Donna Yancey's research while affiliated with Duke University and other places

What is this page?


This page lists the scientific contributions of an author, who either does not have a ResearchGate profile, or has not yet added these contributions to their profile.

It was automatically created by ResearchGate to create a record of this author's body of work. We create such pages to advance our goal of creating and maintaining the most comprehensive scientific repository possible. In doing so, we process publicly available (personal) data relating to the author as a member of the scientific community.

If you're a ResearchGate member, you can follow this page to keep up with this author's work.

If you are this author, and you don't want us to display this page anymore, please let us know.

Publications (11)


Fig. 1. Characterization of Lin - HLA-DR - myeloid cell population in the peripheral blood of renal cell carcinoma patients. A, MDSC were isolated from the PBMC of healthy donors and renal cell carcinoma patients (one representative pair of donor-patient from 9 are shown) by selecting lineage negative (CD3, CD14, CD19, CD56) and HLA-DR ^ negative cells as described previously (22). B, proportion of Lin - HLA-DR - cells in the peripheral blood of renal cell carcinoma patients is significantly increased. Freshly isolated PBMC from renal cell carcinoma patients and healthy donors were stained with CD3, CD14, CD19, CD56, and HLA-DR, and then analyzed by flow cytometry for presence of Lin - HLA-DR - cell population. C, Lin - HLA-DR - MDSC populations were characterized by flow cytometry.White histograms, cell surface marker expression; gray 
Fig. 2. Isolation of CD33 + HLA-DR - MDSC populations. A, PBMC obtained from a 
Fig. 3. Renal cell carcinoma ^ derived CD33 + HLA-DR-cells inhibit T-cell responses. A, ELISPOTassay. MDSC isolated from a HLA-A0201-positive healthy volunteer (Donor) or a renal cell carcinoma patient (RCC) were pulsed with MART-1peptide and incubated with a MART-1 ^ specific CTL clone at a ratio of 1:1for 20 h. Assay was developed as described in Materials and Methods. After washing, spots were counted using an automated ELISPOTcounter. B, CTL assays were done in the presence of MDSC isolated from a renal cell carcinoma patient as described in the legend to Fig. 2B.We show that MDSC isolated from a renal cell carcinoma patient (RCC MDSC), but not from a healthy donor (donor CD33 + cells) inhibited cytolytic activity of MART-1 ^ specifc CTL. Data were obtained from triplicates of single experiment representative of two. C, effect of renal cell carcinoma-derived MDSC on tetanus toxoid-induced T-cell proliferation. Dendritic cells derived from a HLA-A0201-positive healthy volunteer were pulsed with tetanus toxoid (0.1mg/mL for 1h in 37jC), washed and then cultured with autologousTcells (1:5) in round-bottomed 96-well plates for 4 d in the presence or absence of renal cell carcinoma ^ derived MDSC. [ 3 H]thymidine (1 ACi/well) was added 18 h before cell harvest. Thymidine uptake was measured using a liquid scintilla.
Fig. 4. Involvement of reactive oxygen species and NO in mechanisms of immune suppression mediated by CD33 + HLA-DR - cells. To evaluate the role of reactive oxygen species in MDSC-mediated immunosuppression, catalase and/or superoxide dismutase ( SOD ) or uric acid were added to IFN- g ELISPOTassays in the presence of MDSC isolated from a renal cell carcinoma donor ( A ) and a healthy volunteer ( B ). MDSC isolated from an HLA-A0201-positive healthy volunteer (Donor) or a renal cell carcinoma patient (RCC) were pulsed with MART-1peptide and incubated with a MART-1specific CTL clone at a ratio of 1:1for 20 h. Assay was developed as described in Material and Methods. After washing, spots were counted using an automated ELISPOTcounter. Antigen-specific production of reactive oxygen species and NO was measured by flow cytometry. MDSC were incubated with MART-1specific clone, and after 1h, 5 A mol/L of H 2 DCFDA ( C ) or 5 A mol/L DAF-FM diacetate ( D ) were added for 20 min. After washing, CD3-negative cells were analyzed by fluorescence-activated cell sorting (MDSC incubated with MART-1 ^ specific CTL in the absence of fluorogenic probes are shown as gray histograms). Results of representative experiment out of two are shown. 
Fig. 5. Reverse transcription-PCR analysis of renal cell carcinoma-derived MDSC. Total RNA from dendritic cells (DC), HL-60 cell line, MDSC, and lineage-negative/ DR+ renal cell carcinoma-derived cells (Lin-/DR+) were isolated usingTrizol reagent (Invitrogen). Reverse transcription was done at 25 j C for 10 min, 42 j C for 2 h, and 72 j C for 5 min from 100 ng of total RNA using Superscript II reverse transcriptase (Invitrogen) and random primers (3 A g/ A L; Invitrogen). A, expression of receptors for ATRA; B, expression of MDSC - related genes. 

+1

Reversal of Myeloid Cell - Mediated Immunosuppression in Patients with Metastatic Renal Cell Carcinoma
  • Article
  • Full-text available

January 2009

·

547 Reads

·

279 Citations

Clinical Cancer Research

·

Zhen Su

·

·

[...]

·

Johannes Vieweg

Tumor-induced immunosuppression remains a significant obstacle that limits the efficacy of biological therapy for renal cell carcinoma. Here we evaluate the role of CD33 myeloid-derived suppressor cells (MDSC) in the regulation of T-cell responses in renal cell carcinoma patients. We also examine effect of all-trans-retinoic acid (ATRA) on MDSC-mediated immune suppression. CD33-positive myeloid cells were isolated from the peripheral blood of renal cell carcinoma patients with magnetic beads and tested in vitro for their ability to inhibit T-cell responses. T-cell function was evaluated using ELISPOT and CTL assays. MDSC isolated from renal cell carcinoma patients, but not from healthy donors, were capable of suppressing antigen-specific T-cell responses in vitro through the secretion of reactive oxygen species and nitric oxide upon interaction with CTL. MDSC-mediated immune suppression and IFN-gamma down-regulation was reversible in vitro by exposing cells to the reactive oxygen species inhibitors. Moreover, ATRA was capable of abrogating MDSC-mediated immunosuppression and improving T-cell function by direct differentiation into antigen-presenting cell precursors. These results may have significant implications regarding the future design of active immunotherapy protocols that may include differentiation agents as part of a multimodal approach to renal cell carcinoma immunotherapy.

Download
Share


Dannull J, Su Z, Rizzieri D, Yang BK, Coleman D, Yancey D, Zhang A, Dahm P, Chao N, Gilboa E, Vieweg JEnhancement of vaccine-mediated antitumor immunity in cancer patients after depletion of regulatory T cells. J Clin Invest 115: 3623-3633

January 2006

·

136 Reads

·

838 Citations

The Journal of clinical investigation

In this study, we investigated whether elimination of CD4+/CD25+ Tregs using the recombinant IL-2 diphtheria toxin conjugate DAB(389)IL-2 (also known as denileukin diftitox and ONTAK) is capable of enhancing the immunostimulatory efficacy of tumor RNA-transfected DC vaccines. We show that DAB(389)IL-2 is capable of selectively eliminating CD25-expressing Tregs from the PBMCs of cancer patients without inducing toxicity on other cellular subsets with intermediate or low expression of CD25. DAB(389)IL-2-mediated Treg depletion resulted in enhanced stimulation of proliferative and cytotoxic T cell responses in vitro but only when DAB(389)IL-2 was omitted during T cell priming. DAB(389)IL-2 significantly reduced the number of Tregs present in the peripheral blood of metastatic renal cell carcinoma (RCC) patients and abrogated Treg-mediated immunosuppressive activity in vivo. Moreover, DAB(389)IL-2-mediated elimination of Tregs followed by vaccination with RNA-transfected DCs significantly improved the stimulation of tumor-specific T cell responses in RCC patients when compared with vaccination alone. Our findings may have implications in the design of immune-based strategies that may incorporate the Treg depletion strategy to achieve potent antitumor immunity with therapeutic impact.


Telomerase mRNA-Transfected Dendritic Cells Stimulate Antigen-Specific CD8+ and CD4+ T Cell Responses in Patients with Metastatic Prostate Cancer

April 2005

·

139 Reads

·

378 Citations

The Journal of Immunology

Telomerase reverse transcriptase (hTERT) represents an attractive target for cancer immunotherapy because hTERT is reactivated in most human tumors. A clinical trial was initiated in which hTERT mRNA-transfected dendritic cells (DC) were administered to 20 patients with metastatic prostate cancer. Nine of these subjects received DC transfected with mRNA encoding a chimeric lysosome-associated membrane protein-1 (LAMP) hTERT protein, allowing for concomitant induction of hTERT-specific CD8+ and CD4+ T cell responses. Treatment was well tolerated. Intense infiltrates of hTERT-specific T cells were noted at intradermal injection sites after repeated vaccination. In 19 of 20 subjects, expansion of hTERT-specific CD8+ T cells was measured in the peripheral blood of study subjects, with 0.9-1.8% of CD8+ T cells exhibiting Ag specificity. Patients immunized with the chimeric LAMP hTERT vaccine developed significantly higher frequencies of hTERT-specific CD4+ T cells than subjects receiving DC transfected with the unmodified hTERT template. Moreover, CTL-mediated killing of hTERT targets was enhanced in the LAMP hTERT group, suggesting that an improved CD4+ response could augment a CTL response. Vaccination was further associated with a reduction of prostate-specific Ag velocity and molecular clearance of circulating micrometastases. Our findings provide a rationale for further development of hTERT-transfected DC vaccines in the treatment of prostate and other cancers.


Fig. 1. Expression of TAAs by tumor and benign renal tissues. Analysis of TAA gene expression using RT-PCR. cDNA, generated from histologically confirmed renal tumor and the corresponding benign tissues, was amplified by 30 cycles of PCR and subjected to electrophoresis on 1.5% agarose gels. PCR products were visualized by ethidium bromide staining. As controls, RNA species extracted from HeLa cells ( H ; G250 positive , 
Fig. 2. Increase of tumor-specific T cells after vaccination with renal tumor RNA-transfected DCs. T cells isolated from pre- and postvaccination peripheral blood samples were 
Fig. 4. SSCP analysis of TCR V ␤ utilization. PBMCs from a renal cancer patient were stimulated twice with autologous DCs transfected with renal tumor RNA ( RCC ), hTERT RNA ( hTERT ), G250 RNA ( G250 ), or OFA RNA ( OFA ) to generate CTLs. PCR using V ␤ -specific primers and a fluorescent C ␤ -specific primer was performed followed by SSCP analysis using cDNA from CTL lines. Autologous renal tumor tissue ( TIL ) or CTLs generated against GFP were used as controls. As exemplified for V ␤ 11, V ␤ 22, and V ␤ 19, the following SSCP banding patterns were identified (see arrows ): 1, patterns unique for RCC-specific CTLs; 2, patterns present in both hTERT- and RCC-specific CTLs; 3, patterns present in RCC-specific CTLs, OFA-specific CTLs, and TILs. 
Immunological and clinical responses in metastatic renal cancer patients vaccinated with tumor RNA-transfected dendritic cells

June 2003

·

412 Reads

·

356 Citations

Cancer Research

Autologous dendritic cells transfected with total renal tumor RNA have been shown to be potent stimulators of CTLs and antitumor immunity in vitro. A Phase I trial was conducted to evaluate this strategy for feasibility, safety, and efficacy to induce tumor-specific T-cell responses in subjects with metastatic renal cell carcinoma. Renal tumor RNA-transfected dendritic cells were administered to 10 evaluable study patients with no evidence of dose-limiting toxicity or vaccine-related adverse effects including autoimmunity. In six of seven evaluable subjects, expansion of tumor-specific T cells was detected after immunization. The vaccine-induced T-cell reactivities were directed against a broad set of renal tumor-associated antigens, including telomerase reverse transcriptase, G250, and oncofetal antigen, but not against self-antigens expressed by normal renal tissues. Although most patients underwent secondary therapies after vaccination, tumor-related mortality of the study subjects was unexpectedly low with only 3 of 10 patients dying from disease after a mean follow-up of 19.8 months. These data provide a scientific rationale for continued clinical investigation of this polyvalent vaccine strategy in the treatment of metastatic renal cell carcinoma and, potentially, other cancers.


Enhanced induction of telomerase-specific CD4(+) T cells using dendritic cells transfected with RNA encoding a chimeric gene product

October 2002

·

58 Reads

·

114 Citations

Cancer Research

Dendritic cells (DCs) transfected with mRNA encoding human telomerase reverse transcriptase (hTERT) have been shown to represent potent inducers of CTLs and antitumor immunity. However, it has become widely accepted that not only CTLs but also CD4(+) T helper cells are critical to the generation, as well as to the maintenance, of potent antitumor responses in vivo. In this study, we sought to determine whether human DCs transfected with mRNA encoding a chimeric hTERT/lysosome-associated membrane protein (LAMP-1) protein, carrying the endosomal/lysosomal sorting signal of the LAMP-1, are capable of stimulating concomitant hTERT-specific CD8(+) and CD4(+) T-cell responses in vitro. We show that processing of hTERT/LAMP-1 transcripts leads to enhanced stimulation of hTERT-specific CD4(+) T cells and does not negatively affect intracellular generation and subsequent presentation of MHC class I epitopes, hence, generating a CTL response. These findings provide a preclinical rationale of using DCs transfected with the chimeric hTERT/LAMP-1 RNA in vaccine trials to facilitate generation of antigen-specific CD4(+) T-cell responses that may be required to stimulate and maintain an optimal CD8(+) CTL response in vivo.


Figure 1 Incubation of immature DCs with PSA RNA provides a maturation stimulus. (a) Upregulation of CD83 expression by immature DCs in the presence of increasing PSA mRNA concentrations. (b–e) Immature DCs were incubated for 45 minutes in the presence of 20 µg/ml of PSA mRNA and analyzed after 48 hours by flow cytometry for expression of the DC-specific marker CD1a and the DC maturation marker CD83, respectively. (b) Staining with isotype immunoglobulins IgG 2b -FITC and IgG 1 -PE (isotype control). (c) DCs incubated in the absence of PSA mRNA (untreated). (d) DCs incubated with PSA mRNA that had been treated for 1 hour at 37°C with 10 µg/ml RNase A (PSA-RNA + RNase A), and (e) DCs incubated in the presence of PSA mRNA. One representative experiment out of three independent experiments is presented.  
Table 1 Characteristics of patients enrolled, including status before vaccination and response to vaccination
Figure 2 In vivo induction of PSA-specific T cell responses. PBMCs obtained at baseline (PSA, KK pre) and after three vaccination cycles (PSA, KK post) using PSA RNA-transfected DCs were cultured overnight with PSA or kallikrein protein (5 µg/ml). The specific T cell frequencies in each of the evaluable patients treated at a low dose level (10 7 cells; patient 2, 3, and 5), medium dose level (3 × 10 7 cells; patient 6, 7, and 9), or high dose level (5 × 10 7 cells; patient 11 and 13) is expressed as the number of spot-forming cells per 5 × 10 5 PBMCs seeded in each well.
Figure 4 PSA velocities after vaccination with PSA RNA-transfected DCs. Pretherapy (pre slope) and post-treatment (post slope) serum PSA kinetics of study candidates vaccinated with PSA RNA-transfected DCs were calculated as described previously (13). A linear regression model was used to obtain estimates on the change of serum PSA over time, and the differences between pre-and post-therapy log slope estimates were computed for each patient. (a-c) The results on the serum PSA kinetics in all seven patients who were eligible for analysis before (pre) and after (post) vaccine therapy. In one of seven patients available for analysis (a), a decrease in the log slope PSA was calculated upon initiation of therapy. In five of the seven evaluable patients we could demonstrate a significant reduction of the log slope PSA (patient 3, 5, 7, 9, 10) (b), whereas one patient (patient 2) exhibited unchanged (c) PSA velocities after treatment with PSA RNA-pulsed DCs. (d) Pre-(white bars) and post-therapy (black bars) log slope PSA values for the entire group. Asterisk indicates statistically significant differences between the pre-and post-treatment slope.
Autologous dendritic cells transfected with prostate-specific antigen RNA stimulate CTL responses against metastatic prostate tumors

March 2002

·

176 Reads

·

519 Citations

The Journal of clinical investigation

Autologous dendritic cells (DCs) transfected with mRNA encoding prostate-specific antigen (PSA) are able to stimulate potent, T cell-mediated antitumor immune responses in vitro. A phase I trial was performed to evaluate this strategy for safety, feasibility, and efficacy to induce T cell responses against the self-protein PSA in patients with metastatic prostate cancer. In 13 study subjects, escalating doses of PSA mRNA-transfected DCs were administered with no evidence of dose-limiting toxicity or adverse effects, including autoimmunity. Induction of PSA-specific T cell responses was consistently detected in all patients, suggesting in vivo bioactivity of the vaccine. Vaccination was further associated with a significant decrease in the log slope PSA in six of seven subjects; three patients that could be analyzed exhibited a transient molecular clearance of circulating tumor cells. The demonstration of vaccine safety, successful in vivo induction of PSA-specific immunity, and impact on surrogate clinical endpoints provides a scientific rationale for further clinical investigation of RNA-transfected DCs in the treatment of human cancer.


Fig. 1. Autologous DCs transfected with autologous tumor RNA are capable of recognizing renal tumor targets. PBMCs from a renal cancer patient (patient A) were stimulated twice with autologous DCs transfected with autologous renal tumor RNA (DC RCC RNA) and tested for the presence of antigen-specific T cells. DCs transfected with autologous tumor RNA (bottom left panel) or cultured primary tumor cells derived from the same patients (top right panel) were used as target cells. DCs transfected with RNA from autologous PBMCs or GFP served as control targets. Blocking studies (bottom left panel) were performed using CD8, HLA class II, and isotype control antibodies added during incubation of tumor-specific effector cells and RCC RNA-transfected DC targets (E:T ratio, 40:1). Using identical experimental conditions, tumor-specific T cells were reacted against their cognate targets (RCC RNA-transfected DCs), K562 cells, or naı¨venaı¨ve DCs (bottom right panel).  
Fig. 2. Autologous DCs transfected with autologous tumor RNA stimulate CTLs that recognize autologous tumor but not normal tissue antigens. PBMCs from a renal cancer patient were stimulated twice with autologous DCs transfected with various autologous RNA preparations and tested for the presence of antigen-specific CTLs. For stimulations, DCs were transfected with cellular RNA derived from the following autologous sources: RCC (A), RE (B), SM (C), and PBMCs (D). DCs transfected with autologous RCC RNA, RE RNA, SM RNA, PBMC RNA, or in vitro synthesized GFP RNA were used as targets. These results are representative of two experiments. Fig. 3. Autologous DCs transfected with autologous RCC RNA stimulate CTLs that recognize antigens shared with other RCC patients. DCs were generated from two renal cancer patients (patient B, left panel; patient C, right panel) and transfected with autologous tumor RNA (RCC RNA) to stimulate CTLs from PBMCs. After two rounds of stimulation, PBMCs were assayed for the presence of CTLs recognizing and lysing DC targets transfected with the following RNA preparations: autologous RCC (patient B), allogeneic RCC (patient C), and allogeneic RCC (patient A). DCs pulsed with RNA from autologous or allogeneic RE or with RNA from autologous or allogeneic PBMCs (data not shown) derived from patient C or patient A served as control target cells.  
Fig. 5. Autologous DCs transfected with RNA prepared from the primary tumor provide antimetastatic activity. PBMCs from a renal cancer patient were stimulated twice with autologous DCs transfected with cellular RNA extracted from the following autologous sources: primary RCC (RCC RNA, A); LN metastasis (LN RNA, B); and BM (BM RNA, C). The RNA-transfected DCs were used to stimulate CTLs and as targets. Additional targets included DCs transfected with autologous PBMC RNA, TERT RNA, and GFP RNA.  
Fig. 4. Autologous DCs transfected with RNA from autologous primary tumor stimulate CTLs that recognize tumor and TERT antigens. Renal tumor or TERT-specific CTLs were generated by stimulating PBMCs with autologous DCs transfected with autologous renal tumor RNA (left panel) or TERT RNA (right panel). On DC stimulation, PBMCs were tested for the presence of CTLs recognizing and lysing DCs transfected with autologous RCC RNA or transfected with TERT RNA. DCs transfected with either PBMC RNA or GFP RNA were used as control targets.  
Human dendritic cells transfected with renal tumor RNA stimulate polyclonal T-cell response against antigens expressed by primary and metastatic tumors.

May 2001

·

95 Reads

·

155 Citations

Cancer Research

Although renal cell carcinoma has been shown to respond to immunotherapy, renal cell carcinoma-specific rejection antigens and their corresponding CTL epitopes have rarely been described. The use of dendritic cells (DCs) transfected with mRNA isolated from tumor cells may allow specific immunotherapy even in cancers for which potent rejection antigens have not been identified. Here we show that DCs transfected with RNA isolated from renal cancer tissue are remarkably effective in stimulating tumor-specific T-cell response in vitro but do not cross-react with normal tissue antigens including antigens expressed by renal parenchyma. In contrast, the tumor-specific CTLs lysed allogeneic tumor but not allogeneic normal tissue targets, suggesting the presence of shared albeit unidentified antigens among renal carcinomas. CTL responses against telomerase reverse transcriptase (TERT) accounted in part for the reactivities against allogeneic tumors because renal tumor RNA-transfected DCs stimulated polyclonal CTL responses, which encompassed as a subcomponent a response against TERT. Nonetheless, the tumor-specific CTLs were consistently superior to the CTLs stimulated with TERT RNA-transfected DCs in recognizing and lysing tumor targets, suggesting that tumor-specific CTLs represent a polyclonal response providing more effective antitumor activity than T-cell responses directed against a single antigen in the form of TERT. Tumor RNA-transfected DCs were capable of stimulating T-cell reactivities not only against the primary tumor but also against metastatic tumors, although discrete differences in the antigenic repertoire expressed by these tissues were apparent. Thus, total tumor RNA-transfected DCs may represent a broadly applicable vaccine strategy to induce polyclonal and potentially therapeutic T-cell responses in renal cancer patients.


Induction of Polyclonal Prostate Cancer-Specific CTL Using Dendritic Cells Transfected with Amplified Tumor RNA

April 2001

·

81 Reads

·

190 Citations

The Journal of Immunology

Polyvalent cancer vaccines targeting the entire antigenic spectrum on tumor cells may represent a superior therapeutic strategy for cancer patients than vaccines solely directed against single Ags. In this study, we show that autologous dendritic cells (DC) transfected with RNA amplified from microdissected tumor cells are capable of stimulating CTL against a broad set of unidentified and critical prostate-specific Ags. Although the polyclonal CTL responses generated with amplified tumor RNA-transfected DC encompassed as a subcomponent a response against prostate-specific Ag (PSA) as well as against telomerase reverse transcriptase, the tumor-specific CTL were consistently more effective than PSA or telomerase reverse transcriptase CTL to lyse tumor targets, suggesting the superiority of the polyclonal response. Although tumor RNA-transfected DC stimulated CTL, which recognized not only tumor but also self-Ags expressed by benign prostate tissue, these cross-reactive CTL were exclusively specific for the PSA, indicating an immunodominant role of PSA in the prostate cancer-specific immune response. Our data suggest that tumor RNA-transfected DC may represent a broadly applicable, potentially clinically effective vaccine strategy for prostate cancer patients, which is not limited by tumor tissue availability for Ag preparation and may minimize the risk of clonal tumor escape.


Human Dendritic Cells Transfected with RNA Encoding Prostate-Specific Antigen Stimulate Prostate-Specific CTL Responses In Vitro

June 2000

·

135 Reads

·

195 Citations

The Journal of Immunology

Although immunological tolerance to self Ags represents an important mechanism to prevent normal tissue injury, there is growing evidence that tolerance to tumor Ags, which often represent normal peripherally expressed proteins, is not absolute and can be effectively reverted. Prostate-specific Ag (PSA) is a self Ag expressed by both normal and malignant prostatic epithelium, and therefore offers a unique opportunity to examine the ability of self Ags to serve as specific CTL targets. In this study, we investigated the efficacy of autologous dendritic cells (DC) transfected with mRNA encoding PSA to stimulate CTL against PSA Ags in vitro. Ag in form of RNA carries the advantage to encode multiple epitopes for many HLA alleles, thus permitting induction of CTL responses among many cancer patients independent of their HLA repertoire. In this study, we show that PSA mRNA-transfected DC were capable of stimulating primary CTL responses against PSA Ags in vitro. The PSA-specific CTL did not cross-react with kallikrein Ags, a protein, which shares significant homology with PSA, suggesting that harmful autoimmune toxicity may not represent a significant problem with this approach. PSA RNA-transfected DC generated from male or female healthy volunteers or from cancer patients were equally effective in stimulating PSA-specific CTL in vitro, implying that neither natural tolerance to PSA Ags nor tumor-mediated T cell anergy may represent major barriers for CTL generation against the self Ag PSA. This study provides a preclinical rationale for using PSA RNA-transfected DC in active or adoptive immunization protocols.


Citations (10)


... Tregs were defined as CD4 + CD25 + FoxP3 + cells, while MDSCs were defined as CD11b + CD14 + CD33 + cells, according to previous studies [24][25][26]. The following immune cell subsets were analyzed using flow cytometry and the listed Abs. ...

Reference:

Baseline interleukin-6 is a prognostic factor for patients with metastatic breast cancer treated with eribulin
Reversal of Myeloid Cell - Mediated Immunosuppression in Patients with Metastatic Renal Cell Carcinoma

Clinical Cancer Research

... In some studies for dissociation of bladder tissue 1:1 collagenase II and dispase enzymes are used at 37°C for 12 hours [40][41][42][43]. Also there is a need to monitor tissue digestion process for every 2 hours by gently shaking the digestion mixture by checking the viability of cells under the microscope [50][51][52][53]. ...

Isolation and characterization of a novel human bladder cancer cell line: BK10
  • Citing Article
  • January 1998

In Vitro Cellular & Developmental Biology - Animal

... PSA, a serin-protease similar to kallikrein, is usually displayed in prostate epithelial cells [72][73][74] . Studies have identified PSA-derived peptides that activate tumor-reactive CD8+ cytotoxic T lymphocytes (CTLs) when presented by human leukocyte antigen-A2 (HLA-A2) and HLA-A3 [75][76][77] . Simultaneous induction of tumor-reactive CTLs and HLA-A2/A3-restricted epitopes of PSA has been achieved using specific oligopeptides [78][79][80] . ...

Human Dendritic Cells Transfected with RNA Encoding Prostate-Specific Antigen Stimulate Prostate-Specific CTL Responses In Vitro

The Journal of Immunology

... The company developing this vaccine opted to pursue a similar approach using autologous DCs as a vaccine but loaded ex vivo with autologous tumour lysates rather than peptides based on results from a non-randomized clinical trial in which prolonged OS (18.3 months) had been observed in patients with glioblastoma 70 . In another approach, DCs isolated from patients with prostate cancer and transfected with mRNA encoding PSA were shown to stimulate a potent antigen-specific cytotoxic T cell response 71 . Similarly, in a small study including 13 patients with metastatic prostate cancer, autologous DCs were loaded with mRNA encoding PSA 72 , with the results showing a decreased PSA slope and increased PSA-specific T cell responses in all patients; unfortunately, this approach has not advanced further, for unclear reasons. ...

Induction of Polyclonal Prostate Cancer-Specific CTL Using Dendritic Cells Transfected with Amplified Tumor RNA

The Journal of Immunology

... Heiser et al. isolated RNA from renal cancer tissue and transfected DCs with tumor RNA. The authors demonstrated that DCs transfected with tumor RNA also stimulated a tumor-specific CTL response in patients with RCC [134]. Indeed, DCs transfected with tumor RNA were more effective than DCs transfected with TAA-RNA [131]. ...

Human dendritic cells transfected with renal tumor RNA stimulate polyclonal T-cell response against antigens expressed by primary and metastatic tumors.

Cancer Research

... first reported that naked-mRNA-encoded luciferase, chloramphenicol acetyltransferase, or beta-galactosidase was separately injected into skeletal muscle and then immediately produced protein in mice. Since then, synthetic in vitro mRNA has been widely utilized in vaccinations for treating cancer and infectious diseases [20][21][22][23][24][25][26][27] . However, mRNA strategy has not been considered as a therapeutic approach for other diseases due to mRNA instability and immunogenicity; thus, the application of synthetic mRNA for gene replacement therapy has been delayed. ...

Autologous dendritic cells transfected with prostate-specific antigen RNA stimulate CTL responses against metastatic prostate tumors

The Journal of clinical investigation

... Since the peptides from loaded mRNAs must be coupled with the MHC compound to mount the anti-tumoral immune responses, the chimeric lysosome-associated membrane protein-1 (LAMP-1) with the tumoral antigen can facilitate this process [68]. Similarly, Su et al. have shown that the mRNA of tumoral antigen/LAMP-1 can enhance the peptide/MHC-I presentation and the anti-tumoral immune responses in affected patients [69]. However, Kyte et al. [70] vaccinated melanoma patients with autologous tumor-mRNA loaded DCs in a phase I/II trial. ...

Enhanced induction of telomerase-specific CD4(+) T cells using dendritic cells transfected with RNA encoding a chimeric gene product
  • Citing Article
  • October 2002

Cancer Research

... Targeting telomerase activity alongside ionising radiation predominantly involves biological agents, including interfering RNA, such as nucleoside analogues (6-thio-dG, AZT), non-nucleoside inhibitors (BIBR1532), and antisense oligonucleotide inhibitors GRN163L [14][15][16]. Furthermore, immunotherapy with hTERT polypeptide GV1001, secreted polypeptide Vx-001, and dendritic cells GRVAC1 has entered clinical trials, demonstrating promising therapeutic outcomes in phase I and phase II clinical trials [17][18][19]. ...

Telomerase mRNA-Transfected Dendritic Cells Stimulate Antigen-Specific CD8+ and CD4+ T Cell Responses in Patients with Metastatic Prostate Cancer

The Journal of Immunology

... Because the majority of Tregs express high levels of CD25, it has been hypothesized that the greatest immunologic impact of denileukin diftitox may be on depleting Treg cells. A previous study of single dose denileukin diftitox followed by vaccination with tumor RNA-transfected dendritic cells noted a decrease in circulating CD4 + CD25 + T cells with a greater magnitude of the specific immune response to the vaccine relative to no denileukin diftitox treatment [10] . In contrast , denileukin diftitox administered on a 5 day schedule failed to deplete CD4 + CD25 + T cells from the blood of subjects with melanoma, [11] suggesting that a difference in dose or schedule could be important. ...

Dannull J, Su Z, Rizzieri D, Yang BK, Coleman D, Yancey D, Zhang A, Dahm P, Chao N, Gilboa E, Vieweg JEnhancement of vaccine-mediated antitumor immunity in cancer patients after depletion of regulatory T cells. J Clin Invest 115: 3623-3633

The Journal of clinical investigation

... Referring to DNA and mRNA vaccines, the delivery of hTERT-encoded DNA plasmids with or without interleukin-12 plasmid via intramuscular electroporation could display favorable safety and efficacy in patients with pancreatic cancer, in association with positive immune responses represented by the increased production of hTERT-specific IFN-γ and activation of hTERT-specific T cells. 86 Analogously, intradermal injection of naked mRNA coding for tumorassociated antigens such as telomerase, with GM-CSF as adjuvant, could perform well in renal cell cancer patients, with median survival of 24-26 months. 89 As to DC vaccines, several clinical trials have demonstrated that the vaccinations with transfected telomerase RNA or the telomerase peptide pulsed DC could be well tolerated and exert beneficial clinical effects in patients with renal cancer, 89 ...

Immunological and clinical responses in metastatic renal cancer patients vaccinated with tumor RNA-transfected dendritic cells

Cancer Research