1
|
Dai W, Li Y, Wu S, Wang Q, Zheng X, Zhang J, Han X, Zhou Y. Identification of MAGE-A10 specific T cell receptor promising in immunotherapy of hepatocellular carcinoma. Int J Biol Macromol 2025:144243. [PMID: 40379175 DOI: 10.1016/j.ijbiomac.2025.144243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2025] [Revised: 05/12/2025] [Accepted: 05/13/2025] [Indexed: 05/19/2025]
Abstract
Due to the limitations of current treatment strategies, hepatocellular carcinoma (HCC) continues to impose a severe burden on people's health. In the process of exploring novel therapies, T cell receptor-engineered T cell (TCR-T) therapy has been extensively developed in HCC immunotherapy. Melanoma-associated antigen family A member 10 (MAGE-A10) is a cancer-testis antigen (CTA), specifically expressed on HCC cells. However, the identification of TCR-T targeting MAGE-A10 in HCC remains rarely discussed. In this study, single-cell RNA sequencing (scRNA-seq) and TCR sequencing (scTCR-seq) were performed on samples from HCC patients. The cellular landscape of HCC was illustrated through a single-cell atlas. Reactive T cells were defined based on the matched T cells. Additionally, most reactive T cells were enriched in CD4_CD69_Th, CD4_FOXP3_Treg, CD4_CXCL13_TEX, and CD8_CXCL13_TEX. GLIPH2 was utilized to cluster TCRs from reactive T cells, enabling the identification of reactive TCRs. TCRMatch predicted MAGE-A10 as a specific antigen recognized by one of the reactive TCRs. Furthermore, the affinity assessments between human leukocyte antigen (HLA), epitope of MAGE-A10, and the identified TCR were performed with NetMHCpan and DLpTCR. Finally, cytotoxicity assays indicated the specific recognition and killing of MAGE-A10-TCR-T cells against HCC cells, paving the way for TCR-T immunotherapy in HCC.
Collapse
Affiliation(s)
- Wei Dai
- Tumor Biological Diagnosis and Treatment Center, The Third Affiliated Hospital of Soochow University, Changzhou 213003, China; Jiangsu Engineering Research Center for Tumor Immunotherapy, Changzhou 213003, China; Institute of Cell Therapy, Soochow University, Changzhou 213003, China
| | - Yuanqi Li
- Tumor Biological Diagnosis and Treatment Center, The Third Affiliated Hospital of Soochow University, Changzhou 213003, China; Jiangsu Engineering Research Center for Tumor Immunotherapy, Changzhou 213003, China; Institute of Cell Therapy, Soochow University, Changzhou 213003, China
| | - Shaoxian Wu
- Tumor Biological Diagnosis and Treatment Center, The Third Affiliated Hospital of Soochow University, Changzhou 213003, China; Jiangsu Engineering Research Center for Tumor Immunotherapy, Changzhou 213003, China; Institute of Cell Therapy, Soochow University, Changzhou 213003, China
| | - Qi Wang
- Tumor Biological Diagnosis and Treatment Center, The Third Affiliated Hospital of Soochow University, Changzhou 213003, China; Jiangsu Engineering Research Center for Tumor Immunotherapy, Changzhou 213003, China; Institute of Cell Therapy, Soochow University, Changzhou 213003, China
| | - Xiao Zheng
- Tumor Biological Diagnosis and Treatment Center, The Third Affiliated Hospital of Soochow University, Changzhou 213003, China; Jiangsu Engineering Research Center for Tumor Immunotherapy, Changzhou 213003, China; Institute of Cell Therapy, Soochow University, Changzhou 213003, China
| | - Jinping Zhang
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou 215123, China
| | - Xiao Han
- Institute of Pediatrics, Guangzhou Women and Children's Medical Centre, Guangzhou Medical University, Guangzhou 510623, China.
| | - You Zhou
- Tumor Biological Diagnosis and Treatment Center, The Third Affiliated Hospital of Soochow University, Changzhou 213003, China; Jiangsu Engineering Research Center for Tumor Immunotherapy, Changzhou 213003, China; Institute of Cell Therapy, Soochow University, Changzhou 213003, China.
| |
Collapse
|
2
|
Mahmoud AM. Cancer testis antigens as immunogenic and oncogenic targets in breast cancer. Immunotherapy 2018; 10:769-778. [PMID: 29926750 PMCID: PMC6462849 DOI: 10.2217/imt-2017-0179] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2017] [Accepted: 03/16/2018] [Indexed: 01/07/2023] Open
Abstract
Breast cancer cells frequently express tumor-associated antigens that can elicit immune responses to eradicate cancer. Cancer-testis antigens (CTAs) are a group of tumor-associated antigens that might serve as ideal targets for cancer immunotherapy because of their cancer-restricted expression and robust immunogenicity. Previous clinical studies reported that CTAs are associated with negative hormonal status, aggressive tumor behavior and poor survival. Furthermore, experimental studies have shown the ability of CTAs to induce both cellular and humoral immune responses. They also demonstrated the implication of CTAs in promoting cancer cell growth, inhibiting apoptosis and inducing cancer cell invasion and migration. In the current review, we attempt to address the immunogenic and oncogenic potential of CTAs and their current utilization in therapeutic interventions for breast cancer.
Collapse
Affiliation(s)
- Abeer M Mahmoud
- Department of Physical Therapy, College of Applied Health Sciences, University of Illinois at Chicago, Chicago, IL 60612, USA
- Department of Pathology, South Egypt Cancer Institute, Assiut University, Assiut 71111, Egypt
| |
Collapse
|
3
|
Neek M, Tucker JA, Kim TI, Molino NM, Nelson EL, Wang SW. Co-delivery of human cancer-testis antigens with adjuvant in protein nanoparticles induces higher cell-mediated immune responses. Biomaterials 2017; 156:194-203. [PMID: 29202325 DOI: 10.1016/j.biomaterials.2017.11.022] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Revised: 11/07/2017] [Accepted: 11/19/2017] [Indexed: 12/19/2022]
Abstract
Nanoparticles have attracted considerable interest as cancer vaccine delivery vehicles for inducing sufficient CD8+ T cell-mediated immune responses to overcome the low immunogenicity of the tumor microenvironment. Our studies described here are the first to examine the effects of clinically-tested human cancer-testis (CT) peptide epitopes within a synthetic nanoparticle. Specifically, we focused on two significant clinical CT targets, the HLA-A2 restricted epitopes of NY-ESO-1 and MAGE-A3, using a viral-mimetic packaging strategy. Our data shows that simultaneous delivery of a NY-ESO-1 epitope (SLLMWITQV) and CpG using the E2 subunit assembly of pyruvate dehydrogenase (E2 nanoparticle), resulted in a 25-fold increase in specific IFN-γ secretion in HLA-A2 transgenic mice. This translated to a 15-fold increase in lytic activity toward target cancer cells expressing the antigen. Immunization with a MAGE-A3 epitope (FLWGPRALV) delivered with CpG in E2 nanoparticles yielded an increase in specific IFN-γ secretion and cell lysis by 6-fold and 9-fold, respectively. Furthermore, combined delivery of NY-ESO-1 and MAGE-A3 antigens in E2 nanoparticles yielded an additive effect that increased lytic activity towards cells bearing NY-ESO-1+ and MAGE-A3+. Our investigations demonstrate that formulation of CT antigens within a nanoparticle can significantly enhance antigen-specific cell-mediated responses, and the combination of the two antigens in a vaccine can preserve the increased individual responses that are observed for each antigen alone.
Collapse
Affiliation(s)
- Medea Neek
- Department of Chemical Engineering and Materials Science, University of California, Irvine, CA 92697, USA
| | - Jo Anne Tucker
- Department of Medicine, University of California, Irvine, CA 92697, USA
| | - Tae Il Kim
- Department of Biomedical Engineering, University of California, Irvine, CA 92697, USA
| | - Nicholas M Molino
- Department of Chemical Engineering and Materials Science, University of California, Irvine, CA 92697, USA
| | - Edward L Nelson
- Department of Medicine, University of California, Irvine, CA 92697, USA; Chao Family Comprehensive Cancer Center, University of California, Irvine, CA 92697, USA; Institute for Immunology, University of California, Irvine, CA 92697, USA
| | - Szu-Wen Wang
- Department of Chemical Engineering and Materials Science, University of California, Irvine, CA 92697, USA; Department of Biomedical Engineering, University of California, Irvine, CA 92697, USA; Chao Family Comprehensive Cancer Center, University of California, Irvine, CA 92697, USA.
| |
Collapse
|
4
|
Peper JK, Bösmüller HC, Schuster H, Gückel B, Hörzer H, Roehle K, Schäfer R, Wagner P, Rammensee HG, Stevanović S, Fend F, Staebler A. HLA ligandomics identifies histone deacetylase 1 as target for ovarian cancer immunotherapy. Oncoimmunology 2015; 5:e1065369. [PMID: 27467910 PMCID: PMC4910750 DOI: 10.1080/2162402x.2015.1065369] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Revised: 06/16/2015] [Accepted: 06/17/2015] [Indexed: 10/28/2022] Open
Abstract
The recent approval of clincially effective immune checkpoint inhibitors illustrates the potential of cancer immunotherapy. A challenging task remains the identification of specific targets guiding immunotherapy. Facilitated by technical advances, the direct identification of physiologically relevant targets is enabled by analyzing the HLA ligandome of cancer cells. Since recent publications demonstrate the immunogenicity of ovarian cancer (OvCa), immunotherapies, including peptide-based cancer vaccines, represent a promising treatment approach. To identify vaccine peptides, we employed a combined strategy of HLA ligandomics in high-grade serous OvCa samples and immunogenicity analysis. Only few proteins were naturally presented as HLA ligands on all samples analyzed, including histone deacetylase (HDAC) 1 and 2. In vitro priming of CD8(+) T cells demonstrated that two HDAC1/2-derived HLA ligands can induce T-cell responses, capable of killing HLA-matched tumor cells. High HDAC1 expression shown by immunohistochemistry in 136 high-grade serous OvCa patients associated with significantly reduced overall survival (OS), whereas patients with high numbers of CD3(+) tumor-infiltrating lymphocytes (TILs) in the tumor epithelium and CD8(+) TILs in the tumor stroma showed improved OS. However, correlating HDAC1 expression with TILs, high levels of TILs abrogated the impact of HDAC1 on OS. This study strengthens the role of HDAC1/2 as an important tumor antigen in OvCa, demonstrating its impact on OS in a large cohort of OvCa patients. We further identified two immunogenic HDAC1-derived peptides, which frequently induce multi-functional T-cell responses in many donors, suitable for future multi-peptide vaccine trials in OvCa patients.
Collapse
Affiliation(s)
- Janet Kerstin Peper
- Department of Immunology, Institute of Cell Biology, University of Tübingen , Tübingen, Germany
| | | | - Heiko Schuster
- Department of Immunology, Institute of Cell Biology, University of Tübingen , Tübingen, Germany
| | - Brigitte Gückel
- Department of Obstetrics and Gynecology, University Hospital of Tübingen , Tübingen, Germany
| | - Helen Hörzer
- Department of Obstetrics and Gynecology, University Hospital of Tübingen , Tübingen, Germany
| | - Kevin Roehle
- Department of Immunology, Institute of Cell Biology, University of Tübingen , Tübingen, Germany
| | - Richard Schäfer
- Department of Clinical and Experimental Transfusion Medicine, University Hospital of Tübingen , Tübingen, Germany
| | - Philipp Wagner
- Department of Obstetrics and Gynecology, University Hospital of Tübingen , Tübingen, Germany
| | - Hans-Georg Rammensee
- Department of Immunology, Institute of Cell Biology, University of Tübingen, Tübingen, Germany; German Cancer Consortium (DKTK), DKFZ partner site Tübingen, Tübingen, Germany
| | - Stefan Stevanović
- Department of Immunology, Institute of Cell Biology, University of Tübingen, Tübingen, Germany; German Cancer Consortium (DKTK), DKFZ partner site Tübingen, Tübingen, Germany
| | - Falko Fend
- Institute of Pathology, University Hospital of Tübingen, Tübingen, Germany; German Cancer Consortium (DKTK), DKFZ partner site Tübingen, Tübingen, Germany
| | - Annette Staebler
- Institute of Pathology, University Hospital of Tübingen , Tübingen, Germany
| |
Collapse
|
5
|
Vassilev L, Ranki T, Joensuu T, Jäger E, Karbach J, Wahle C, Partanen K, Kairemo K, Alanko T, Turkki R, Linder N, Lundin J, Ristimäki A, Kankainen M, Hemminki A, Backman C, Dienel K, von Euler M, Haavisto E, Hakonen T, Juhila J, Jäderberg M, Priha P, Vuolanto A, Pesonen S. Repeated intratumoral administration of ONCOS-102 leads to systemic antitumor CD8 + T-cell response and robust cellular and transcriptional immune activation at tumor site in a patient with ovarian cancer. Oncoimmunology 2015; 4:e1017702. [PMID: 26140248 PMCID: PMC4485730 DOI: 10.1080/2162402x.2015.1017702] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Revised: 02/05/2015] [Accepted: 02/06/2015] [Indexed: 12/04/2022] Open
Abstract
Adenoviruses are excellent immunotherapeutic agents with a unique ability to prime and boost immune responses. Recombinant adenoviruses cause immunogenic cancer cell death and subsequent release of tumor antigens for antigen presenting cells, resulting in the priming of potent tumor-specific immunity. This effect may be further enhanced by immune-stimulating transgenes expressed by the virus. We report a case of a 38-year-old female with Stage 3 metastatic micropapillary serous carcinoma of the ovary. She was treated in a Phase I study with a granulocyte-macrophage colony stimulating factor (GMCSF)-expressing oncolytic adenovirus, Ad5/3-D24-GMCSF (ONCOS-102). The treatment resulted in progressive infiltration of CD8+ lymphocytes into the tumor and concomitant systemic induction of several tumor-specific CD8+ T-cell populations. The patient was alive at the latest follow up more than 20 months after initiation of the study.
Collapse
Affiliation(s)
| | - T Ranki
- Oncos Therapeutics ; Helsinki, Finland
| | - T Joensuu
- Docrates Cancer Center ; Helsinki, Finland
| | - E Jäger
- Hämatologie-Onkologie; Krankenhaus Nordwest ; Frankfurt, Germany
| | - J Karbach
- Hämatologie-Onkologie; Krankenhaus Nordwest ; Frankfurt, Germany
| | - C Wahle
- Hämatologie-Onkologie; Krankenhaus Nordwest ; Frankfurt, Germany
| | - K Partanen
- Docrates Cancer Center ; Helsinki, Finland
| | - K Kairemo
- Docrates Cancer Center ; Helsinki, Finland
| | - T Alanko
- Docrates Cancer Center ; Helsinki, Finland
| | - R Turkki
- Institute for Molecular Medicine Finland (FIMM) ; Helsinki, Finland
| | - N Linder
- Institute for Molecular Medicine Finland (FIMM) ; Helsinki, Finland
| | - J Lundin
- Institute for Molecular Medicine Finland (FIMM) ; Helsinki, Finland
| | - A Ristimäki
- Division of Pathology; HUSLAB and Haartman Institute; Helsinki University Central Hospital ; Helsinki, Finland ; Genome-Scale Biology; Research Programs Unit; University of Helsinki ; Helsinki, Finland
| | - M Kankainen
- Institute for Molecular Medicine Finland (FIMM) ; Helsinki, Finland
| | - A Hemminki
- University of Helsinki and Helsinki University Central Hospital; Cancer Gene Therapy Group ; Helsinki, Finland
| | - C Backman
- Oncos Therapeutics ; Helsinki, Finland
| | - K Dienel
- Oncos Therapeutics ; Helsinki, Finland
| | | | | | - T Hakonen
- Oncos Therapeutics ; Helsinki, Finland
| | - J Juhila
- Oncos Therapeutics ; Helsinki, Finland
| | | | - P Priha
- Oncos Therapeutics ; Helsinki, Finland
| | | | - S Pesonen
- Oncos Therapeutics ; Helsinki, Finland
| |
Collapse
|
6
|
Identification of two novel HLA-A*0201-restricted CTL epitopes derived from MAGE-A4. Clin Dev Immunol 2011; 2010:567594. [PMID: 21350607 PMCID: PMC3042617 DOI: 10.1155/2010/567594] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2010] [Revised: 10/08/2010] [Accepted: 12/05/2010] [Indexed: 11/24/2022]
Abstract
MAGE-A antigens belong to cancer/testis (CT) antigens that are expressed in tumors but not in normal tissues except testis and placenta. MAGE-A antigens and their epitope peptides have been used in tumor immunotherapy trials. MAGE-A4 antigen is extensively expressed in various histological types of tumors, so it represents an attractive target for tumor immunotherapy. In this study, we predicted HLA-A*0201-restricted cytotoxic T lymphocyte (CTL) epitopes of MAGE-A4, followed by peptide/HLA-A*0201 affinity and complex stability assays. Of selected four peptides (designated P1, P2, P3, and P4), P1 (MAGE-A4286-294, KVLEHVVRV) and P3 (MAGE-A4272-280, FLWGPRALA) could elicit peptide-specific CTLs both in vitro from HLA-A*0201-positive PBMCs and in HLA-A*0201/Kb transgenic mice. And the induced CTLs could lyse target cells in an HLA-A*0201-restricted fashion, demonstrating that the two peptides are HLA-A*0201-restricted CTL epitopes and could serve as targets for therapeutic antitumoral vaccination.
Collapse
|
7
|
Marmé A, Zimmermann HP, Moldenhauer G, Schorpp-Kistner M, Müller C, Keberlein O, Giersch A, Kretschmer J, Seib B, Spiess E, Hunziker A, Merchán F, Möller P, Hahn U, Kurek R, Marmé F, Bastert G, Wallwiener D, Ponstingl H. Loss of Drop1 expression already at early tumor stages in a wide range of human carcinomas. Int J Cancer 2008; 123:2048-56. [PMID: 18709643 DOI: 10.1002/ijc.23763] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
In a study on gene deregulation in ovarian carcinoma we found a mRNA coding for a 350 kDa protein, Drop1, to be downregulated 20- to 180-fold in the majority of ovarian and mammary carcinomas. The mRNA is encoded by a set of exons in the 5' region of the SYNE1 gene. Immunohistochemical staining for Drop1 protein by a specific monoclonal antibody corresponds to the pattern seen for the mRNA. cDNA arrays of matched pairs of tumor and normal tissue and in situ hybridizations confirmed the drastic loss of Drop1 mRNA as a common feature in uterus, cervix, kidney, lung, thyroid and pancreas carcinomas, already at early tumor stages and in all metastases. Two-hybrid studies suggest a role of this deficiency in the malignant progression of epithelial tumors.
Collapse
Affiliation(s)
- Alexander Marmé
- Department of Obstetrics and Gynecology, University of Heidelberg, Heidelberg, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
8
|
Conrad H, Gebhard K, Krönig H, Neudorfer J, Busch DH, Peschel C, Bernhard H. CTLs Directed against HER2 Specifically Cross-React with HER3 and HER4. THE JOURNAL OF IMMUNOLOGY 2008; 180:8135-45. [DOI: 10.4049/jimmunol.180.12.8135] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
9
|
Gückel B, Rentzsch C, Nastke MD, Marmé A, Gruber I, Stevanović S, Kayser S, Wallwiener D. Pre-existing T-cell immunity against mucin-1 in breast cancer patients and healthy volunteers. J Cancer Res Clin Oncol 2005; 132:265-74. [PMID: 16374613 DOI: 10.1007/s00432-005-0064-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2005] [Accepted: 11/11/2005] [Indexed: 01/28/2023]
Abstract
PURPOSE There is evidence that some tumor patients are able to generate tumor-associated antigen (TAA)-specific T-cell immunity spontaneously. However, little is understood about the existence and nature of self-reactive T-cells that recognize TAA in healthy donors (HD). METHODS Human mucin (MUC-1), a highly glycosylated transmembrane protein, is a well characterized TAA expressed by epithelial tumors. We compared endogenous MUC-1-specific T-cell immunity of breast cancer patients (BCP) and healthy volunteers using two MUC-1-derived HLA-A*0201-restricted peptides (MUC-1(950-958), MUC-1(12-20)). Antigen-dependent interferon (IFN)-gamma and Granzyme B expression of T-cells were analysed by a reverse transcription-polymerase chain reaction (qRT-PCR)-based assay. RESULTS A 32% of BCP and 43% of healthy volunteers revealed pre-existent CD8+ T-cells specific for MUC-1(950-958) but not for MUC-1(12-20). In patients, MUC-1-specific T-cells have been detected mainly in early stage disease prior adjuvant therapy. Those T-cells showed MUC-1-dependent IFN-gamma production after short-term stimulation but no clear Granzyme B expression. However, after repetitive in vitro stimulations using peptide-pulsed CD40-stimulated B-cell lines as autologous antigen presenting cells (APC) T-cell lines exhibited lytic capacity against HLA-A*0201+/MUC-1+ tumor cells. CONCLUSION MUC-1(950-958) is a dominant tumor antigen against which CD8+ T-cells were found frequently in BCP as well as in HD. Until now, this was only known for MelanA/MART-1. In contrast to previous reports, MUC-1-specific immunity was not linked to gender or number of pregnancies in women. Whether MUC-1(950-958)-related immunity highlights a yet unknown cross-reactivity in HD remains unclear. The presence of MUC-1-specific T-cells in some BCP may reflect a balance between immune tolerance and immune defence during aetiopathology.
Collapse
Affiliation(s)
- Brigitte Gückel
- Department of Obstetrics and Gynecology, University of Tübingen, Calwerstrasse 7, 72076, Tübingen, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
10
|
Gückel B, Stumm S, Rentzsch C, Marmé A, Mannhardt G, Wallwiener D. A CD80-transfected human breast cancer cell variant induces HER-2/neu-specific T cells in HLA-A*02-matched situations in vitro as well as in vivo. Cancer Immunol Immunother 2005; 54:129-40. [PMID: 15365776 PMCID: PMC11034349 DOI: 10.1007/s00262-004-0583-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2004] [Accepted: 06/14/2004] [Indexed: 10/26/2022]
Abstract
Adjuvant treatment is still only working in a small percentage of breast cancer patients. Therefore, new strategies need to be developed. Immunotherapies are a very promising approach because they could successfully attack tumor cells in the stage of dormancy. To assess the feasibility of using an allogeneic approach for vaccination of breast cancer patients, we selected a CD80-transfected breast cancer cell line based on its immunogenic properties. Using CD80+ KS breast cancer cells and human leukocyte antigen (HLA)-A*02-matched peripheral blood mononuclear cells (PBMCs) of breast cancer patients in allogeneic mixed lymphocyte-tumor cell cultures (MLTCs), it was possible to isolate HLA-A*02-restricted cytotoxic T cells (CTLs). Furthermore, a genetically modified KS variant expressing influenza A matrix protein serving as a surrogate tumor-associated antigen (TAA) was able to stimulate flu peptide-specific T cells alongside the induction of alloresponses in MLTCs. KS breast cancer cells were demonstrated to express already known TAAs such as CEA, MUC-1, MAGE-1, MAGE-2, and MAGE-3. To further improve antigenicity, HER-2/neu was added to this panel as a marker antigen known to elicit HLA-A*02-restricted CTLs in patients with breast cancer. Thus, the antigen-processing and antigen-presentation capacity of KS cells was further demonstrated by the stimulation of HER-2/neu-specific CD8+ T cells in PBMCs of breast cancer patients in vitro. These results gave a good rationale for a phase I/II trial, where the CD80+ HER-2/neu-overexpressing KS variant is actually used as a cellular vaccine in patients with metastatic breast cancer. As a proof of principle, we present data from two patients where a significant increase of interferon-gamma (IFN-gamma) release was detected when postvaccination PBMCs were stimulated by allogeneic vaccine cells as well as by HLA-A*02-restricted HER-2/neu epitopes. In whole cell vaccine trials, monitoring is particularly challenging because of strong alloresponses and limited knowledge of TAAs. In this study, a panel of HER-2/neu epitopes, together with the quantitative real time (qRT)-PCR method to analyze vaccine-induced cytokines secreted by T cells, proved to be highly sensitive and feasible to perform an "immunological staging" following vaccination.
Collapse
Affiliation(s)
- Brigitte Gückel
- Department of Gynecology and Obstetrics, University of Tübingen, Calwerstrasse 7, 72076, Tübingen, Germany.
| | | | | | | | | | | |
Collapse
|