scholarly journals Immune responses and long-term disease recurrence status after telomerase-based dendritic cell immunotherapy in patients with acute myeloid leukemia

Cancer ◽  
2017 ◽  
Vol 123 (16) ◽  
pp. 3061-3072 ◽  
Author(s):  
Hanna J. Khoury ◽  
Robert H. Collins ◽  
William Blum ◽  
Patrick S. Stiff ◽  
Laurence Elias ◽  
...  
Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 2651-2651 ◽  
Author(s):  
Arjan A van de Loosdrecht ◽  
Sandra van Wetering ◽  
Saskia Santegoeds ◽  
Satwinder Kaur Singh ◽  
Malika Koppes ◽  
...  

Abstract Vaccines against tumor associated antigens represent an appealing strategy for preventing tumor recurrence. A novel immunotherapy platform is represented by a dendritic cell vaccine that originates from a human myeloid leukemia cell line, DCOne, which endogenously expresses a range of leukemia associated antigens, including PRAME and WT-1, and can be differentiated into mature dendritic cells (DCs). Mature DC derived from DCOne are being developed to replace patient-derived DC vaccines. The first indication in which vaccination with mature DCs derived from DCOne has been tested clinically is Acute Myeloid Leukemia (AML) because of the high unmet medical need and substantial evidence that AML is a suitable target for immunotherapy. A Phase I/IIa study enrolled 12 AML patients (age range 58-71) who were either in CR1/CR2 (n=5) or had smoldering disease (n=7). Patients had received all available standard care, were at high risk of relapse and ineligible for all available post-remission therapies, including allogeneic stem cell transplantation. Patients were selected for the study because they had an estimated life expectancy of 3-6 months. A standard 3+3 design was used, starting with 4 bi-weekly intradermal DCOne DC vaccinations of 10E6 (n=3), 25E6 (n=3) or 50E6 (n=6) cells. Patients were monitored for clinical and immunological responses for 126 days and surviving patients underwent long-term follow-up after study completion. Primary endpoints were safety and feasibility; secondary endpoints were clinical and immunological responses. Treatment was well tolerated in all patients, with related adverse events mainly limited to injection site reactions. During the 5 months duration of the study 3 patients died: 2 from infections and 1 from leukemia. Clear evidence for induction of multi-functional immune responses was obtained, including increased post vaccination delayed type hypersensitivity reactions, increases in CD4+ and CD8+ T cell proliferative responses and/or sero-conversion to DCOne DCs and/or AML blasts in 6 out of 9 patients. Three of 7 patients who were evaluable by IFNgELISpots showed vaccination-induced reactive T cell responses to WT-1 and/or PRAME, antigens which are present in DCOne. The patients who survived more than 6 months post-vaccination showed strongly prolonged survival. Four patients are still alive 28, 22, 12 and 10 months after study entry and 1 patient survived for 23 months. Patients in CR1 or CR2 at study entry were all in CR at the end of the study and these patients were more likely to exhibit prolonged survival. Patients not in CR at study entry, with one exception, all had persistence of disease at the end of the study and died due to disease progression. Together, these results suggest that patients who have a capable immune system can induce a multi-functional and lasting immune response to the vaccine. As expected this most likely translates into long-term clinical benefit if patients are in CR at the time of vaccination. This is in keeping with the expected kinetics of cancer immunotherapy. We conclude that vaccination with DCOne derived DCs is safe and feasible in elderly AML patients, and generates both cellular and humoral immune responses. The hypothesis that DCOne-derived DCs induce immune responses against the patients’ leukemia cells, translating into clinical benefit in patients with a capable immune system who are in CR at the time of vaccination, will now be investigated in a multi-center randomized Phase II trial in AML patients in first remission. Disclosures: van Wetering: DCPrime: Employment. Kaur Singh:DCPrime: Employment. Hall:DCPrime: Consultancy. Kruisbeek:DCPrime: Employment.


2016 ◽  
Vol 24 ◽  
pp. S110
Author(s):  
Jane S. Lebkowski ◽  
Hanna J. Khoury ◽  
Robert H. Collins ◽  
William Blum ◽  
Patrick Stiff ◽  
...  

2021 ◽  
Vol 9 (Suppl 3) ◽  
pp. A232-A232
Author(s):  
Haoxiao Zuo ◽  
Satwinder Kaur Singh ◽  
Marie-José Van Lierop ◽  
Jorn Kaspers ◽  
Remco Bos ◽  
...  

BackgroundDCP-001 is a cancer relapse vaccine derived from the DCOne® human leukemic cell line. During manufacturing, DCOne® cells are shifted towards a mature dendritic cell (mDC) phenotype, combining an endogenous tumor antigen repertoire (e.g. WT-1, RHAMM and PRAME) with a mDC costimulatory profile and providing the basis for the highly immunogenic vaccine DCP-001. In a phase I clinical study in acute myeloid leukemia (AML), DCP-001 demonstrated to be safe and to induce multifunctional antitumor immune responses.1 It has also been reported that DCP-001 induces antitumor immunity against multiple myeloma cells in peripheral blood mononuclear cells (PBMC) from multiple myeloma patients and that DCP-001 antigenic material is transferred to host antigen presenting cells (APC), possibly via extracellular vesicles.2 However, the possibility of direct interactions between DCP-001 and host APC has not yet been investigated.MethodsTo further elucidate the mode of action of DCP-001, we studied the interactions of DCP-001 with human PBMC and isolated immature monocyte-derived DCs (iMoDC) in in vitro co-culture studies. A human skin explant model was used to determine uptake of DCP-001 by migrating skin DCs after intradermal injection.ResultsWe found that DCP-001 stimulates the secretion of various proinflammatory cytokines (IL-1β, GM-CSF, IFN-γ, IL-2, TNF-α, and IL-6) and chemokines (IL-8 and RANTES) in PBMC. In addition, we demonstrate that DCP-001 is efficiently taken up by iMoDC via direct cell-cell interactions and that this phagocytic process is influenced by ”eat-me” and ”don’t eat me” signaling pathways. Blocking of the ”eat-me” signals calreticulin and phosphatidylserine inhibited the uptake of DCP-001, whereas blockade of the ”don’t eat me” signal CD47 enhanced DCP-001 uptake. After intradermal injection of DCP-001 in an ex-vivo human skin model, its uptake by skin-emigrating DCs was demonstrated as well as simultaneous activation of these DCs.ConclusionsOur data suggest a key role for host antigen presenting cells in the triggering of immune responses upon DCP-001 vaccination. In addition, the data provide rationale for potential combination therapies based on DCP-001 and inhibitors of the CD47 pathway.Referencesvan de Loosdrecht AA, et al. A novel allogeneic off-the-shelf dendritic cell vaccine for post-remission treatment of elderly patients with acute myeloid leukemia. Cancer Immunol Immunother 2018;67(10):1505–1518.Leaf RK, et al. DCOne as an allogeneic cell-based vaccine for multiple myeloma. J Immunother 2017;40(9):315–322.


Author(s):  
Michael Heuser ◽  
B. Douglas Smith ◽  
Walter Fiedler ◽  
Mikkael A. Sekeres ◽  
Pau Montesinos ◽  
...  

AbstractThis analysis from the phase II BRIGHT AML 1003 trial reports the long-term efficacy and safety of glasdegib + low-dose cytarabine (LDAC) in patients with acute myeloid leukemia ineligible for intensive chemotherapy. The multicenter, open-label study randomized (2:1) patients to receive glasdegib + LDAC (de novo, n = 38; secondary acute myeloid leukemia, n = 40) or LDAC alone (de novo, n = 18; secondary acute myeloid leukemia, n = 20). At the time of analysis, 90% of patients had died, with the longest follow-up since randomization 36 months. The combination of glasdegib and LDAC conferred superior overall survival (OS) versus LDAC alone; hazard ratio (HR) 0.495; (95% confidence interval [CI] 0.325–0.752); p = 0.0004; median OS was 8.3 versus 4.3 months. Improvement in OS was consistent across cytogenetic risk groups. In a post-hoc subgroup analysis, a survival trend with glasdegib + LDAC was observed in patients with de novo acute myeloid leukemia (HR 0.720; 95% CI 0.395–1.312; p = 0.14; median OS 6.6 vs 4.3 months) and secondary acute myeloid leukemia (HR 0.287; 95% CI 0.151–0.548; p < 0.0001; median OS 9.1 vs 4.1 months). The incidence of adverse events in the glasdegib + LDAC arm decreased after 90 days’ therapy: 83.7% versus 98.7% during the first 90 days. Glasdegib + LDAC versus LDAC alone continued to demonstrate superior OS in patients with acute myeloid leukemia; the clinical benefit with glasdegib + LDAC was particularly prominent in patients with secondary acute myeloid leukemia. ClinicalTrials.gov identifier: NCT01546038.


Sign in / Sign up

Export Citation Format

Share Document