1
|
Peptide Modification Diminishes HLA Class II-restricted CD4 + T Cell Recognition of Prostate Cancer Cells. Int J Mol Sci 2022; 23:ijms232315234. [PMID: 36499557 PMCID: PMC9738740 DOI: 10.3390/ijms232315234] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Revised: 11/27/2022] [Accepted: 11/28/2022] [Indexed: 12/08/2022] Open
Abstract
Prostate cancer poses an ongoing problem in the western world accounting for significant morbidity and mortality in the male population. Current therapy options are effective in treating most prostate cancer patients, but a significant number of patients progress beyond a manageable disease. For these patients, immunotherapy has emerged as a real option in the treatment of the late-stage metastatic disease. Unfortunately, even the most successful immunotherapy strategies have only led to a four-month increase in survival. One issue responsible for the shortcomings in cancer immunotherapy is the inability to stimulate helper CD4+ T cells via the HLA class II pathway to generate a potent antitumor response. Obstacles to proper HLA class II stimulation in prostate cancer vaccine design include the lack of detectable class II proteins in prostate tumors and the absence of defined class II specific prostate tumor antigens. Here, for the first time, we show that the insertion of a lysosomal thiol reductase (GILT) into prostate cancer cells directly enhances HLA class II antigen processing and results in increased CD4+ T cell activation by prostate cancer cells. We also show that GILT insertion does not alter the expression of prostate-specific membrane antigen (PSMA), an important target in prostate cancer vaccine strategies. Our study suggests that GILT expression enhances the presentation of the immunodominant PSMA459 epitope via the HLA class II pathway. Biochemical analysis showed that the PSMA459 peptide was cysteinylated under a normal physiologic concentration of cystine, and this cysteinylated form of PSMA459 inhibited T cell activation. Taken together, these results suggest that GILT has the potential to increase HLA class II Ag presentation and CD4+ T cell recognition of prostate cancer cells, and GILT-expressing prostate cancer cells could be used in designing cell therapy and/or vaccines against prostate cancer.
Collapse
|
2
|
Qin L, Wang S, Dominguez D, Long A, Chen S, Fan J, Ahn J, Skakuj K, Huang Z, Lee A, Mirkin C, Zhang B. Development of Spherical Nucleic Acids for Prostate Cancer Immunotherapy. Front Immunol 2020; 11:1333. [PMID: 32733447 PMCID: PMC7362897 DOI: 10.3389/fimmu.2020.01333] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Accepted: 05/26/2020] [Indexed: 01/02/2023] Open
Abstract
Although the strategy of therapeutic vaccination for the treatment of prostate cancer has advanced to and is available in the clinic (Sipuleucel-T), the efficacy of such therapy remains limited. Here, we develop Immunostimulatory Spherical Nucleic Acid (IS-SNA) nanostructures comprised of CpG oligonucleotides as adjuvant and prostate cancer peptide antigens, and evaluate their antitumor efficacy in syngeneic mouse models of prostate cancer. IS-SNAs with the specific structural feature of presenting both antigen and adjuvant CpG on the surface (hybridized model (HM) SNAs) induce stronger cytotoxic T lymphocyte (CTL) mediated antigen-specific killing of target cells than that for IS-SNAs with CpG on the surface and antigen encapsulated within the core (encapsulated model (EM) SNAs). Mechanistically, HM SNAs increase the co-delivery of CpG and antigen to dendritic cells over that for EM SNAs or admixtures of linear CpG and peptide, thereby improving cross-priming of antitumor CD8+ T cells. As a result, vaccination with HM SNAs leads to more effective antitumor immune responses in two prostate cancer models. These data demonstrate the importance of the structural positioning of peptide antigens together with adjuvants within IS-SNAs to the efficacy of IS-SNA-based cancer immunotherapy.
Collapse
Affiliation(s)
- Lei Qin
- Division of Hematology/Oncology, Department of Medicine, Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Shuya Wang
- Interdisciplinary Biological Sciences Graduate Program, Northwestern University, Evanston, IL, United States
| | - Donye Dominguez
- Division of Hematology/Oncology, Department of Medicine, Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Alan Long
- Division of Hematology/Oncology, Department of Medicine, Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Siqi Chen
- Division of Hematology/Oncology, Department of Medicine, Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Jie Fan
- Division of Hematology/Oncology, Department of Medicine, Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Jihae Ahn
- Division of Hematology/Oncology, Department of Medicine, Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Kacper Skakuj
- Department of Chemistry, Northwestern University, Evanston, IL, United States
| | - Ziyin Huang
- Department of Materials Science and Engineering, Northwestern University, Evanston, IL, United States
| | - Andrew Lee
- Department of Chemical and Biological Engineering, Northwestern University, Evanston, IL, United States
| | - Chad Mirkin
- Department of Chemistry, Northwestern University, Evanston, IL, United States.,The International Institute for Nanotechnology, Northwestern University, Evanston, IL, United States
| | - Bin Zhang
- Division of Hematology/Oncology, Department of Medicine, Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| |
Collapse
|
3
|
Ji D, Yi H, Zhang D, Zhan T, Li Z, Li M, Jia J, Qiao M, Xia J, Zhai Z, Song C, Gu J. Somatic Mutations and Immune Alternation in Rectal Cancer Following Neoadjuvant Chemoradiotherapy. Cancer Immunol Res 2018; 6:1401-1416. [PMID: 30282671 DOI: 10.1158/2326-6066.cir-17-0630] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Revised: 04/04/2018] [Accepted: 09/28/2018] [Indexed: 11/16/2022]
Affiliation(s)
- Dengbo Ji
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Surgery III, Peking University Cancer Hospital and Institute, Haidian District, Beijing, China
| | - Haizhao Yi
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Surgery III, Peking University Cancer Hospital and Institute, Haidian District, Beijing, China
- Department of General Surgery 1, Affiliated Hospital of Chengde Medical College, Chengde, China
| | - Dakui Zhang
- Department of General Surgery, China-Japan Friendship Hospital, Beijing, China
| | - Tiancheng Zhan
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Surgery III, Peking University Cancer Hospital and Institute, Haidian District, Beijing, China
| | - Zhaowei Li
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Surgery III, Peking University Cancer Hospital and Institute, Haidian District, Beijing, China
| | - Ming Li
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Surgery III, Peking University Cancer Hospital and Institute, Haidian District, Beijing, China
| | - Jinying Jia
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Surgery III, Peking University Cancer Hospital and Institute, Haidian District, Beijing, China
| | - Meng Qiao
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Surgery III, Peking University Cancer Hospital and Institute, Haidian District, Beijing, China
| | - Jinhong Xia
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Surgery III, Peking University Cancer Hospital and Institute, Haidian District, Beijing, China
| | - Zhiwei Zhai
- Department of Gastrointestinal Surgery, Chaoyang Hospital, Beijing, China
| | - Can Song
- School of Life Sciences, Tsinghua University, Beijing, China
- Peking-Tsinghua Center for Life Sciences, Beijing, China
| | - Jin Gu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Surgery III, Peking University Cancer Hospital and Institute, Haidian District, Beijing, China.
- Peking-Tsinghua Center for Life Sciences, Beijing, China
- Peking University S.G. Hospital, Beijing, China
| |
Collapse
|
4
|
Anti-tumor effect of the alphavirus-based virus-like particle vector expressing prostate-specific antigen in a HLA-DR transgenic mouse model of prostate cancer. Vaccine 2015; 33:5386-5395. [PMID: 26319744 DOI: 10.1016/j.vaccine.2015.08.062] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2015] [Revised: 06/23/2015] [Accepted: 08/18/2015] [Indexed: 12/21/2022]
Abstract
The goal of this study was to determine if an alphavirus-based vaccine encoding human Prostate-Specific Antigen (PSA) could generate an effective anti-tumor immune response in a stringent mouse model of prostate cancer. DR2bxPSA F1 male mice expressing human PSA and HLA-DRB1(*)1501 transgenes were vaccinated with virus-like particle vector encoding PSA (VLPV-PSA) followed by the challenge with Transgenic Adenocarcinoma of Mouse Prostate cells engineered to express PSA (TRAMP-PSA). PSA-specific cellular and humoral immune responses were measured before and after tumor challenge. PSA and CD8 reactivity in the tumors was detected by immunohistochemistry. Tumor growth was compared in vaccinated and control groups. We found that VLPV-PSA could infect mouse dendritic cells in vitro and induce a robust PSA-specific immune response in vivo. A substantial proportion of splenic CD8 T cells (19.6 ± 7.4%) produced IFNγ in response to the immunodominant peptide PSA(65-73). In the blood of vaccinated mice, 18.4 ± 4.1% of CD8 T cells were PSA-specific as determined by the staining with H-2D(b)/PSA(65-73) dextramers. VLPV-PSA vaccination also strongly stimulated production of IgG2a/b anti-PSA antibodies. Tumors in vaccinated mice showed low levels of PSA expression and significant CD8+ T cell infiltration. Tumor growth in VLPV-PSA vaccinated mice was significantly delayed at early time points (p=0.002, Gehan-Breslow test). Our data suggest that TC-83-based VLPV-PSA vaccine can efficiently overcome immune tolerance to PSA, mediate rapid clearance of PSA-expressing tumor cells and delay tumor growth. The VLPV-PSA vaccine will undergo further testing for the immunotherapy of prostate cancer.
Collapse
|
5
|
Abstract
Prostate cancer is the second most diagnosed cancer in men and current treatment of advanced prostate cancer is ineffective. Immunotherapy has emerged as a promising treatment option for metastatic prostate cancer but its clinical application is still in the early stages of development. In order to treat metastatic prostate tumors, new directions must be taken to improve current immunotherapeutic strategies. These include the identification of effective tumor antigens (Ags), the induction of the HLA class II pathway for Ag processing and CD4+ T cell activation, and the ability of tumor cells to act like Ag presenting cells. In this review, we suggest a model for tumor Ag selection, epitope modification and self-processing for presentation by class II proteins as a means of restoring immune activation and tumor clearance. We also outline the importance of a Gamma-IFN-inducible Lysosomal Thiol reductase (GILT) in Ag and modified peptide processing by tumor cells, generation of functional epitopes for T cell recognition, and inclusion of immune checkpoint blockers in cancer immunotherapy. Taken together, this review provides a framework for the future development of novel cancer vaccines and the improvement of existing immunotherapeutics in prostate cancer.
Collapse
Affiliation(s)
- Bently P Doonan
- Department of Microbiology and Immunology, and Hollings Cancer Center, Medical University of South Carolina, 173 Ashley Avenue, Charleston, SC 29425, USA
| | - Azizul Haque
- Department of Microbiology and Immunology, and Hollings Cancer Center, Medical University of South Carolina, 173 Ashley Avenue, Charleston, SC 29425, USA
| |
Collapse
|
6
|
Riabov V, Kim D, Chhina S, Alexander RB, Klyushnenkova EN. Immunostimulatory early phenotype of tumor-associated macrophages does not predict tumor growth outcome in an HLA-DR mouse model of prostate cancer. Cancer Immunol Immunother 2015; 64:873-83. [PMID: 25893810 PMCID: PMC11028626 DOI: 10.1007/s00262-015-1697-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2014] [Accepted: 04/11/2015] [Indexed: 01/17/2023]
Abstract
Tumor-associated macrophages (TAM) were shown to support the progression of many solid tumors. However, anti-tumor properties of TAM were also reported in several types of cancer. Here, we investigated the phenotype and functions of TAM in two transgenic mouse models of prostate cancer that display striking differences in tumor growth outcome. Mice expressing prostate-specific antigen (PSA) as a self-antigen specifically in prostate (PSAtg mice) rejected PSA-expressing transgenic adenocarcinoma of mouse prostate (TRAMP) tumors. However, the introduction of HLA-DRB1*1501 (DR2b) transgene presenting PSA-derived peptides in a MHC class II-restricted manner exacerbated the growth of TRAMP-PSA tumors in DR2bxPSA F 1 mice. Despite the difference in tumor growth outcome, tumors in both strains were equally and intensively infiltrated by macrophages on the first week after tumor challenge. TAM exhibited mixed M1/M2 polarization and simultaneously produced pro-inflammatory (TNFα, IL1β) and anti-inflammatory (IL10) cytokines. TAM from both mouse strains demonstrated antigen-presenting potential and pronounced immunostimulatory activity. Moreover, they equally induced apoptosis of tumor cells. In vivo depletion of macrophages in DR2bxPSA F 1 but not PSAtg mice aggravated tumor growth suggesting that macrophages more strongly contribute to anti-tumor immunity when specific presentation of PSA to CD4+ T cells is possible. In summary, we conclude that in the early stages of tumor progression, the phenotype and functional properties of TAM did not predict tumor growth outcome in two transgenic prostate cancer models. Furthermore, we demonstrated that during the initial stage of prostate cancer development, TAM have the potential to activate T cell immunity and mediate anti-tumor effects.
Collapse
Affiliation(s)
- Vladimir Riabov
- Department of Surgery, Division of Urology, University of Maryland, 10S Pine Street, MSTF-4, Baltimore, MD, 21201, USA,
| | | | | | | | | |
Collapse
|
7
|
Klyushnenkova EN, Riabov VB, Kouiavskaia DV, Wietsma A, Zhan M, Alexander RB. Breaking immune tolerance by targeting CD25+ regulatory T cells is essential for the anti-tumor effect of the CTLA-4 blockade in an HLA-DR transgenic mouse model of prostate cancer. Prostate 2014; 74:1423-32. [PMID: 25111463 DOI: 10.1002/pros.22858] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2013] [Accepted: 06/17/2014] [Indexed: 11/11/2022]
Abstract
INTRODUCTION Recent studies suggest that the cancer immunotherapy based on the blockade of the CTLA-4-mediated inhibitory pathway is efficacious only in select populations, predominantly for immunogenic tumors or when delivered in combination with modalities that can break immunologic tolerance to tumor antigens. METHODS We studied the effect of CD25+ cell depletion and CTLA-4 blockade on the growth of Transgenic Mouse Adenocarcinoma of Prostate (TRAMP)-PSA tumor cells in DR2bxPSA F1 mice. In these mice, immunological tolerance to PSA was established in a context of the HLA-DRB1*1501(DR2b) allele. RESULTS In our model, single administration of anti-CD25 antibody prior to tumor inoculation significantly increased IFN-γ production in response to the CD8 T cell epitope PSA65-73 , and delayed TRAMP-PSA tumor growth compared to mice treated with isotype control antibodies. In contrast, the anti-tumor effect of the anti-CTLA-4 antibody as a monotherapy was marginal. The combinatory treatment with anti-CD25/anti-CTLA-4 antibodies significantly enhanced anti-tumor immunity and caused more profound delay in tumor growth compared to each treatment alone. The proportion of tumor-free animals was higher in the group that received combination treatment (21%) compared to other groups (2-7%). The enhanced anti-tumor immunity in response to the CD25 depletion or CTLA-4 blockade was only seen in the immunogenic TRAMP-PSA tumor model, whereas the effect was completely absent in mice bearing poorly immunogenic TRAMP-C1 tumors. DISCUSSION Our data suggest that breaking immunological tolerance to "self" antigens is essential for the therapeutic effect of CTLA-4 blockade. Such combinatory treatment may be a promising approach for prostate cancer immunotherapy.
Collapse
Affiliation(s)
- Elena N Klyushnenkova
- Department of Surgery, Division of Urology, University of Maryland, Baltimore, Maryland; VA Maryland Health Care System, Baltimore, Maryland
| | | | | | | | | | | |
Collapse
|
8
|
A cytomegalovirus-based vaccine expressing a single tumor-specific CD8+ T-cell epitope delays tumor growth in a murine model of prostate cancer. J Immunother 2013; 35:390-9. [PMID: 22576344 DOI: 10.1097/cji.0b013e3182585d50] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Cytomegalovirus (CMV) is a highly immunogenic virus that results in a persistent, life-long infection in the host typically with no ill effects. Certain unique features of CMV, including its capacity to actively replicate in the presence of strong host CMV-specific immunity, may give CMV an advantage compared with other virus-based vaccine delivery platforms. In the present study, we tested the utility of mouse CMV (mCMV)-based vaccines expressing human prostate-specific antigen (PSA) for prostate cancer immunotherapy in double-transgenic mice expressing PSA and HLA-DRB1*1501 (DR2bxPSA F1 mice). We assessed the capacity of 2 mCMV-based vectors to induce PSA-specific CD8 T-cell responses and affect the growth of PSA-expressing Transgenic Adenocarcinoma of the Mouse Prostate tumors (TRAMP-PSA). In the absence of tumor challenge, immunization with mCMV vectors expressing either a H2-D(b)-restricted epitope PSA(65-73) (mCMV/PSA(65-73)) or the full-length gene for PSA (mCMV/PSA(FL)) induced comparable levels of CD8 T-cell responses that increased (inflated) with time. Upon challenge with TRAMP-PSA tumor cells, animals immunized with mCMV/PSA(65-73) had delay of tumor growth and increased PSA-specific CD8 T-cell responses, whereas animals immunized with mCMV/PSA(FL) showed progressive tumor growth and no increase in number of splenic PSA(65-73)-specific T cells. The data show that a prototype CMV-based prostate cancer vaccine can induce an effective antitumor immune response in a "humanized" double-transgenic mouse model. The observation that mCMV/PSA(FL) is not effective against TRAMP-PSA is consistent with our previous findings that HLA-DRB1*1501-restricted immune responses to PSA are associated with suppression of effective CD8 T-cell responses to TRAMP-PSA tumors.
Collapse
|
9
|
Prostate Cancer and Immunoproteome: Awakening and Reprogramming the Guardian Angels. Arch Immunol Ther Exp (Warsz) 2012; 60:191-8. [DOI: 10.1007/s00005-012-0169-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2011] [Accepted: 03/07/2012] [Indexed: 12/24/2022]
|
10
|
Sui H, Bai Y, Wang K, Li X, Song C, Fu F, Zhang Y, Li L. The anti-tumor effect of Newcastle disease virus HN protein is influenced by differential subcellular targeting. Cancer Immunol Immunother 2010; 59:989-99. [PMID: 20130861 PMCID: PMC11030028 DOI: 10.1007/s00262-010-0821-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2009] [Accepted: 01/19/2010] [Indexed: 12/30/2022]
Abstract
BACKGROUND Immunotherapy is emerging as a major player in the current standard of care for aggressive cancers such as non-small cell lung cancer (NSCLC). The Newcastle disease virus with its tumor-specific replicative and oncolytic abilities is a promising immunotherapeutic candidate. A DNA vaccine expressing the major immunogenic hemagglutinin-neuraminidase (HN) protein of this virus has shown promising results as an immunotherapeutic agent. METHODS In the present study, three different DNA vaccine constructs encoding differentially targeted HN proteins (cytoplasmic or Cy-HN, secreted or Sc-HN and membrane-anchored or M-HN) were generated to evaluate their anti-tumor effect in vitro and in vivo. RESULTS Although all three DNA constructs elicited an immune response, tumor-bearing mice intratumorally injected with M-HN demonstrated a significantly better anti-tumor effect than those injected with Cy-HN or Sc-HN. We also showed that this anti-tumor effect was mediated by higher lymphocyte proliferative response and CTL activity in mice intratumorally injected with M-HN. CONCLUSION The membrane-anchored form of the HN protein appears to be an ideal candidate to develop as an immunotherapeutic agent for NSCLC.
Collapse
MESH Headings
- Animals
- Apoptosis/immunology
- CD8-Positive T-Lymphocytes/cytology
- CD8-Positive T-Lymphocytes/immunology
- Cancer Vaccines/administration & dosage
- Cancer Vaccines/immunology
- Cancer Vaccines/metabolism
- Carcinoma, Non-Small-Cell Lung/drug therapy
- Carcinoma, Non-Small-Cell Lung/immunology
- Carcinoma, Non-Small-Cell Lung/pathology
- Cell Line, Tumor
- Cell Membrane/immunology
- Cell Survival/immunology
- Cytoplasm/immunology
- Cytotoxicity, Immunologic/immunology
- Female
- HN Protein/genetics
- HN Protein/immunology
- Humans
- Lung Neoplasms/drug therapy
- Lung Neoplasms/immunology
- Lung Neoplasms/pathology
- Mice
- Mice, Inbred BALB C
- Mice, Nude
- Microscopy, Electron, Transmission
- Newcastle disease virus/genetics
- Newcastle disease virus/immunology
- T-Lymphocytes, Cytotoxic
- Transfection
- Vaccines, DNA/administration & dosage
- Vaccines, DNA/immunology
- Vaccines, DNA/metabolism
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Hong Sui
- Medical Department, The Tumor Hospital Affiliated Harbin Medical University, Harbin, 150040 Heilongjiang China
| | - Yuxian Bai
- Medical Department, The Tumor Hospital Affiliated Harbin Medical University, Harbin, 150040 Heilongjiang China
| | - Kaibing Wang
- Interventional Department, The Second Hospital Affiliated Harbin Medical University, Harbin, Heilongjiang China
| | - Xi Li
- Division of Swine Disease, National Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Medicine, Harbin, Heilongjiang China
| | - Chun Song
- The Key Laboratory of Cell Transplantation of Ministry of Health, The First Hospital Affiliated Harbin Medical University, Harbin, Heilongjiang China
| | - Fang Fu
- Division of Swine Disease, National Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Medicine, Harbin, Heilongjiang China
| | - Yongxin Zhang
- Division of Swine Disease, National Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Medicine, Harbin, Heilongjiang China
| | - Lejing Li
- Medical Department, The Tumor Hospital Affiliated Harbin Medical University, Harbin, 150040 Heilongjiang China
| |
Collapse
|
11
|
Lekka E, Gritzapis AD, Perez SA, Tsavaris N, Missitzis I, Mamalaki A, Papamichail M, Baxevanis CN. Identification and characterization of a HER-2/neu epitope as a potential target for cancer immunotherapy. Cancer Immunol Immunother 2010; 59:715-27. [PMID: 19904532 PMCID: PMC11030771 DOI: 10.1007/s00262-009-0791-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2009] [Accepted: 10/22/2009] [Indexed: 12/11/2022]
Abstract
Our aim is to develop peptide vaccines that stimulate tumor antigen-specific T-lymphocyte responses against frequently detected cancers. We describe herein a novel HLA-A*0201-restricted epitope, encompassing amino acids 828-836 (residues QIAKGMSYL), which is naturally presented by various HER-2/neu (+) tumor cell lines. HER-2/neu(828-836), [HER-2(9(828))], possesses two anchor residues and stabilized HLA-A*0201 on T2 cells in a concentration-dependent Class I binding assay. This peptide was stable for 3.5 h in an off-kinetic assay. HER-2(9(828)) was found to be immunogenic in HLA-A*0201 transgenic (HHD) mice inducing peptide-specific and functionally potent CTL and long-lasting anti-tumor immunity. Most important, using HLA-A*0201 pentamer analysis we could detect increased ex vivo frequencies of CD8(+) T-lymphocytes specifically recognizing HER-2(9(828)) in 8 out of 20 HLA-A*0201(+) HER-2/neu (+) breast cancer patients. Moreover, HER-2(9(828))-specific human CTL recognized the tumor cell line SKOV3.A2 as well as the primary RS.A2.1.DR1 tumor cell line both expressing HER-2/neu and HLA-A*0201. Finally, therapeutic vaccination with HER-2(9(828)) in HHD mice was proven effective against established transplantable ALC.A2.1.HER tumors, inducing complete tumor regression in 50% of mice. Our data encourage further exploitation of HER-2(9(828)) as a promising candidate for peptide-based cancer vaccines.
Collapse
Affiliation(s)
- Eftychia Lekka
- Cancer Immunology and Immunotherapy Center, Saint Savas Cancer Hospital, Athens, Greece.
| | | | | | | | | | | | | | | |
Collapse
|
12
|
Sfanos KS, Bruno TC, Meeker AK, De Marzo AM, Isaacs WB, Drake CG. Human prostate-infiltrating CD8+ T lymphocytes are oligoclonal and PD-1+. Prostate 2009; 69:1694-703. [PMID: 19670224 PMCID: PMC2782577 DOI: 10.1002/pros.21020] [Citation(s) in RCA: 191] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
BACKGROUND Prostate-infiltrating CD8(+) T lymphocytes (CD8(+) PIL) are prevalent in men with prostate cancer (PCa), however, it is unclear whether the presence of such cells reflects a non-specific immune infiltrate or an oligoclonal, antigen-driven adaptive immune response. METHODS We investigated the complexity of the T-cell receptor (TCR) repertoire in the prostate gland by examining the diversity of CD8(+) TCR beta chain variable region (Vbeta) gene sequences in both the peripheral blood and prostates of cancer patients. Vbeta repertoire analysis was performed by family-specific Vbeta spectratyping and flow cytometry, as well as direct sequence analysis (5' RACE and cloning). Programmed cell death 1 (PD-1 or PDCD1) expression on peripheral blood CD8(+) T cells and CD8(+) PIL was analyzed by flow cytometry. RESULTS CD8(+) PIL isolated from cancer patients exhibited restricted TCR Vbeta gene usage, and identical clones were identified in multiple sites within the prostate. Furthermore, CD8(+) PIL express high levels of the inhibitory receptor PD-1, a cell surface protein associated with an "exhausted" CD8(+) T-cell phenotype. CONCLUSIONS CD8(+) PIL appear to have undergone clonal expansion in response to an as yet unidentified antigen; however, due to the high expression of PD-1, these cells are likely incapable of mounting an effective immune response. The results provide an important basis for further efforts aimed at the identification of specific antigens involved in prostatic inflammation, and suggest that PD-1 blockade may be useful in immunotherapy for PCa.
Collapse
MESH Headings
- Adenocarcinoma/immunology
- Adult
- Antigens, CD/biosynthesis
- Antigens, CD/genetics
- Antigens, CD/immunology
- Apoptosis Regulatory Proteins/biosynthesis
- Apoptosis Regulatory Proteins/genetics
- Apoptosis Regulatory Proteins/immunology
- Biopsy
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/pathology
- Clone Cells/immunology
- Clone Cells/pathology
- Flow Cytometry
- Humans
- Lymphocytes, Tumor-Infiltrating/immunology
- Lymphocytes, Tumor-Infiltrating/pathology
- Male
- Middle Aged
- Neoplastic Stem Cells/immunology
- Neoplastic Stem Cells/pathology
- Programmed Cell Death 1 Receptor
- Prostatic Neoplasms/immunology
- Random Amplified Polymorphic DNA Technique
- Receptors, Antigen, T-Cell/biosynthesis
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/immunology
Collapse
Affiliation(s)
- Karen S. Sfanos
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287-2101
| | - Tullia C. Bruno
- Department of Oncology, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287-2101
| | - Alan K. Meeker
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287-2101
| | - Angelo M. De Marzo
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287-2101
- Department of Oncology, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287-2101
- Department of Urology, James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287-2101
| | - William B. Isaacs
- Department of Oncology, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287-2101
- Department of Urology, James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287-2101
| | - Charles G. Drake
- Department of Oncology, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287-2101
- Department of Urology, James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287-2101
- Correspondence to: Charles G. Drake, Johns Hopkins SKCCC, 1650 Orleans Street, CRBI 410, Baltimore, MD 21231. Phone: 410-502-7523; Fax: 443-287-4653;
| |
Collapse
|