Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
JCI Insight ; 3(4)2018 02 22.
Article in English | MEDLINE | ID: mdl-29467338

ABSTRACT

EBV infection is associated with a number of malignancies of clinical unmet need, including Hodgkin lymphoma, nasopharyngeal carcinoma, gastric cancer, and posttransplant lymphoproliferative disease (PTLD), all of which express the EBV protein latent membrane protein 2A (LMP2A), an antigen that is difficult to target by conventional antibody approaches. To overcome this, we utilized phage display technology and a structure-guided selection strategy to generate human T cell receptor-like (TCR-like) monoclonal antibodies with exquisite specificity for the LMP2A-derived nonamer peptide, C426LGGLLTMV434 (CLG), as presented on HLA-A*02:01. Our lead construct, clone 38, closely mimics the native binding mode of a TCR, recognizing residues at position P3-P8 of the CLG peptide. To enhance antitumor potency, we constructed dimeric T cell engaging bispecific antibodies (DiBsAb) of clone 38 and an affinity-matured version clone 38-2. Both DiBsAb showed potent antitumor properties in vitro and in immunodeficient mice implanted with EBV transformed B lymphoblastoid cell lines and human T cell effectors. Clone 38 DiBsAb showed a stronger safety profile compared with its affinity-matured variant, with no activity against EBV- tumor cell lines and a panel of normal tissues, and was less cross-reactive against HLA-A*02:01 cells pulsed with a panel of CLG-like peptides predicted from a proteomic analysis. Clone 38 was also shown to recognize the CLG peptide on other HLA-A*02 suballeles, including HLA-A*02:02, HLA-A*02:04, and HLA-A*02:06, allowing for its potential use in additional populations. Clone 38 DiBsAb is a lead candidate to treat EBV malignancies with one of the strongest safety profiles documented for TCR-like mAbs.


Subject(s)
Antibodies, Bispecific/immunology , Antibodies, Monoclonal/immunology , Epstein-Barr Virus Infections/drug therapy , Neoplasms/drug therapy , Oncogene Proteins, Viral/antagonists & inhibitors , Viral Matrix Proteins/antagonists & inhibitors , Animals , Antibodies, Bispecific/administration & dosage , Antibodies, Bispecific/chemistry , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/chemistry , Blood Buffy Coat , CHO Cells , Cell Line, Tumor , Cricetulus , Cross Reactions , Crystallography, X-Ray , Epstein-Barr Virus Infections/immunology , Epstein-Barr Virus Infections/virology , HEK293 Cells , HLA-A2 Antigen/chemistry , HLA-A2 Antigen/immunology , HLA-A2 Antigen/metabolism , Herpesvirus 4, Human/immunology , Humans , Mice , Models, Molecular , Neoplasms/immunology , Neoplasms/virology , Oncogene Proteins, Viral/immunology , Peptide Library , Receptors, Antigen, T-Cell/chemistry , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Viral Matrix Proteins/chemistry , Viral Matrix Proteins/immunology , Xenograft Model Antitumor Assays
2.
Oncoimmunology ; 6(3): e1267891, 2017.
Article in English | MEDLINE | ID: mdl-28405494

ABSTRACT

T-cell-based therapies have emerged as one of the most clinically effective ways to target solid and non-solid tumors. HER2 is responsible for the oncogenesis and treatment resistance of several human solid tumors. As a member of the HER family of tyrosine kinase receptors, its over-activity confers unfavorable clinical outcome. Targeted therapies directed at this receptor have achieved responses, although development of resistance is common. We explored a novel HER2/CD3 bispecific antibody (HER2-BsAb) platform that while preserving the anti-proliferative effects of trastuzumab, it recruits and activates non-specific circulating T-cells, promoting T cell tumor infiltration and ablating HER2(+) tumors, even when these are resistant to standard HER2-targeted therapies. Its in vitro tumor cytotoxicity, when expressed as EC50, correlated with the surface HER2 expression in a large panel of human tumor cell lines, irrespective of lineage or tumor type. HER2-BsAb-mediated cytotoxicity was relatively insensitive to PD-1/PD-L1 immune checkpoint inhibition. In four separate humanized mouse models of human breast cancer and ovarian cancer cell line xenografts, as well as human breast cancer and gastric cancer patient-derived xenografts (PDXs), HER2-BsAb was highly effective in promoting T cell infiltration and suppressing tumor growth when used in the presence of human peripheral blood mononuclear cells (PBMC) or activated T cells (ATC). The in vivo and in vitro antitumor properties of this BsAb support its further clinical development as a cancer immunotherapeutic.

3.
Clin Cancer Res ; 19(7): 1858-72, 2013 Apr 01.
Article in English | MEDLINE | ID: mdl-23444227

ABSTRACT

PURPOSE: Tumor antigen-specific monoclonal antibodies (mAb) block oncogenic signaling and induce Fcγ receptor (FcγR)-mediated cytotoxicity. However, the role of CD8(+) CTL and FcγR in initiating innate and adaptive immune responses in mAb-treated human patients with cancer is still emerging. EXPERIMENTAL DESIGN: FcγRIIIa codon 158 polymorphism was correlated with survival in 107 cetuximab-treated patients with head and neck cancer (HNC). Flow cytometry was carried out to quantify EGF receptor (EGFR)-specific T cells in cetuximab-treated patients with HNC. The effect of cetuximab on natural killer (NK) cell, dendritic cell (DC), and T-cell activation was measured using IFN-γ release assays and flow cytometry. RESULTS: FcγRIIIa polymorphism did not predict clinical outcome in cetuximab-treated patients with HNC; however, elevated circulating EGFR(853-861)-specific CD8(+) T cells were found in cetuximab-treated patients with HNC (P < 0.005). Cetuximab promoted EGFR-specific cellular immunity through the interaction of EGFR(+) tumor cells and FcγRIIIa on NK cells but not on the polymorphism per se. Cetuximab-activated NK cells induced IFN-γ-dependent expression of DC maturation markers, antigen processing machinery components such as TAP-1/2 and T-helper cell (T(H)1) chemokines through NKG2D/MICA binding. Cetuximab initiated adaptive immune responses via NK cell-induced DC maturation, which enhanced cross-presentation to CTL specific for EGFR as well as another tumor antigen, MAGE-3. CONCLUSION: Cetuximab-activated NK cells promote DC maturation and CD8(+) T-cell priming, leading to tumor antigen spreading and TH1 cytokine release through "NK-DC cross-talk." FcγRIIIa polymorphism did not predict clinical response to cetuximab but was necessary for NK-DC interaction and mAb-induced cross-presentation. EGFR-specific T cells in cetuximab-treated patients with HNC may contribute to clinical response.


Subject(s)
Antibodies, Monoclonal, Humanized/immunology , Antigens, Neoplasm/immunology , Dendritic Cells/immunology , Head and Neck Neoplasms/immunology , Killer Cells, Natural/immunology , T-Lymphocytes/immunology , ATP Binding Cassette Transporter, Subfamily B, Member 2 , ATP-Binding Cassette Transporters/metabolism , Aged , Antibodies, Monoclonal, Humanized/therapeutic use , Antibody-Dependent Cell Cytotoxicity/immunology , Cell Line, Tumor , Cetuximab , Coculture Techniques , Cross-Priming/immunology , Cytokines/biosynthesis , Dendritic Cells/metabolism , ErbB Receptors/immunology , Female , Genotype , Head and Neck Neoplasms/drug therapy , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/mortality , Head and Neck Neoplasms/pathology , Humans , Interferon-gamma/metabolism , Killer Cells, Natural/metabolism , Lymphocyte Activation , Male , Middle Aged , NK Cell Lectin-Like Receptor Subfamily K/metabolism , Neoplasm Staging , Polymorphism, Genetic , Receptor Cross-Talk/immunology , Receptors, IgG/genetics , Receptors, IgG/immunology , T-Lymphocytes, Cytotoxic/immunology , Th1 Cells/immunology , Th1 Cells/metabolism
4.
Immunol Res ; 50(2-3): 248-54, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21717064

ABSTRACT

Tumor antigen (TA)-targeted monoclonal antibodies (mAb), trastuzumab, cetuximab, panitumumab, and rituximab, have been among the most successful new therapies in the present generation. Clinical activity is observed as a single agent, or in combination with radiotherapy or chemotherapy, against metastatic colorectal cancer, head and neck cancer, breast cancer, and follicular lymphoma. However, the activity is seen only in a minority of patients. Thus, an intense need exists to define the mechanism of action of these immunoactive mAb. Here, we discuss some of the likely immunological events that occur in treated patients: antibody-dependent cellular cytotoxicity (ADCC), cross talk among immune cells including NK cells and dendritic cells (DCs), and generation of TA-specific T lymphocyte responses. We present evidence supporting the induction of "NK:DC cross talk," leading to priming of TA-specific cellular immunity. These observations show that mAb-mediated NK cell activation can be greatly enhanced by the action of stimulatory cytokines and surface molecules on maturing DC and that NK:DC interaction facilitates the recruitment of both NK cells and DC to the tumor site(s). The cooperative, reciprocal stimulatory activity of both NK cells and DC can modulate both the innate immune response in the local tumor microenvironment and the adaptive immune response in secondary lymphoid organs. These events likely contribute to clinical activity, as well as provide a potential biomarker of response to mAb therapy.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antigens, Neoplasm/immunology , Antineoplastic Agents/pharmacology , Dendritic Cells , Killer Cells, Natural , Neoplasms , T-Lymphocytes/immunology , Antibody-Dependent Cell Cytotoxicity/immunology , Dendritic Cells/drug effects , Dendritic Cells/immunology , Dendritic Cells/metabolism , Humans , Immunity, Cellular/immunology , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Neoplasms/immunology , Neoplasms/metabolism
5.
J Immunother ; 33(1): 83-91, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19952953

ABSTRACT

Therapeutic targeting of the epidermal growth factor receptor (EGFR), which is highly overexpressed and correlated with poor prognosis in colorectal and head and neck squamous cell carcinoma (SCCHN), has shown clinical efficacy using the blocking mAbs, cetuximab or panitumumab, but only in 10% to 20% of patients. Clinical responsiveness is correlated with certain Fcgamma receptor genotypes, suggesting immune activity may contribute to therapeutic efficacy. In addition, cetuximab-resistant tumor cells exhibit ubiquitination and degradation of EGFR, which would increase its processing as a tumor antigen for cytotoxic T lymphocyte (CTL) lysis. Thus, T cell-based immunotherapy might enhance the antitumor efficacy of EGFR-specific mAbs, but CTL epitopes are poorly defined. To permit combinatorial EGFR-targeted immunotherapy, we identified a novel immunogenic wild-type sequence peptide, EGFR853-861 and modified its anchor sequence to enhance HLA-A*0201 binding and stimulation of cross-reactive anti-wild-type EGFR853-861-specific CTL. Cross-reactivity was also observed with HER2861-869. EGFR853-861-specific CTL recognition of SCCHN cells was increased by incubation of tumor cells with cetuximab, which led to EGFR degradation. In addition, EGFR853-861-specific CTLs were elevated in the circulation of SCCHN patients as compared with healthy control peripheral blood mononuclear cells. Thus, a novel, immunogenic EGFR-encoded CTL epitope may be incorporated into vaccines and would be useful for combinatorial immunotherapy with EGFR-specific mAbs in cancer patients.


Subject(s)
Epitopes, T-Lymphocyte/immunology , ErbB Receptors/immunology , HLA-A Antigens/immunology , Head and Neck Neoplasms/immunology , Blotting, Western , Cell Line, Tumor , Cell Separation , Flow Cytometry , HLA-A2 Antigen , Humans , T-Lymphocytes, Cytotoxic/immunology
6.
Int J Cancer ; 125(8): 1912-20, 2009 Oct 15.
Article in English | MEDLINE | ID: mdl-19610063

ABSTRACT

The MAGE antigens are frequently expressed cancer vaccine targets. However, quantitative analysis of MAGE expression in upper aerodigestive tract (UADT) tumor cells and its association with T-cell recognition has not been performed, hindering the selection of appropriate candidates for MAGE-specific immunotherapy. Using quantitative RT-PCR (QRT-PCR), we evaluated the expression of MAGE-3/6 in 65 UADT cancers, 48 normal samples from tumor matched sites and 7 HLA-A*0201+ squamous cell carcinoma of the head and neck (SCCHN) cell lines. Expression results were confirmed using Western blot. HLA-A*0201:MAGE-3- (271-279) specific cytotoxic T lymphocytes (MAGE-CTL) from SCCHN patients and healthy donors showed that MAGE-3/6 expression was highly associated with CTL recognition in vitro. On the basis of the MAGE-3/6 expression, we could identify 31 (47%) of the 65 UADT tumors, which appeared to express MAGE-3/6 at levels that correlated with efficient CTL recognition. To confirm that the level of MAGE-3 expression was responsible for CTL recognition, 2 MAGE-3/6 mRNA(high) SCCHN cell lines, PCI-13 and PCI-30, were subjected to MAGE-3/6-specific knockdown. RNAi-transfected cells showed that MAGE expression and MAGE-CTL recognition were significantly reduced. Furthermore, treatment of cells expressing low MAGE-3/6 mRNA with a demethylating agent, 5-aza-2'-deoxycytidine (DAC), increased the expression of MAGE-3/6 and CTL recognition. Thus, using QRT-PCR UADT cancers frequently express MAGE-3/6 at levels sufficient for CTL recognition, supporting the use of a QRT-PCR-based assay for the selection of candidates likely to respond to MAGE-3/6 immunotherapy. Demethylating agents could increase the number of patients amenable for targeting epigenetically modified tumor antigens in vaccine trials.


Subject(s)
Antigens, Neoplasm/metabolism , Carcinoma, Squamous Cell/metabolism , Esophageal Neoplasms/metabolism , Head and Neck Neoplasms/metabolism , Lung Neoplasms/metabolism , Neoplasm Proteins/metabolism , Blotting, Western , Carcinoma, Squamous Cell/pathology , Case-Control Studies , Cytotoxicity, Immunologic , DNA Methylation , Esophageal Neoplasms/pathology , Flow Cytometry , Gene Expression Regulation, Neoplastic , Head and Neck Neoplasms/pathology , Humans , Immunoenzyme Techniques , Interferon-gamma/metabolism , Lung Neoplasms/pathology , Peptide Fragments/metabolism , Prognosis , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/pathology
7.
J Immunother ; 32(5): 465-73, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19609238

ABSTRACT

Ability to cross-present exogenous antigens in the human leukocyte antigen class I pathway is key to the antigen presenting function of mature tumor cell-loaded dendritic cells (DC). Conditions of DC maturation have been shown to be important for DCs ability to produce proinflammatory cytokines and induce T cell effector functions. However, it remains unknown if the different pathways of maturation are associated with modulation of the ability of mature DCs to cross-present tumor antigens (TA). Here, we compare DC matured with 3 clinically relevant cytokine combinations including interleukin (IL)-1 beta, tumor necrosis factor-alpha, IL-6 (termed DC-0), DC-0 cells incubated with prostaglandin-2 (termed DC-0+prostaglandin-2), or DC treated with interferon-gamma, interferon-alpha, tumor necrosis factor-alpha, Poly I:C, and IL1-beta (termed DC-1). We found that these DC vary in their ability to cross-present TA to cytotoxic T lymphocytes (CTL), with the DC-1 cytokine combination being significantly more effective than the other 2. TA cross presentation and CTL priming were strongly correlated with level of expression of the antigen processing machinery components, TAP1 and TAP2, indicating that these components could be used as biomarkers to standardize DC preparations for optimal function. However, the up-regulation of TAP1/TAP2 was not sufficient to explain the enhanced cross-presentation ability of DC-1 cells, as the use of IFN-gamma alone to up-regulate TAP1/TAP2 did not generate DC as effective at cross-presentation as the full DC-1 maturation cytokine combination. These data indicate for the first time that the pathways of DC maturation modulate antigen processing machinery component expression to different extents and that differently matured DC vary in the ability to cross-present TA to human leukocyte antigen class I-restricted CTL.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Antigen Presentation , Carcinoma, Small Cell/immunology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Melanoma/immunology , ATP Binding Cassette Transporter, Subfamily B, Member 2 , ATP Binding Cassette Transporter, Subfamily B, Member 3 , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/immunology , Antigens, Neoplasm/immunology , Antigens, Neoplasm/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/pathology , Cancer Vaccines , Carcinoma, Small Cell/pathology , Cell Differentiation/drug effects , Cell Differentiation/immunology , Cell Line, Tumor , Cytokines/metabolism , Cytokines/pharmacology , Dendritic Cells/pathology , Gene Expression Regulation/drug effects , Gene Expression Regulation/immunology , HLA-A Antigens/metabolism , HLA-A2 Antigen , Humans , Lymphocyte Activation , MART-1 Antigen , Melanoma/pathology , Neoplasm Proteins/immunology , Neoplasm Proteins/metabolism , Peptide Fragments/immunology , Peptide Fragments/metabolism , Prostaglandins/metabolism , Prostaglandins/pharmacology
8.
Oral Oncol ; 45(9): 747-51, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19442565

ABSTRACT

Overall survival for patients with squamous cell carcinoma of the head and neck (SCCHN) has not improved appreciably over the past few decades. Novel therapeutic approaches, such as immunotherapy, are under clinical investigation since the standard treatments are toxic and have not successfully controlled this disease with sufficiently high success rates. Cancer immunotherapy describes various techniques to expand and activate the immune system to control tumor growth in vivo, and clinical evaluation has so far demonstrated low toxicity. Immunotherapy appears to have the most applicability in settings of minimal residual disease and to reduce distant metastases after other therapeutic interventions, and its potential clinical value is now receiving intensive evaluation. Emerging forms of SCCHN immunotherapy involve both the use of monoclonal antibodies (mAb) that target growth factor receptors where immune activation appears to contribute to tumor cell lysis, as well as various forms of active vaccination strategies which activate and direct the patient's cellular immunity against the tumor. This article reviews immunotherapeutic strategies currently in clinical trials or under development for patients with SCCHN.


Subject(s)
Cancer Vaccines/therapeutic use , Carcinoma, Squamous Cell/therapy , Head and Neck Neoplasms/therapy , Immunotherapy/methods , Antibodies, Monoclonal/therapeutic use , Carcinoma, Squamous Cell/immunology , Dendritic Cells/immunology , Head and Neck Neoplasms/immunology , Humans , Immunologic Factors/therapeutic use , Peptides/therapeutic use , Receptors, Growth Factor/drug effects , Treatment Outcome , Vaccines, DNA/therapeutic use
9.
Cancer Immunol Immunother ; 58(11): 1853-64, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19319529

ABSTRACT

Immunotherapy with the EGFR-specific mAb cetuximab is clinically effective in 10-20% of patients with squamous cell carcinoma of the head and neck (SCCHN). Little information is available about the mechanism(s) underlying patients' differential clinical response to cetuximab-based immunotherapy, although this information may contribute to optimizing the design of cetuximab-based immunotherapy. Our understanding of these mechanisms would benefit from the characterization of the variables which influence the extent of cell dependent-lysis of SCCHN cells incubated with cetuximab in vitro. Therefore, in this study we have investigated the role of FcgammaR IIIa-158 genotype expressed by effector NK cells, cetuximab concentration, and EGFR expression level by SCCHN cells in the extent of their in vitro lysis and in the degree of NK cell activation. PBMC or purified CD56+ NK cells genotyped at IIIa codon 158 and SCCHN cell lines expressing different levels of EGFR have been used as effectors and targets, respectively, in antibody dependent cellular cytotoxicity (ADCC) assays. Furthermore, supernatants from ADCC assays were analyzed for cytokine and chemokine levels using multiplexed ELISA. We found that the extent of lysis of SCCHN cells was influenced by the EGFR expression level, cetuximab concentration, and FcgammaR polymorphism. Effector cells expressing the FcgammaR IIIa-158 VV allele were significantly (P < 0.0001) more effective than those expressing FcgammaR IIIa VF and FF [corrected] alleles in mediating lysis of SCCHN cells expressed higher levels of the activation markers CD69 and CD107a, and secreted significantly (P < 0.05) larger amounts of inflammatory cytokines and chemokines. IL-2 or IL-15 treatment increased cetuximab-mediated ADCC by poor binding FcgammaR IIIa 158 FF expressing NK cells. The importance of the FcgammaR IIIa-158 polymorphism in cytotoxicity of SCCHN cells by NK cells supports a potential role for immune activation and may explain patient variability of cetuximab mediated clinical responses. Cellular and secreted immune profiles and FcgammaR genotypes from patients' lymphocytes may provide clinically useful biomarkers of immune activation in cetuximab treated patients.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antineoplastic Agents/pharmacology , Carcinoma, Squamous Cell/drug therapy , ErbB Receptors/physiology , Head and Neck Neoplasms/drug therapy , Killer Cells, Natural/immunology , Polymorphism, Genetic , Receptors, IgG/genetics , Antibodies, Monoclonal, Humanized , Antibody-Dependent Cell Cytotoxicity , Carcinoma, Squamous Cell/immunology , Cell Line, Tumor , Cetuximab , Cytokines/biosynthesis , ErbB Receptors/analysis , ErbB Receptors/genetics , Genotype , Head and Neck Neoplasms/immunology , Humans , Lymphocyte Activation
10.
Curr Oncol Rep ; 11(2): 156-62, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19216848

ABSTRACT

Monoclonal antibodies (mAbs) are now commonly used therapeutic agents in cancer patients. Since US Food and Drug Administration approval of cetuximab for head and neck squamous cell carcinoma, it has been used increasingly in this disease. Several other mAbs also are in development or in clinical -trials. Recently, evidence has accumulated that mAbs induce activation of cellular immunity, including natural killer and T cells and that this may contribute to clinical response. mAbs have been shown to mediate antibody-dependent cellular cytotoxicity, complement-dependent lysis, and activation of tumor antigen-specific T cells. Various patient and tumor factors, such as polymorphisms in Fcgamma receptors expressed by immune cells, activity of T-regulatory cells, and tumor escape through downregulation of antigen-processing machinery in tumor cells, are likely to modulate the immune activation mediated by therapeutic mAbs. Understanding the interplay of these factors is likely to improve the selection of the most appropriate candidates for mAb-based immunotherapy, prediction of clinical response, and our understanding of mechanisms of tumor escape from therapeutic mAbs.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antigens, Neoplasm/immunology , Carcinoma, Squamous Cell/therapy , Head and Neck Neoplasms/therapy , Carcinoma, Squamous Cell/immunology , Complement Pathway, Classical , Head and Neck Neoplasms/immunology , Humans , Immunotherapy , Lymphocyte Activation , Receptors, IgG/genetics , T-Lymphocytes/immunology
11.
Arch Otolaryngol Head Neck Surg ; 133(12): 1277-81, 2007 Dec.
Article in English | MEDLINE | ID: mdl-18086972

ABSTRACT

OBJECTIVE: To determine if the epidermal growth factor receptor (EGFR)-specific monoclonal antibodies (mAbs) cetuximab or panitumumab mediate in vitro immune activation against squamous cell carcinoma of the head and neck (SCCHN) cell lines. DESIGN: In vitro study. SETTING: Basic science research laboratory. INTERVENTION: Squamous cell carcinoma of the head and neck cell lines were treated with the Food and Drug Administration-approved EGFR-specific mAbs cetuximab or panitumumab in the presence or absence of peripheral blood mononuclear cells from healthy donors. MAIN OUTCOME MEASURES: Cetuximab and panitumumab were compared in terms of their cytotoxic effects, ability to induce apoptosis, bind to EGFR, and block phosphorylation of this receptor in SCCHN cell lines. RESULTS: We demonstrate that both cetuximab and panitumumab have similar levels of EGFR binding, induction of apoptosis, cell lysis, and inhibition of phospho-EGFR in SCCHN cell lines, suggesting similar direct effects. However, neither of these mAbs demonstrated in vitro antitumor activity when used alone. In contrast, in the presence of peripheral blood lymphocytes, either of them can mediate antibody-dependent cell cytotoxicity in vitro when used in doses similar to those found in patients receiving them clinically. CONCLUSION: We propose that antibody-dependent cell cytotoxicity may constitute an important antitumor mechanism that could contribute to overall clinical effectiveness of EGFR-specific antibodies.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/therapeutic use , Carcinoma, Squamous Cell/drug therapy , ErbB Receptors/immunology , Head and Neck Neoplasms/drug therapy , Immunity, Cellular/drug effects , Antibodies, Monoclonal, Humanized , Apoptosis/drug effects , Carcinoma, Squamous Cell/immunology , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Cetuximab , Head and Neck Neoplasms/immunology , Head and Neck Neoplasms/pathology , Humans
12.
J Immunol ; 176(6): 3402-9, 2006 Mar 15.
Article in English | MEDLINE | ID: mdl-16517708

ABSTRACT

Squamous cell carcinoma of the head and neck (SCCHN) cells are poorly recognized in vitro by CTL despite expressing the restricting HLA class I allele and the targeted tumor Ag (TA). Several lines of evidence indicate that the lack of SCCHN cell recognition by CTL reflects defects in targeted TA peptide presentation by HLA class I Ag to CTL because of Ag-processing machinery (APM) dysfunction. First, lack of recognition of SCCHN cells by CTL is associated with marked down-regulation of the IFN-gamma-inducible APM components low-m.w. protein 2, TAP1, TAP2, and tapasin. Second, SCCHN cell recognition by CTL is restored by pulsing cells with exogenous targeted TA peptide. Third, the restoration of CTL recognition following incubation of SCCHN cells with IFN-gamma is associated with a significant (p = 0.001) up-regulation of the APM components TAP1, TAP2, and tapasin. Lastly, and most conclusively, SCCHN cell recognition by CTL is restored by transfection with wild-type TAP1 cDNA. Our findings may explain the association between APM component down-regulation and poor clinical course of the disease in SCCHN. Furthermore, the regulatory nature of the APM defects in SCCHN cells suggests that intralesional administration of IFN-gamma may have a beneficial effect on the clinical course of the disease and on T cell-based immunotherapy of SCCHN by restoring SCCHN cell recognition by CTL.


Subject(s)
Antigen Presentation/immunology , Carcinoma, Squamous Cell/immunology , Head and Neck Neoplasms/immunology , T-Lymphocytes, Cytotoxic/immunology , Antigens, Neoplasm/immunology , Antigens, Neoplasm/metabolism , Carcinoma, Squamous Cell/metabolism , Cell Adhesion Molecules/metabolism , Cell Line, Tumor , Gene Expression Regulation , HLA-A2 Antigen/metabolism , Head and Neck Neoplasms/metabolism , Histocompatibility Antigens Class I/immunology , Humans , Interferon-gamma/pharmacology , Neoplasm Proteins/metabolism , Receptor, ErbB-2/metabolism , T-Lymphocytes, Cytotoxic/drug effects , T-Lymphocytes, Cytotoxic/metabolism , Transfection
13.
Cancer Res ; 65(23): 11146-55, 2005 Dec 01.
Article in English | MEDLINE | ID: mdl-16322265

ABSTRACT

Human papillomavirus (HPV)-associated squamous cell carcinoma of the head and neck (SCCHN) seems to be a suitable target for cancer vaccination. HPV-encoded oncogenic proteins, such as E7, are promising tumor-specific antigens and are obligatory for tumor growth. Because few immunologic studies have analyzed the endogenous HPV-specific immune response in this subset of SCCHN patients, we studied T-cell frequencies against HPV-16 E7(11-20) or E7(86-93) in tumor-bearing, human leukocyte antigen (HLA)-A*0201+ SCCHN patients, whose tumors were either HPV-16+ or HPV-16-. In HPV-16+ SCCHN patients, frequencies of T cells against either peptide were significantly elevated (P < 0.005) compared with HPV-16- patients or healthy volunteers. Tetramer+ T cells showed evidence of terminally differentiated phenotype (CD45RA+CCR7-) and an elevated level of CD107a staining for degranulation. Despite detectable expression of the restricting HLA class I allele, HLA-A*0201-E7(11-20)- or HLA-A*0201-E7(86-93)-specific CTL obtained by in vitro stimulation of healthy donor peripheral blood mononuclear cells only recognize a naturally HPV-16-transformed, HLA-A*0201+ SCCHN cell line after pretreatment with IFN-gamma. This cell line had little or no expression of LMP2, TAP1, and tapasin, critical components of the HLA class I antigen-processing machinery, which were up-regulated by IFN-gamma treatment. Immunohistochemistry of HPV-16+ SCCHN tumors showed that these antigen-processing machinery components are down-regulated in tumors in vivo compared with adjacent normal squamous epithelium. Thus, immunity to HPV-16 E7 is associated with the presence of HPV-16 infection and presentation of E7-derived peptides on SCCHN cells, which show evidence of immune escape. These findings support further development of E7-specific immunotherapy and strategies for up-regulation of antigen-processing machinery components in HPV-associated SCCHN.


Subject(s)
Carcinoma, Squamous Cell/immunology , Carcinoma, Squamous Cell/virology , Head and Neck Neoplasms/immunology , Head and Neck Neoplasms/virology , Oncogene Proteins, Viral/immunology , Animals , Carcinoma, Squamous Cell/therapy , Dendritic Cells/immunology , Epitopes, T-Lymphocyte/immunology , HLA-A Antigens/immunology , HLA-A2 Antigen , Head and Neck Neoplasms/therapy , Human papillomavirus 16/immunology , Humans , Immunotherapy, Adoptive/methods , Interferon-gamma/pharmacology , Male , Mice , Mice, Transgenic , Papillomavirus E7 Proteins , Papillomavirus Infections/complications , Papillomavirus Infections/immunology , T-Lymphocytes, Cytotoxic/immunology
14.
Eur J Cancer ; 41(5): 807-15, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15763658

ABSTRACT

Uncertainty regarding the causality of human papillomaviruses (HPVs) in squamous cell carcinoma of the head and neck (SCCHN) necessitates better in vitro models. We carried out molecular analyses of a novel, naturally HPV-16-transformed SCCHN cell line (UPCI:SCC090) and show high copy number of HPV-16 DNA, present in a head to tail, tandemly repeated integrated state. Sequence analysis of the HPV-16 long control region (LCR) in UPCI:SCC090 revealed a deletion of 163 bp, removing a portion of the enhancer sequence, including the binding sites for the transcription factors YY1 and NF1. The E6 and E7 oncogenes of HPV-16 are expressed at high levels in this cell lines, as determined by quantitative reverse transcriptase-polymerase chain reaction (RT-PCR). UPCI:SCC090 contains wild-type tumour suppressor TP53 gene, and undetectable p53 protein, except after treatment with cisplatin, specific proteasome inhibitors or by E6 RNA interference, suggesting E6-dependent degradation of p53 in this cell line. The results of our studies are consistent with a causative role of HPV-16 in the pathogenesis of SCCHN.


Subject(s)
Carcinoma, Squamous Cell/virology , Head and Neck Neoplasms/virology , Papillomaviridae , Papillomavirus Infections/complications , Blotting, Southern , Cell Line, Tumor , DNA, Viral/analysis , Humans , Models, Biological , RNA, Viral/analysis , Reverse Transcriptase Polymerase Chain Reaction/methods , Transcription, Genetic , Tumor Suppressor Protein p53/metabolism
15.
Life Sci ; 76(3): 263-80, 2004 Dec 03.
Article in English | MEDLINE | ID: mdl-15531379

ABSTRACT

Though the nicotinic acetylcholine receptor (nAChR) subunits alpha9 and alpha 10 have been thoroughly characterized within hair cells of the organ of Corti in the inner ear, prior studies have shown that they are also expressed in lymphocytes. In this report, we sought to more definitively characterize the nAChR subunits alpha9 and alpha10 within various populations of human lymphocytes. Using a combination of techniques, including RT-PCR, single-cell RT-PCR, Northern and western blot analysis, and immunofluorescence, expression of both alpha9 and alpha 10 was demonstrated in purified populations of T-cells (CD3+, CD4+, CD8+ and the Jurkat, MT2 and CEM T-cell lines) and B-cells (CD19+, CD80+ and EBV-immortalized B-cells). Single-lymphocyte recording techniques failed to identify an ionic current in response to applied acetylcholine in either T-cells or B-cells. These results clearly demonstrate the presence of these nicotinic receptor subunits within several populations of human lymphocytes, implicating their role in the immune response. However, a lack of demonstrated response to applied acetylcholine using standard single-cell recording techniques suggests a physiology different than that seen in hair cells of the inner ear.


Subject(s)
B-Lymphocyte Subsets/metabolism , Receptors, Nicotinic/metabolism , T-Lymphocyte Subsets/metabolism , Acetylcholine/pharmacology , B-Lymphocyte Subsets/drug effects , Cell Separation , Flow Cytometry , Humans , Jurkat Cells , RNA, Messenger/metabolism , Receptors, Nicotinic/analysis , Receptors, Nicotinic/genetics , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocyte Subsets/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL