Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Zhiyong Qiu is active.

Publication


Featured researches published by Zhiyong Qiu.


Proceedings of the National Academy of Sciences of the United States of America | 2008

Antigen kinetics determines immune reactivity

Pål Johansen; Tazio Storni; Lorna Rettig; Zhiyong Qiu; Ani Der-Sarkissian; Kent Andrew Smith; Vania Manolova; Karl S. Lang; Beat Müllhaupt; Tilman Gerlach; Roberto F. Speck; Adrian Bot; Thomas M. Kündig

A current paradigm in immunology is that the strength of T cell responses is governed by antigen dose, localization, and costimulatory signals. This study investigates the influence of antigen kinetics on CD8 T cell responses in mice. A fixed cumulative antigen dose was administered by different schedules to produce distinct dose-kinetics. Antigenic stimulation increasing exponentially over days was a stronger stimulus for CD8 T cells and antiviral immunity than a single dose or multiple dosing with daily equal doses. The same was observed for dendritic cell vaccination, with regard to T cell and anti-tumor responses, and for T cells stimulated in vitro. In conclusion, stimulation kinetics per se was shown to be a separate parameter of immunogenicity. These findings warrant a revision of current immunization models and have implications for vaccine development and immunotherapy.


Journal of Immunotherapy | 2008

Phase 1 Trial of Intranodal Injection of a Melan-A/MART-1 DNA Plasmid Vaccine in Patients With Stage IV Melanoma

Jeffrey S. Weber; William D. Boswell; John C. Smith; Evan M. Hersh; Jolie Snively; Mella Diaz; Sabrina Miles; Xiding Liu; Mihail Obrocea; Zhiyong Qiu; Adrian Bot

Nineteen patients with stage IV melanoma were treated in an escalating dose, phase 1 trial of a DNA plasmid vaccine pSEM. The plasmid encoded T-cell epitopes from differentiation antigens Melan-A/melanoma antigen recognized by T cells (MART)-1 and tyrosinase, encompassing amino acids 26-35 and 31-70 from Melan-A/MART-1, and 1-9 as well as 369-377 from tyrosinase. End points of the trial were safety, tolerability, and melanoma antigen-specific immunity by tetramer assay. Intralymph nodal infusions of the vaccine were given 4 times, every 2 weeks over 96 hours each to groin lymph nodes. Vaccine doses were 500, 1000, and 1500 μg of DNA per infusion. Disease evaluation was performed 8 weeks after treatment initiation. The vaccine was well tolerated, with only grade I/II toxicity observed and no dose limiting toxicity at the highest dose of 1500 μg per infusion. Immune response defined prospectively was seen in 4/19 patients, and 5/19 had evidence of preexisting immunity to Melan-A/MART-1. No immune responses to tyrosinase was seen. There was a correlation between time to progression (TTP) and Melan-A/MART-1 immunity (preexisting or induced) for all patients. There was no association between TTP and immune competence assayed by ex vivo polyclonal stimulation of peripheral blood mononuclear cells. No clinical responses were seen. DNA plasmid pSEM vaccine was well tolerated when administered intranodally by 96-hour infusion to patients with stage IV melanoma, and was immunogenic, but did not induce regression of established disease. The association of TTP with preexisting or induced Melan-A immunity supports future attempts to induce potent immunity to this antigen.


Clinical Cancer Research | 2011

Intra-Lymph Node Prime-Boost Vaccination Against Melan A and Tyrosinase for the Treatment of Metastatic Melanoma: Results of a Phase 1 Clinical Trial

Antoni Ribas; Jeffrey S. Weber; Bartosz Chmielowski; Begonya Comin-Anduix; David Lu; Michael Douek; Nagesh Ragavendra; Steve Raman; Elizabeth Seja; Darlene Rosario; Sabrina Miles; David C. Diamond; Zhiyong Qiu; Mihail Obrocea; Adrian Bot

Purpose: The goal of this study was to test the safety and activity of a therapeutic vaccine, MKC1106-MT, in patients with metastatic melanoma. Experimental Design: MKC1106-MT comprises a plasmid (pMEL-TYR) and two peptides (E-MEL and E-TYR), corresponding to Melan A and tyrosinase, administered by intra–lymph node injection in a prime-boost sequence. All 18 patients were HLA-A*0201 positive and received a fixed priming dose of plasmid and a low or a high peptide dose. Enumeration of antigen-specific T cells was done prior to and throughout the treatment. Patients who did not exhibit disease progression remained on study and could receive up to eight cycles of treatment. Results: The MKC1106-MT regimen was well tolerated and resulted in an overall immune response rate of 50%. The treatment showed disease control, defined as stable disease that lasted for 8 weeks or more in 6 of 18 (33%) of the patients: 14% and 46% in the low and high peptide dose, respectively. Interestingly, four patients, all with tumor burden largely confined to lymph nodes and Melan A–specific T cells at baseline, showed durable disease control associated with radiologic evidence of tumor regression. There was no noticeable correlation between the expansion of antigen-specific T cells in blood and the clinical outcome; yet, there was evidence of active tumor-infiltrating lymphocytes (TIL) in two regressing lesions. Conclusions: MKC1106-MT showed immunogenicity and evidence of disease control in a defined patient population. These findings support further development of this investigational agent and the concept of therapeutic vaccination in metastatic melanoma. Clin Cancer Res; 17(9); 2987–96. ©2011 AACR.


Journal of Immunotherapy | 2011

A phase 1 study of a vaccine targeting preferentially expressed antigen in melanoma and prostate-specific membrane antigen in patients with advanced solid tumors.

Jeffrey S. Weber; Nicholas J. Vogelzang; Marc S. Ernstoff; Oscar B. Goodman; Lee D. Cranmer; John L. Marshall; Sabrina Miles; Dar Rosario; David C. Diamond; Zhiyong Qiu; Mihail Obrocea; Adrian Bot

Preferentially expressed antigen in melanoma (PRAME) and prostate-specific membrane antigen (PSMA) are tumor-associated antigens implicated in cellular differentiation, genetic stability, and angiogenesis. MKC1106-PP is an immunotherapeutic regimen cotargeting PRAME and PSMA, comprised of a recombinant plasmid (pPRA-PSM encoding fragments derived from both antigens) and 2 peptides (E-PRA and E-PSM derived from PRAME and PSMA, respectively). This multicenter study evaluated MKC1106-PP with a fixed plasmid dose and 2 different peptide doses, administered by intralymph node injection in a prime-boost sequence in human leukocyte antigen-A*0201 and tumor-antigen-positive patients with progressing metastatic solid tumors who had failed standard therapy. Immune monitoring was done by tetramer and enzymatic-linked immune spot analysis. The treatment was well tolerated, with no significant differences in safety, immune response, and clinical outcome relative to peptide doses. Fifteen of 24 evaluable patients showed an immune response, as defined by the expansion of PRAME-specific or PSMA-specific T cells in the blood. There were no partial or complete responses by the Response Evaluation Criteria in Solid Tumors. Seven patients showed stable disease (SD) for 6 months or longer, or prostate specific antigen decline: 4 of 10 with prostate carcinoma, 2 of 2 with renal clear cell carcinoma, and 1 of 10 with metastatic melanoma. In addition, there was an association between the induction and persistence of antigen-specific T cells in blood above baseline levels and disease control, defined as SD for 6 months or longer. These results support further development of MKC1106-PP in specific clinical indications.


Vaccine | 2009

Enhancing DNA vaccination by sequential injection of lymph nodes with plasmid vectors and peptides.

Kent Andrew Smith; Victor Tam; Raymond Wong; Robb R. Pagarigan; Brenna L. Meisenburg; Diljeet K. Joea; Xiping Liu; Christiana Sanders; David C. Diamond; Thomas M. Kündig; Zhiyong Qiu; Adrian Bot

DNA vaccines or peptides are capable of inducing specific immunity; however, their translation to the clinic has generally been problematic, primarily due to the reduced magnitude of immune response and poor pharmacokinetics. Herein, we demonstrate that a novel immunization strategy, encompassing sequential exposure of the lymph node milieu to plasmid and peptide in a heterologous prime-boost fashion, results in considerable MHC class I-restricted immunity in mice. Plasmid-primed antigen expression was essential for the generation of a population of central memory T cells, expressing CD62L and low in PD-1, with substantial capability to expand and differentiate to peripheral memory and effector cells, following subsequent exposure to peptide. These vaccine-induced T cells dominated the T cell repertoire, were able to produce large amounts of chemokines and pro-inflammatory cytokines, and recognized tumor cells effectively. In addition to outlining a feasible and effective method to transform plasmid DNA vaccination into a potentially viable immunotherapeutic approach for cancer, this study sheds light on the mechanism of heterologous prime-boost and the considerable heterogeneity of MHC class I-restricted T cell responses.


Clinical Cancer Research | 2009

Lymph Node–Targeted Immunotherapy Mediates Potent Immunity Resulting in Regression of Isolated or Metastatic Human Papillomavirus–Transformed Tumors

Kent Andrew Smith; Brenna L. Meisenburg; Victor Tam; Robb R. Pagarigan; Raymond Sm Wong; Diljeet K. Joea; Liz Lantzy; Mayra A. Carrillo; Todd M. Gross; Uriel M. Malyankar; Chih-Sheng Chiang; Diane M. Da Silva; Thomas M. Kündig; W. Martin Kast; Zhiyong Qiu; Adrian Bot

Purpose: The goal of this study was to investigate the therapeutic potential of a novel immunotherapy strategy resulting in immunity to localized or metastatic human papillomavirus 16–transformed murine tumors. Experimental Design: Animals bearing E7-expressing tumors were coimmunized by lymph node injection with E7 49-57 antigen and TLR3-ligand (synthetic dsRNA). Immune responses were measured by flow cytometry and antitumor efficacy was evaluated by tumor size and survival. In situ cytotoxicity assays and identification of tumor-infiltrating lymphocytes and T regulatory cells were used to assess the mechanisms of treatment resistance in bulky disease. Chemotherapy with cyclophosphamide was explored to augment immunotherapy in late-stage disease. Results: In therapeutic and prophylactic settings, immunization resulted in a considerable expansion of E7 49-57 antigen–specific T lymphocytes in the range of 1/10 CD8+ T cells. The resulting immunity was effective in suppressing disease progression and mortality in a pulmonary metastatic disease model. Therapeutic immunization resulted in control of isolated tumors up to a certain volume, and correlated with antitumor immune responses measured in blood. In situ analysis showed that within bulky tumors, T-cell function was affected by negative regulatory mechanisms linked to an increase in T regulatory cells and could be overcome by cyclophosphamide treatment in conjunction with immunization. Conclusions: This study highlights a novel cancer immunotherapy platform with potential for translatability to the clinic and suggests its potential usefulness for controlling metastatic disease, solid tumors of limited size, or larger tumors when combined with cytotoxic agents that reduce the number of tumor-infiltrating T regulatory cells. (Clin Cancer Res 2009;15(19):6167–76)


Immunology Letters | 2009

TLR-9 signaling and TCR stimulation co-regulate CD8+ T cell-associated PD-1 expression

Raymond Wong; Kent Andrew Smith; Victor Tam; Robb R. Pagarigan; Brenna L. Meisenburg; Angeline M. Quach; Mayra A. Carrillo; Zhiyong Qiu; Adrian Bot

Elevated Programmed Death-1 (PD-1) expression can inhibit T cell activity and is a potential barrier to achieving persisting and optimal immunity via therapeutic vaccination. Using a direct lymph node-targeted vaccination procedure that enabled uncoupling of synthetic peptide (signal 1, TCR-mediated) and adjuvant (signal 2, non-TCR-mediated), we evaluated the impact of varied doses of Toll-like receptor (TLR)-9 ligand CpG oligodeoxynucleotide (ODN) adjuvant on epitope-specific CD8(+) T cell-associated PD-1 expression. Peptide vaccination without adjuvant yielded CD8(+) T cells with significantly elevated PD-1 expression. This conferred impaired function ex vivo, but was reversible by antibody-mediated PD-1 blockade. By comparison, peptide vaccination with escalating doses of CpG ODN adjuvant yielded higher magnitudes of CD8(+) T cells with progressively lower PD-1 expression and greater ex vivo function. CpG ODN adjuvant in context of titrated peptide doses for vaccination yielded the lowest overall PD-1 expression levels, demonstrating that fine-tuning both TCR-independent (adjuvant dose) and -dependent (antigen dose) stimuli can synergize to co-regulate PD-1 expression on epitope-specific CD8(+) T cells. These data hint at strategies to elicit PD-1(low) CD8(+) T cells using TLR-9 ligand adjuvants, and also shed light on the PD-1-regulated homeostasis of CD8(+) T cells.


Journal of Translational Medicine | 2010

Programmed cell death-1 (PD-1) at the heart of heterologous prime-boost vaccines and regulation of CD8 + T cell immunity

Adrian Bot; Zhiyong Qiu; Raymond Wong; Mihail Obrocea; Kent Andrew Smith

Developing new vaccination strategies and optimizing current vaccines through heterologous prime-boost carries the promise of integrating the benefits of different yet synergistic vectors. It has been widely thought that the increased immunity afforded by heterologous prime-boost vaccination is mainly due to the minimization of immune responses to the carrier vectors, which allows a progressive build up of immunity against defined epitopes and the subsequent induction of broader immune responses against pathogens. Focusing on CD8+ T cells, we put forward a different yet complementary hypothesis based primarily on the systematic analysis of DNA vaccines as priming agents. This hypothesis relies on the finding that during the initiation of immune response, acquisition of co-inhibitory receptors such as programmed cell death-1 (PD-1) is determined by the pattern of antigen exposure in conjunction with Toll-like receptor (TLR)-dependent stimulation, critically affecting the magnitude and profile of secondary immunity. This hypothesis, based upon the acquisition and co-regulation of pivotal inhibitory receptors by CD8+ T cells, offers a rationale for gene-based immunization as an effective priming strategy and, in addition, outlines a new dimension to immune homeostasis during immune reaction to pathogens. Finally, this model implies that new and optimized immunization approaches for cancer and certain viral infections must induce highly efficacious T cells, refractory to a broad range of immune-inhibiting mechanisms, rather than solely or primarily focusing on the generation of large pools of vaccine-specific lymphocytes.


Cancer Research | 2010

Abstract 2398: Intra-lymph node DNA vaccination as a platform for safe and effective immunotherapy of cancer

Zhiyong Qiu; Kent Andrew Smith; Raymond Wong; Victor Tam; Begonya Comin-Anduix; Anthony Ribas; Darlene Rosario; Mihail Obrocea; Adrian Bot

The purpose of this study was to advance a novel immunotherapy platform technology that would achieve objective clinical response and allow subsequent elucidation of immune correlates of clinical responses. To this aim, we tested an immunotherapy approach encompassing sequential intra-lymph node administration of DNA vectors and peptide analogues to elicit multivalent CD8+ T cell immunity against cancer cell and stromal antigens. This included the assessment of safety, immune response and clinical outcome following treatment with two investigational regimens in patients with metastatic solid tumors such as melanoma, prostate, and renal clear cell carcinoma. Strikingly, T cell transcriptomal analysis by gene array and flow cytometry analysis in a preclinical model showed that DNA vaccination induced a distinct immunophenotype, compared to peptide vaccination. DNA vaccine-primed CD8+ T cells showed a coordinated, lower expression of co-inhibitory molecule PD-1 and retention of the lymph node homing molecule CD62L, leading to a significant persistence of antigen-specific T cells, cellular expansion and functional conversion to effector cells, upon peptide boosting. Secondly, both objective tumor regression and elevation of antigen-specific T cells were observed in immunized patients, although there was no strict correlation between these two outcomes. Instead, there was a disease stage dependency of the presence of T cells specific for Melan A / MART-1 in melanoma patients, with all stage IV M1a (lymphatic metastatic disease) patients exhibiting pre-existing T cells and responding to treatment through durable tumor regression. Significantly, T cells specific for both immunizing antigens, Melan A/MART-1 and Tyrosinase, were identifiable within regressing tumor lesions in these M1a patients. In contrast, the preponderance of pre-existing Melan A/MART-1 T cells in melanoma patients with visceral metastatic disease was less than 50% and no apparent tumor regression upon administration was observed. In a second parallel trial, co-immunization against PRAME and PSMA generated clinical response through disease stabilization in several patients with carcinoma and was mirrored by a persisting T cell immunity. In summary, these results lead to three major conclusions: first, design of synthetic active immunotherapies affording objective tumor regression is possible, thereby refuting the paradigm that such approaches can be effective only in earlier disease stage; second, a systemic, hypothesis-generating approach must be employed to uncover appropriate biomarker correlates; and third, DNA-based vaccination has the potential of being translated to safe and effective therapies for cancer. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 101st Annual Meeting of the American Association for Cancer Research; 2010 Apr 17-21; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2010;70(8 Suppl):Abstract nr 2398.


Archive | 2004

Expression vectors encoding epitopes of target-associated antigens and methods for their design

John J. L. Simard; David C. Diamond; Zhiyong Qiu; Xiang-Dong Lei

Collaboration


Dive into the Zhiyong Qiu's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Kent J. Smith

Medical University of South Carolina

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Anthony Ribas

University of California

View shared research outputs
Top Co-Authors

Avatar

Antoni Ribas

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

David Lu

University of California

View shared research outputs
Top Co-Authors

Avatar

Diane M. Da Silva

University of Southern California

View shared research outputs
Top Co-Authors

Avatar

Elizabeth Seja

University of California

View shared research outputs
Researchain Logo
Decentralizing Knowledge