scholarly journals Recent Advances in Experimental Dendritic Cell Vaccines for Cancer

2021 ◽  
Vol 11 ◽  
Author(s):  
Ivan Y. Filin ◽  
Kristina V. Kitaeva ◽  
Catrin S. Rutland ◽  
Albert A. Rizvanov ◽  
Valeriya V. Solovyeva

The development of immunotherapeutic methods for the treatment of oncological diseases have made it possible to improve the effectiveness of standard therapies. There was no breakthrough after first using of personalized therapeutic vaccines based on dendritic cells in clinical practice. A deeper study of the biology of dendritic cells, as well as the use of new approaches and agents for antigenic work, have made it possible to expand the field of application of dendritic cell (DC) vaccines and improve the indicators of cancer patients. In addition, the low toxicity of DC vaccines in clinical trials makes it possible to use promising predictions of their applicability in wider clinical practice. This review examines new approaches and recent advances of the DC vaccine in clinical trials.

2021 ◽  
Vol 23 (Supplement_6) ◽  
pp. vi173-vi173
Author(s):  
Kelly Hotchkiss ◽  
Kristen Batich ◽  
Mrinaj Janampalli ◽  
Pam Norberg ◽  
John Sampson

Abstract INTRODUCTION Dendritic cell (DC) vaccines have shown marginal success in treating glioblastoma (GBM), with inefficient vaccine migration a major limitation. Prior evidence from our clinical trials demonstrated that tetanus diphtheria (Td) preconditioning produced greater DC migration to vaccine draining lymph nodes (VDLNs) and long-term survival. Greater DC numbers reaching VDLNs was also associated with long-term survival. We found from preclinical studies and our patients that increased DC migration was dependent upon the chemokine (C-C motif) ligand 3 (CCL3). METHODS The effect of systemic CCL3 treatment on DC vaccine migration (n=5), antigen-specific T cell responses (n=5) and efficacy against orthotopic GL261-OVA and SMA560 tumors (n=10) was studied in C57Bl/6 and VMdK mice. DCs were electroporated with OVA-mRNA or pulsed with ODC1 neoantigen peptide. Median overall survival (mOS) was measure in days (d) post-intracranial implantation. RESULTS Intravenous CCL3 at the time of intradermal DC vaccination resulted in a dose-dependent increase in migration to VDLN (10ug p=0.036, 20ug p< 0.0001, 50ug p< 0.0001). Mean migration levels following CCL3 treatment were similar to Td-preconditioning (p=0.52) but showed significantly less variability between mice. Combined CCL3 and DC vaccination generated more tumor antigen-specific CD8+IFNγ+ T cells 7 days compared to DC vaccine alone (p=0.0045). CCL3+OVA-DC treatment resulted in significantly greater survival compared to OVA-DC alone (mOS 37 vs 19.5 d; p=0.0174) in established GL261-OVA. CCL3 treatment increased survival in mice with established SMA560 tumors treated with neoantigen ODC1 peptide-pulsed DCs (Tumor alone mOS: 21d, DCvac: 25d, CCL3+DCvac: 48d, p=0.002). CONCLUSIONS These data combined with previous success of our DC vaccine clinical trials reflect the potency of CCL3 to enhance DC vaccine-specific migration, immune responses and survival. CCL3 is a novel and safe adjuvant to overcome prior limitations in DC vaccine therapy and may be translatable to increase heterogeneous tumor antigen presentation following vaccine-targeted tumor killing.


2007 ◽  
Vol 25 (18_suppl) ◽  
pp. 8547-8547
Author(s):  
J. W. Fay ◽  
J. Banchereau ◽  
K. Palucka

8547 Background: Previous clinical trials have demonstrated safety and tolerability of cancer antigen-loaded dendritic cells (DCs) in the treatment of metastatic melanoma. Further, DCs administered to patients with metastatic melanoma are immunogenic and induce durable clinical responses even in patients who failed previous cytotoxic therapy. This report is an analysis of long term clinical outcomes of DC vaccinated patients from our institution. Methods: Between March 1999 and February 2005 seventy patients with metastatic melanoma were treated with DC vaccines in four sequential phase I-IIa clinical trials. Most patients had M1b or M1c metastatic disease. DCs were generated either from CD34+ hematopoietic progenitors or from monocytes. Forty nine HLA-A*0201+ patients received vaccines pulsed with melanoma antigen derived peptides (MART-1, MAGE-3, TYR and gp100). Twenty one patients received DCs loaded with killed allogeneic melanoma cells. KLH was used as helper antigen and, in HLA-A*0201+ patients, Flu-Matrix peptide was used as control antigen. Patients received up to eight DC vaccinations over a maximum of 7 months. Results: DC vaccinations were safe and tolerable. Fifteen out of 70 (21%) patients are alive as of November 2006. The median survival in this group of surviving patients is 46 months (range 22–92 months). Three patients had no evidence of disease upon completion of DC vaccinations by clinical and positron emission tomography scanning. Four patients had experienced objective clinical responses by RECIST criteria (2 CRs and 2 PRs) based on clinical examination and computerized tomography or magnetic resonance imaging. These included two patients that had failed previous cytotoxic and cytokine therapy. Eight patients are alive with disease. Five of 15 long term survivors had no additional therapy other than DC vaccinations including a patient with CR of liver lesions who remains free of disease 80 months post DC vaccination. Conclusions: DC vaccines bring clinical benefit and elicit durable responses in a fraction of patients with metastatic melanoma. These vaccines may provide a survival advantage. Future studies are underway to improve DC vaccine efficacy for treatment of metastatic melanoma. No significant financial relationships to disclose.


2021 ◽  
Vol 23 (Supplement_6) ◽  
pp. vi51-vi51
Author(s):  
Kristen Batich ◽  
Duane Mitchell ◽  
Patrick Healy ◽  
James Herndon ◽  
Gloria Broadwater ◽  
...  

Abstract INTRODUCTION Vaccination with dendritic cells (DCs) fares poorly in primary and recurrent glioblastoma (GBM). Moreover, GBM vaccine trials are often underpowered due to limited sample size. METHODS To address these limitations, we conducted three sequential clinical trials utilizing Cytomegalovirus (CMV)-specific DC vaccines in patients with primary GBM. Autologous DCs were generated and electroporated with mRNA encoding for the CMV protein pp65. Serial vaccination was given throughout adjuvant temozolomide cycles, and 111Indium radiolabeling was implemented to assess migration efficiency of DC vaccines. Patients were followed for median overall survival (mOS) and OS. RESULTS Our initial study was the phase II ATTAC study (NCT00639639; total n=12) with 6 patients randomized to vaccine site preconditioning with tetanus-diphtheria (Td) toxoid. This led to an expanded cohort trial (ATTAC-GM; NCT00639639) of 11 patients receiving CMV DC vaccines containing granulocyte-macrophage colony-stimulating factor (GM-CSF). Follow-up data from ATTAC and ATTAC-GM revealed 5-year OS rates of 33.3% (mOS 38.3 months; CI95 17.5-undefined) and 36.4% (mOS 37.7 months; CI95 18.2-109.1), respectively. ATTAC additionally revealed a significant increase in DC migration to draining lymph nodes following Td preconditioning (P=0.049). Increased DC migration was associated with OS (Cox proportional hazards model, HR=0.820, P=0.023). Td-mediated increased migration has been recapitulated in our larger confirmatory trial ELEVATE (NCT02366728) of 43 patients randomized to preconditioning (Wilcoxon rank sum, Td n=24, unpulsed DC n=19; 24h, P=0.031 and 48h, P=0.0195). In ELEVATE, median follow-up of 42.2 months revealed significantly longer OS in patients randomized to Td (P=0.026). The 3-year OS for Td-treated patients in ELEVATE was 34% (CI95 19-63%) compared to 6% given unpulsed DCs (CI95 1-42%). CONCLUSION We report reproducibility of our findings across three sequential clinical trials using CMV pp65 DCs. Despite their small numbers, these successive trials demonstrate consistent survival outcomes, thus supporting the efficacy of CMV DC vaccine therapy in GBM.


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 571-571
Author(s):  
Sarah Herby ◽  
Matthew Milliron ◽  
Crystal Mackall ◽  
Terry Fry

Abstract Background: Although allogeneic graft versus tumor responses represent the most potent form of immunotherapy, recurrent malignancy remains a major cause of post-transplant mortality. Thus, strategies to enhance GVT are needed to improve outcomes. Dendritic cell (DC) vaccines can potently enhance anti-tumor immunity in the autologous setting. The impact of alloreactivity on DC vaccine responses is not known, but the potency of the GVT effect suggests that mild alloreactivity might enhance vaccine responses to third party antigens. Methods: To determine the effect of a minor histocompatibility mismatch on DC vaccine responses, lethally irradiated female C3H.SWxC57BL/6 and C3H.SW recipients were transplanted with T cell depleted (TCD) C3H.SW bone marrow (BM). On days 14 and 28, recipients received donor-type lymph node cells containing mature T cells (DLI, 1 x 10^6, 5 x 10^6, or 10 x 10^6 IV). To immunize against the male antigenic complex (HY), BM-derived, anti-CD40 activated donor strain male DC were administered IP at the same time as the DLI. At Day 35, HY specific immune responses were measured using interferon γ ELISPOT against the dominant (UTY) and subdominant (SMCY) class I epitopes and dominant class II epitope (DBY). In some experiments, rhIL-7 and rhIL-15 were administered (5 mcg/day IP) from day 14 to 35. Results: In syngeneic recipients, robust HY responses were observed at all DLI doses, consistent with rapid T cell reconstitution via the thymus. Allogeneic recipients demonstrated mild weight loss and decreased splenic cellularity as the DLI dose was increased, consistent with the development of mild GVHD. However, even recipients receiving the highest DLI dose recovered completely and had none of the major physical symptoms of GVHD. Increasing DLI dose from 1 x 10^6 to 10 x 10^6 paradoxically led to a marked decrease in the median total number of T cells/spleen responding to UTY, SMCY, and DBY (9196 at 1 x 10^6 vs 897 at 10 x 10^6, p = .01; 15662 vs 2854, p= .02; 10257 vs 4227, p = .02). To test whether the presence of alloreactive T cells were necessary for this effect, C3H.SW minor antigen reactive T cells were deleted from B6-derived DLI. TCD B6 BM was transplanted into C3H.SWxC57BL/6 mice and at 5 weeks LN cells were collected and transferred as DLI. Strikingly, transfer of 10 x 10^6 tolerized DLI did not result in a loss of HY vaccine responses when compared to transfer of non-tolerized DLI (UTY, 3888 vs 21075, p= .05; SMCY, 7135 vs 21126, p = .018; DBY, 7117 vs 29201, p= .006). Although IL-7 and IL-15 can potently enhance DC vaccine responses in the syngeneic setting, these cytokines did not improve vaccine responses in allogeneic recipients. Conclusions: Using a minor mismatched allogeneic transplant model, we have shown that mild GVHD can diminish the ability to respond to third party antigens expressed on activated DC vaccines. This suggests that the potency of GVT does not result from the alloreactive milieu, but rather from the nature of the antigens targeted. The mechanisms involved in the suppression of DC vaccine responses in allogeneic recipients are currently under study. These findings have important implications for the use of tumor antigen expressing vaccines as a modality to improve the graft versus tumor effect.


2021 ◽  
Vol 2 (1) ◽  
Author(s):  
Rui Zhang ◽  
Lin Tang ◽  
Qing Li ◽  
Yaomei Tian ◽  
Binyan Zhao ◽  
...  

AbstractDendritic cell (DC)-based cancer vaccines have so far achieved good therapeutic effects in animal experiments and early clinical trials for certain malignant tumors. However, the overall objective response rate in clinical trials rarely exceeds 15%. The poor efficiency of DC migration to lymph nodes (LNs) (< 5%) is one of the main factors limiting the effectiveness of DC vaccines. Therefore, increasing the efficiency of DC migration is expected to further enhance the efficacy of DC vaccines. Here, we used DP7-C (cholesterol modified VQWRIRVAVIRK), which can promote DC migration, as a medium. Through multiomics sequencing and biological experiments, we found that it is the metabolite pantothenic acid (PA) that improves the migration and effectiveness of DC vaccines. We clarified that both DP7-C and PA regulate DC migration by regulating the chemokine receptor CXCR2 and inhibiting miR-142a-3p to affect the NF-κB signaling pathway. This study will lay the foundation for the subsequent use of DP7-C as a universal substance to promote DC migration, further enhance the antitumor effect of DC vaccines, and solve the bottleneck problem of the low migration efficiency and unsatisfactory clinical response rate of DC vaccines.


Vaccines ◽  
2020 ◽  
Vol 8 (1) ◽  
pp. 25 ◽  
Author(s):  
Caroline Boudousquié ◽  
Valérie Boand ◽  
Emilie Lingre ◽  
Laeticia Dutoit ◽  
Klara Balint ◽  
...  

With the emergence of immune checkpoint inhibitors and adoptive T-cell therapies, there is a considerable interest in using personalized autologous dendritic cell (DC) vaccines in combination with T cell-targeting immunotherapies to potentially maximize the therapeutic impact of DC vaccines. Here, we describe the development and optimization of a Good Manufacturing Practice (GMP)-compliant manufacturing process based on tumor lysate as a tumor antigen source for the production of an oxidized tumor cell lysate loaded DC (OC-DC) vaccine. The manufacturing process required one day for lysate preparation and six days for OC-DC vaccine production. Tumor lysate production was standardized based on an optimal tumor digestion protocol and the immunogenicity was improved through oxidation using hypochloric acid prior to freeze-thaw cycles resulting in the oxidized tumor cell lysate (OC-L). Next, monocytes were selected using the CliniMACS prodigy closed system and were placed in culture in cell factories in the presence of IL-4 and GM-CSF. Immature DCs were loaded with OC-L and matured using MPLA-IFNγ. After assessing the functionality of the OC-DC cells (IL12p70 secretion and COSTIM assay), the OC-DC vaccine was cryopreserved in multiple doses for single use. Finally, the stability of the formulated doses was tested and validated. We believe this GMP-compliant DC vaccine manufacturing process will facilitate access of patients to personalized DC vaccines, and allow for multi-center clinical trials.


2019 ◽  
Vol 21 (Supplement_6) ◽  
pp. vi4-vi4
Author(s):  
Maryam Rahman ◽  
Ashley Ghiaseddin ◽  
Oleg Yegorov ◽  
Changlin Yang ◽  
Anjelika Dechkovskaia ◽  
...  

Abstract BACKGROUND We initiated a Phase 2, randomized, placebo-controlled, clinical trial evaluating the efficacy of autologous CMV pp65-LAMP RNA pulsed dendritic cell vaccines mixed with GM-CSF and administered during cycles of adjuvant dose-intensified temozolomide (ATTAC II, NCT02465268). STUDY OBJECTIVE A patient with partially resected GBM experienced a sustained complete radiographic response after receiving five DC vaccines. Peripheral blood responses were characterized to examine possible immunologic biomarkers concordant with a sustained clinical response. METHODS Patients with newly diagnosed glioblastoma undergoing surgical resection are eligible and randomized 2:1 to DC vaccine arms vs placebo (PBMCs in saline). DC vaccines consist of CMV pp65 RNA conjugated either to the full-length lysosomal associated membrane protein (LAMP) or to the short LAMP signal sequence. Patients undergo leukapheresis for DC generation prior to standard chemoradiation and receive cycle 1 of dose-intensified temozolomide (100 mg/m^2 x 21 days) before first three biweekly intradermal DC vaccines admixed with GM-CSF. Subsequent DC vaccines (total of 10) are given monthly with each diTMZ cycle with an intradermal tetanus-diphtheria booster given 6-24hrs before the third, fifth, seventh, and ninth DC vaccines. PBMCs and serum are collected for immune monitoring. RESULTS A 58-year old white male with partially-resected, MGMT-unmethlyated, p53 mutant, H3.3 mutant, midline glioblastoma was enrolled on the ATTAC II study and experienced a complete radiographic response after the fifth DC vaccine that has been sustained > 10 months. Immune monitoring by Elispot, cytokine array, and single-cell RNA sequencing have revealed significant expansion of CMV pp65-specific immune responses, increased circulating IFNg, and marked systemic expansion of cytotoxic T cells and iNKT cells during vaccination. These responses were sustained through cycles of diTMZ despite profound lymphopenia. CONCLUSIONS CMV pp65-LAMP RNA-pulsed DC vaccination was associated with profound immunologic and clinical response in a patient with MGMT unmethylated midline glioblastoma.


2013 ◽  
Vol 31 (15_suppl) ◽  
pp. 3049-3049
Author(s):  
K Chai ◽  
Yq Ai ◽  
Lw Jiang

3049 Background: There are few choices of treatments for advanced pancreatic carcinoma (PC) due to the resistances to chemo- or radio-therapy. Immunotherapy based on dendritic cell (DC) vaccines and p53-based gene therapy are two promising therapeutic modalities. They also demonstrated favorable safety profiles. In this study, we compared the immunological and clinical response between DC vaccine therapy and DC vaccine combined with recombinant adenovirus-p53 (rAd-p53) gene therapy. Methods: Thirty-six patients with a stage IV pancreatic cancer, 21 men and 15 women with an average age of 56.2 years old, were included in this study and randomly assigned to two groups: 16 patients in DC group (DCG) and 20 in DC plus rAd-p53 group (DPG). The DCG patients received autologous antigen-loaded DC (antigen from isolated pancreatic cancer cells) and the DPG patients received both DC and rAd-p53. DC vaccines were injected intra-dermally once every week for 4 injections and rAd-p53 were given by intravenous injections once per 3 days for 5 times at a dose of 3 x 1012viral particles. The response, safety and peripheral blood lymphocyte subsets were investigated. Results: The post-treatment CD3+, CD3+CD4+, CD4+/CD8+ ratio of patients’ peripheral blood in both groups were increased. But the percent of CD4+CD25+ regulatory T cells were significantly decreases. In DPG, 5 patients had a partial response (PR) and 4 patients had stable disease (SD) according to the RECIST standard. The 3 and 3 DCG patients achieved a PR and SD, respectively. The disease control rates (PR+SD) were 45.0% and 37.5% for DPG and DCG, respectively. The 6-month overall survival rates were 50.0% and 43.8% for DPG and DCG, respectively. The median survival times were 6.8 and 5.5 months for DPG and DCG, respectively. Mild to medium grade fever was observed in most of the patients in the two groups. No serious adverse events were found. Conclusions: DC-based immunotherapy and p53 gene therapy are safe and appropriate treatments for patients with advanced pancreatic carcinoma. The combined treatments showed more beneficial results than the DC immunotherapy alone.


2017 ◽  
Vol 2017 ◽  
pp. 1-9 ◽  
Author(s):  
J. C. Rangel-Reyes ◽  
J. C. Chimal-Eguía ◽  
E. Castillo-Montiel

Therapeutic protocols in immunotherapy are usually proposed following the intuition and experience of the therapist. In order to deduce such protocols mathematical modeling, optimal control and simulations are used instead of the therapist’s experience. Clinical efficacy of dendritic cell (DC) vaccines to cancer treatment is still unclear, since dendritic cells face several obstacles in the host environment, such as immunosuppression and poor transference to the lymph nodes reducing the vaccine effect. In view of that, we have created a mathematical murine model to measure the effects of dendritic cell injections admitting such obstacles. In addition, the model considers a therapy given by bolus injections of small duration as opposed to a continual dose. Doses timing defines the therapeutic protocols, which in turn are improved to minimize the tumor mass by an optimal control algorithm. We intend to supplement therapist’s experience and intuition in the protocol’s implementation. Experimental results made on mice infected with melanoma with and without therapy agree with the model. It is shown that the dendritic cells’ percentage that manages to reach the lymph nodes has a crucial impact on the therapy outcome. This suggests that efforts in finding better methods to deliver DC vaccines should be pursued.


2012 ◽  
Vol 2012 ◽  
pp. 1-15 ◽  
Author(s):  
Fabian Benencia ◽  
Leslee Sprague ◽  
John McGinty ◽  
Michelle Pate ◽  
Maria Muccioli

Many clinical trials have been carried out or are in progress to assess the therapeutic potential of dendritic-cell- (DC-) based vaccines on cancer patients, and recently the first DC-based vaccine for human cancer was approved by the FDA. Herewith, we describe the general characteristics of DCs and different strategies to generate effective antitumor DC vaccines. In recent years, the relevance of the tumor microenvironment in the progression of cancer has been highlighted. It has been shown that the tumor microenvironment is capable of inactivating various components of the immune system responsible for tumor clearance. In particular, the effect of the tumor microenvironment on antigen-presenting cells, such as DCs, does not only render these immune cells unable to induce specific immune responses, but also turns them into promoters of tumor growth. We also describe strategies likely to increase the efficacy of DC vaccines by reprogramming the immunosuppressive nature of the tumor microenvironment.


Sign in / Sign up

Export Citation Format

Share Document