatory signal. Additionally, IFP-driven signals are delivered only to the T cells concurrently engineered to be tumor-specific, thereby avoiding systemic T cell activation. For FasL-expressing TMEs, we developed an IFP that replaces the Fas intracellular tail with costimulatory 4-1BB. We tested the the Fas-4-1BB IFP in primary human T cells and in immunocompetent murine models of leukemia and pancreatic cancer.ResultsFas-4-1BB IFP expression enhanced primary human T cell function and enhanced lysis of Panc1 pancreatic tumor cells in vitro. Fas-4-1BB IFP-engineered murine T cells exhibited increased pro-survival signaling, proliferation, antitumor function and altered metabolism in vitro. Notably, the Fas ectodomain is trimeric5 and the 4-1BB intracellular domain requires trimerization to signal.6 In contrast, the CD28 domain is dimeric and did not enhance function when paired with 4-1BB.In vivo, Fas-4-1BB increased T cell persistence and function, and Fas-4-1BB T cell ACT significantly improved survival in a murine AML model. When delivered with a mesothelin-specific TCR, Fas-4-1BB T cells prolonged survival in the autochthonous KPC pancreatic cancer model, increasing median survival to 65 from 37 days (with TCR-only, **P=0.0042). Single-cell RNA sequencing revealed differences in the endogenous tumor-infiltrating immune cells, included changes in cell frequency and programming.ConclusionsWe developed an engineering approach to enhance the in vivo persistence and antitumor efficacy of transferred T cells. Our targeted, two-hit strategy uses a single fusion protein to overcome a death signal prevalent in the TME of many cancers and on activated T cells, and to provide a pro-survival costimulatory signal to T cells. Our results suggest that this fusion protein can increase T cell function when combined with murine or human TCRs, and can significantly improve therapeutic efficacy in liquid and solid tumors, supporting clinical translation.ReferencesYamamoto, T.N., et al., T cells genetically engineered to overcome death signaling enhance adoptive cancer immunotherapy. J Clin Invest 2019.Contini P, et al., In vivo apoptosis of CD8(+) lymphocytes in acute myeloid leukemia patients: involvement of soluble HLA-I and Fas ligand. Leukemia 2007;21(2):p. 253–60.Motz GT, et al., Tumor endothelium FasL establishes a selective immune barrier promoting tolerance in tumors. Nat Med 2014;20(6):p. 607–15.Kornmann M, et al., Fas and Fas-ligand expression in human pancreatic cancer. Ann Surg 2000. 231(3): p. 368–79.Villa-Morales M and J Fernandez-Piqueras, Targeting the Fas/FasL signaling pathway in cancer therapy. Expert Opin Ther Targets 2012;16(1):p. 85–101.Wyzgol, A., et al., Trimer stabilization, oligomerization, and antibody-mediated cell surface immobilization improve the activity of soluble trimers of CD27L, CD40L, 41BBL, and glucocorticoid-induced TNF receptor ligand. J Immunol 2009;183(3):p. 1851–61.


Sign in / Sign up

Export Citation Format

Share Document