A human ErbB2-specific T-cell receptor confers potent antitumor effector functions in genetically engineered primary cytotoxic lymphocytes.

The ErbB2 protein is a member of the tyrosine kinase family of growth factor receptors that is overexpressed in cancers of the breast, ovary, stomach, kidney, colon, and lung, and therefore represents an attractive candidate antigen for targeted cancer immunotherapy. Cytotoxic T lymphocytes specific for various immunogenic ErbB2 peptides have been described, but they often exhibit both poor functional avidity and tumor reactivity. In order to generate potent CD8(+) T cells with specificity for the ErbB2(369-377) peptide, we performed one round of in vitro peptide stimulation of CD8(+) T cells isolated from an HLA-A2(+) patient who was previously vaccinated with autologous dendritic cells pulsed with HLA class I ErbB2 peptides. Using this approach, we enriched highly avid ErbB2-reactive T cells with strong ErbB2-specific, antitumor effector functions. We then stimulated these ErbB2-reactive T cells with ErbB2(+) HLA-A2(+) tumor cells in vitro and sorted tumor-activated ErbB2(369-377) peptide T cells, which allowed for the isolation of a novel T-cell receptor (TCR) with ErbB2(369-377) peptide specificity. Primary human CD8(+) T cells genetically modified to express this ErbB2-specific TCR specifically bound ErbB2(369-377) peptide containing HLA-A2 tetramers, and efficiently recognized target cells pulsed with low nanomolar concentrations of ErbB2(369-377) peptide as well as nonpulsed ErbB2(+) HLA-A2(+) tumor cell lines in vitro. In a novel xenograft model, ErbB2-redirected T cells also significantly delayed progression of ErbB2(+) HLA-A2(+) human tumor in vivo. Together, these results support the notion that redirection of normal T-cell specificity by TCR gene transfer can have potential applications in the adoptive immunotherapy of ErbB2-expressing malignancies.

[1]  R. Sandaltzopoulos,et al.  Novel recombinant human b7-h4 antibodies overcome tumoral immune escape to potentiate T-cell antitumor responses. , 2013, Cancer research.

[2]  L. Benson,et al.  Enzymatic Discovery of a HER-2/neu Epitope That Generates Cross-Reactive T Cells , 2013, The Journal of Immunology.

[3]  G. Coukos,et al.  Primary Human Ovarian Epithelial Cancer Cells Broadly Express HER2 at Immunologically-Detectable Levels , 2012, PloS one.

[4]  H. Nisenbaum,et al.  A Novel Dendritic Cell-based Immunization Approach for the Induction of Durable Th1-polarized Anti-HER-2/neu Responses in Women With Early Breast Cancer , 2012, Journal of immunotherapy.

[5]  J. Waxman,et al.  CD3 limits the efficacy of TCR gene therapy in vivo. , 2011, Blood.

[6]  H. Ikeda,et al.  Improved expression and reactivity of transduced tumor-specific TCRs in human lymphocytes by specific silencing of endogenous TCR. , 2009, Cancer research.

[7]  S. Rosenberg,et al.  Gene therapy with human and mouse T-cell receptors mediates cancer regression and targets normal tissues expressing cognate antigen. , 2009, Blood.

[8]  J. Wargo,et al.  Recognition of NY-ESO-1+ tumor cells by engineered lymphocytes is enhanced by improved vector design and epigenetic modulation of tumor antigen expression , 2009, Cancer Immunology, Immunotherapy.

[9]  D. Busch,et al.  CTLs Directed against HER2 Specifically Cross-React with HER3 and HER41 , 2008, The Journal of Immunology.

[10]  Soldano Ferrone,et al.  HLA Class I Antigen Processing Machinery Component Expression and Intratumoral T-Cell Infiltrate as Independent Prognostic Markers in Ovarian Carcinoma , 2008, Clinical Cancer Research.

[11]  P. Romero,et al.  Molecular Design of the Cαβ Interface Favors Specific Pairing of Introduced TCRαβ in Human T Cells1 , 2008, The Journal of Immunology.

[12]  S. Rosenberg,et al.  Enhanced antitumor activity of T cells engineered to express T-cell receptors with a second disulfide bond. , 2007, Cancer research.

[13]  H. Nisenbaum,et al.  Targeting HER-2/neu in early breast cancer development using dendritic cells with staged interleukin-12 burst secretion. , 2007, Cancer research.

[14]  B. Engels,et al.  Redirecting T lymphocyte specificity by T cell receptor gene transfer--a new era for immunotherapy. , 2007, Molecular aspects of medicine.

[15]  S. Rosenberg,et al.  Gene Transfer of Tumor-Reactive TCR Confers Both High Avidity and Tumor Reactivity to Nonreactive Peripheral Blood Mononuclear Cells and Tumor-Infiltrating Lymphocytes1 , 2006, The Journal of Immunology.

[16]  S. Rosenberg,et al.  Cancer Regression in Patients After Transfer of Genetically Engineered Lymphocytes , 2006, Science.

[17]  Yangbing Zhao,et al.  Enhanced antitumor activity of murine-human hybrid T-cell receptor (TCR) in human lymphocytes is associated with improved pairing and TCR/CD3 stability. , 2006, Cancer research.

[18]  S. Rosenberg,et al.  Recognition of Fresh Human Tumor by Human Peripheral Blood Lymphocytes Transduced with a Bicistronic Retroviral Vector Encoding a Murine Anti-p53 TCR , 2005, The Journal of Immunology.

[19]  C. Shriver,et al.  Clinical trial results of a HER2/neu (E75) vaccine to prevent recurrence in high-risk breast cancer patients. , 2005, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[20]  K. Black,et al.  HER-2, gp100, and MAGE-1 Are Expressed in Human Glioblastoma and Recognized by Cytotoxic T Cells , 2004, Cancer Research.

[21]  M. Cheever,et al.  Effect of dose on immune response in patients vaccinated with an her-2/neu intracellular domain protein--based vaccine. , 2004, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[22]  B. Czerniecki,et al.  Rapid High Efficiency Sensitization of CD8+ T Cells to Tumor Antigens by Dendritic Cells Leads to Enhanced Functional Avidity and Direct Tumor Recognition Through an IL-12-Dependent Mechanism 1 , 2003, The Journal of Immunology.

[23]  K. Darcy,et al.  Evaluation of monoclonal humanized anti-HER2 antibody, trastuzumab, in patients with recurrent or refractory ovarian or primary peritoneal carcinoma with overexpression of HER2: a phase II trial of the Gynecologic Oncology Group. , 2003, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[24]  G. Hortobagyi,et al.  Toxicity, immunogenicity, and induction of E75-specific tumor-lytic CTLs by HER-2 peptide E75 (369-377) combined with granulocyte macrophage colony-stimulating factor in HLA-A2+ patients with metastatic breast and ovarian cancer. , 2002, Clinical cancer research : an official journal of the American Association for Cancer Research.

[25]  T. Schumacher,et al.  T-cell-receptor gene therapy , 2002, Nature Reviews Immunology.

[26]  M. Cheever,et al.  Immunization of cancer patients with a HER-2/neu, HLA-A2 peptide, p369-377, results in short-lived peptide-specific immunity. , 2002, Clinical cancer research : an official journal of the American Association for Cancer Research.

[27]  C. Peschel,et al.  Antihuman epidermal growth factor receptor 2 (HER2) monoclonal antibody trastuzumab enhances cytolytic activity of class I-restricted HER2-specific T lymphocytes against HER2-overexpressing tumor cells. , 2002, Cancer research.

[28]  T. Schumacher,et al.  Can the Low-Avidity Self-Specific T Cell Repertoire Be Exploited for Tumor Rejection?1 , 2002, The Journal of Immunology.

[29]  Alessandro Sette,et al.  Identification of New Epitopes from Four Different Tumor-Associated Antigens: Recognition of Naturally Processed Epitopes Correlates with HLA-A∗0201-Binding Affinity1 , 2001, The Journal of Immunology.

[30]  J. Murray,et al.  Peptide priming of cytolytic activity to HER-2 epitope 369-377 in healthy individuals. , 2000, Clinical cancer research : an official journal of the American Association for Cancer Research.

[31]  L. Kanz,et al.  Induction of cytotoxic T-lymphocyte responses in vivo after vaccinations with peptide-pulsed dendritic cells. , 2000, Blood.

[32]  B. Seliger,et al.  HER‐2/neu is expressed in human renal cell carcinoma at heterogeneous levels independently of tumor grading and staging and can be recognized by HLA‐A2.1‐restricted cytotoxic T lymphocytes , 2000, International journal of cancer.

[33]  A Sette,et al.  Identification of new HER2/neu-derived peptide epitopes that can elicit specific CTL against autologous and allogeneic carcinomas and melanomas. , 1999, Journal of immunology.

[34]  S. Rosenberg,et al.  Immunization with a peptide epitope (p369-377) from HER-2/neu leads to peptide-specific cytotoxic T lymphocytes that fail to recognize HER-2/neu+ tumors. , 1998, Cancer research.

[35]  H. Rammensee,et al.  Her-2/neu-derived peptides are tumor-associated antigens expressed by human renal cell and colon carcinoma lines and are recognized by in vitro induced specific cytotoxic T lymphocytes. , 1998, Cancer research.

[36]  M. Connors,et al.  Effects of CD28 costimulation on long-term proliferation of CD4+ T cells in the absence of exogenous feeder cells. , 1997, Journal of immunology.

[37]  S. Rosenberg,et al.  Quantitative correlation between HLA class I allele expression and recognition of melanoma cells by antigen-specific cytotoxic T lymphocytes. , 1995, Cancer research.

[38]  D. Slamon,et al.  Antibody to HER-2/neu receptor blocks DNA repair after cisplatin in human breast and ovarian cancer cells. , 1994, Oncogene.

[39]  F. Marincola,et al.  Locus-specific analysis of human leukocyte antigen class I expression in melanoma cell lines. , 1994, Journal of immunotherapy with emphasis on tumor immunology : official journal of the Society for Biological Therapy.

[40]  G. Steger,et al.  HER-2-Positive Breast Cancer , 2012, BioDrugs.

[41]  V. Kaklamani,et al.  HER2-Positive Breast Cancer , 2012, Drugs.

[42]  Michel Sadelain,et al.  Targeting tumours with genetically enhanced T lymphocytes , 2003, Nature Reviews Cancer.

[43]  R. Debets,et al.  Genetic engineering of T cell specificity for immunotherapy of cancer. , 2003, Human immunology.

[44]  S. Lam,et al.  Prevalence and significance of HER-2/neu amplification in epithelial ovarian cancer. , 1995, Gynecologic and obstetric investigation.