Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
Bioact Mater ; 34: 422-435, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38282968

ABSTRACT

Cell membrane-derived nanoparticles (NPs) have recently gained popularity due to their desirable features in drug delivery such as mimicking properties of native cells, impeding systemic clearance, and altering foreign body responses. Besides NP technology, adoptive immunotherapy has emerged due to its promise in cancer specificity and therapeutic efficacy. In this research, we developed a biomimetic drug carrier based on chimeric antigen receptor (CAR) transduced T-cell membranes. For that purpose, anti-HER2 CAR-T cells were engineered via lentiviral transduction of anti-HER2 CAR coding lentiviral plasmids. Anti-HER2 CAR-T cells were characterized by their specific activities against the HER2 antigen and used for cell membrane extraction. Anti-cancer drug Cisplatin-loaded poly (D, l-lactide-co-glycolic acid) (PLGA) NPs were coated with anti-human epidermal growth factor receptor 2 (HER2)-specific CAR engineered T-cell membranes. Anti-HER2 CAR-T-cell membrane-coated PLGA NPs (CAR-T-MNPs) were characterized and confirmed via fluorescent microscopy and flow cytometry. Membrane-coated NPs showed a sustained drug release over the course of 21 days in physiological conditions. Cisplatin-loaded CAR-T-MNPs also inhibited the growth of multiple HER2+ cancer cells in vitro. In addition, in vitro uptake studies revealed that CAR-T-MNPs showed an increased uptake by A549 cells. These results were also confirmed via in vivo biodistribution and therapeutic studies using a subcutaneous lung cancer model in nude mice. CAR-T-MNPs localized preferentially at tumor areas compared to those of other studied groups and consisted of a significant reduction in tumor growth in tumor-bearing mice. In Conclusion, the new CAR modified cell membrane-coated NP drug-delivery platform has demonstrated its efficacy both in vitro and in vivo. Therefore, CAR engineered membrane-coated NP system could be a promising cell-mimicking drug carrier that could improve therapeutic outcomes of lung cancer treatments.

2.
Article in English | MEDLINE | ID: mdl-32850765

ABSTRACT

Melanoma is one of the most aggressive skin cancers, and the American Cancer Society reports that every hour, one person dies from melanoma. While there are a number of treatments currently available for melanoma (e.g., surgery, chemotherapy, immunotherapy, and radiation therapy), they face several problems including inadequate response rates, high toxicity, severe side effects due to non-specific targeting of anti-cancer drugs, and the development of multidrug resistance during prolonged treatment. To improve chemo-drug therapeutic efficiency and overcome these mentioned limitations, a multifunctional nanoparticle has been developed to effectively target and treat melanoma. Specifically, poly (lactic-co-glycolic acid) (PLGA) nanoparticles (NPs) were coated with a cellular membrane derived from the T cell hybridoma, 19LF6 endowed with a melanoma-specific anti-gp100/HLA-A2 T-cell receptor (TCR) and loaded with an FDA-approved melanoma chemotherapeutic drug Trametinib. T-cell membrane camouflaged Trametinib loaded PLGA NPs displayed high stability, hemo- and cyto-compatibility. They also demonstrated membrane coating dependent drug release profiles with the most sustained release from the NPs proportional with the highest amount of membrane used. 19LF6 membrane-coated NPs produced a threefold increase in cellular uptake toward the melanoma cell line in vitro compared to that of the bare nanoparticle. Moreover, the binding kinetics and cellular uptake of these particles were shown to be membrane/TCR concentration-dependent. The in vitro cancer killing efficiencies of these NPs were significantly higher compared to other NP groups and aligned with binding and uptake characteristics. Particles with the higher membrane content (greater anti-gp100 TCR content) were shown to be more effective when compared to the free drug and negative controls. In vivo biodistribution studies displayed the theragnostic capabilities of these NPs with more than a twofold increase in the tumor retention compared to the uncoated and non-specific membrane coated groups. Based on these studies, these T-cell membrane coated NPs emerge as a potential theragnostic carrier for imaging and therapy applications associated with melanoma.

3.
MAbs ; 9(4): 603-614, 2017.
Article in English | MEDLINE | ID: mdl-28273004

ABSTRACT

The currently marketed antibody-drug conjugates (ADC) destabilize microtubule assembly in cancer cells and initiate apoptosis in patients. However, few tumor antigens (TA) are expressed at high densities on cancer lesions, potentially minimizing the therapeutic index of current ADC regimens. The peptide/human leukocyte antigen (HLA) complex can be specifically targeted by therapeutic antibodies (designated T cell receptor [TCR]-like antibodies) and adequately distinguish malignant cells, but has not been the focus of ADC development. We analyzed the killing potential of TCR-like ADCs when cross-linked to the DNA alkylating compound duocarmycin. Our data comprise proof-of-principle results that TCR-like ADCs mediate potent tumor cytotoxicity, particularly under common scenarios of low TA/HLA density, and support their continued development alongside agents that disrupt DNA replication. Additionally, TCR-like antibody ligand binding appears to play an important role in ADC functionality and should be addressed during therapy development to avoid binding patterns that negate ADC killing efficacy.


Subject(s)
Antibodies, Neoplasm/pharmacology , Drug Delivery Systems/methods , HLA Antigens/immunology , Indoles/pharmacology , Neoplasms/drug therapy , Peptides/immunology , Receptors, Antigen, T-Cell , Animals , Cell Line, Tumor , Duocarmycins , Humans , Mice , Neoplasms/immunology , Neoplasms/pathology , Pyrrolidinones/pharmacology
4.
J Immunol ; 196(10): 4263-73, 2016 05 15.
Article in English | MEDLINE | ID: mdl-27183642

ABSTRACT

Class I HLA molecules mark infected cells for immune targeting by presenting pathogen-encoded peptides on the cell surface. Characterization of viral peptides unique to infected cells is important for understanding CD8(+) T cell responses and for the development of T cell-based immunotherapies. Having previously reported a series of West Nile virus (WNV) epitopes that are naturally presented by HLA-A*02:01, in this study we generated TCR mimic (TCRm) mAbs to three of these peptide/HLA complexes-the immunodominant SVG9 (E protein), the subdominant SLF9 (NS4B protein), and the immunorecessive YTM9 (NS3 protein)-and used these TCRm mAbs to stain WNV-infected cell lines and primary APCs. TCRm staining of WNV-infected cells demonstrated that the immunorecessive YTM9 appeared several hours earlier and at 5- to 10-fold greater density than the more immunogenic SLF9 and SVG9 ligands, respectively. Moreover, staining following inhibition of the TAP demonstrated that all three viral ligands were presented in a TAP-dependent manner despite originating from different cellular compartments. To our knowledge, this study represents the first use of TCRm mAbs to define the kinetics and magnitude of HLA presentation for a series of epitopes encoded by one virus, and the results depict a pattern whereby individual epitopes differ considerably in abundance and availability. The observations that immunodominant ligands can be found at lower levels and at later time points after infection suggest that a reevaluation of the factors that combine to shape T cell reactivity may be warranted.


Subject(s)
Antigen Presentation , CD8-Positive T-Lymphocytes/immunology , Epitopes, T-Lymphocyte/immunology , Immunodominant Epitopes/immunology , West Nile virus/immunology , Animals , Antibodies, Monoclonal/immunology , Antibody Specificity , Cell Line, Tumor , Dendritic Cells/virology , Female , HLA-A Antigens/immunology , Humans , Mice , Mice, Inbred BALB C , Receptors, Antigen, T-Cell/immunology
5.
Mol Cancer Ther ; 15(2): 313-22, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26719579

ABSTRACT

T cells recognize cancer cells via HLA/peptide complexes, and when disease overtakes these immune mechanisms, immunotherapy can exogenously target these same HLA/peptide surface markers. We previously identified an HLA-A2-presented peptide derived from macrophage migration inhibitory factor (MIF) and generated antibody RL21A against this HLA-A2/MIF complex. The objective of the current study was to assess the potential for targeting the HLA-A2/MIF complex in ovarian cancer. First, MIF peptide FLSELTQQL was eluted from the HLA-A2 of the human cancerous ovarian cell lines SKOV3, A2780, OV90, and FHIOSE118hi and detected by mass spectrometry. By flow cytometry, RL21A was shown to specifically stain these four cell lines in the context of HLA-A2. Next, partially matched HLA-A*02:01+ ovarian cancer (n = 27) and normal fallopian tube (n = 24) tissues were stained with RL21A by immunohistochemistry to assess differential HLA-A2/MIF complex expression. Ovarian tumor tissues revealed significantly increased RL21A staining compared with normal fallopian tube epithelium (P < 0.0001), with minimal staining of normal stroma and blood vessels (P < 0.0001 and P < 0.001 compared with tumor cells) suggesting a therapeutic window. We then demonstrated the anticancer activity of toxin-bound RL21A via the dose-dependent killing of ovarian cancer cells. In summary, MIF-derived peptide FLSELTQQL is HLA-A2-presented and recognized by RL21A on ovarian cancer cell lines and patient tumor tissues, and targeting of this HLA-A2/MIF complex with toxin-bound RL21A can induce ovarian cancer cell death. These results suggest that the HLA-A2/MIF complex should be further explored as a cell-surface target for ovarian cancer immunotherapy.


Subject(s)
Biomarkers, Tumor/metabolism , HLA-A2 Antigen/metabolism , Intramolecular Oxidoreductases/metabolism , Macrophage Migration-Inhibitory Factors/metabolism , Ovarian Neoplasms/metabolism , Peptides/metabolism , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/therapeutic use , Antibody Affinity , Antibody Specificity , Cell Line, Tumor , Female , HLA-A2 Antigen/immunology , Humans , Ovarian Neoplasms/pathology
6.
J Vis Exp ; (97)2015 Mar 24.
Article in English | MEDLINE | ID: mdl-25867039

ABSTRACT

According to the American Cancer Society, more than 200,000 women will be diagnosed with invasive breast cancer each year and approximately 40,000 will die from the disease. The human leukocyte antigen (HLA) class I samples peptides derived from proteasomal degradation of cellular proteins and presents these fragments on the cell surface for interrogation by circulating cytotoxic T lymphocytes (CTL). Generation of T-cell receptor mimic (TCRm) monoclonal antibodies (mAbs) which recognize breast cancer specific peptide/HLA-A*02:01 complexes such as those derived from macrophage migration inhibitory factor (MIF19-27) and NY-ESO-1157-165 enable detection and destruction of breast cancer cells in the absence of an effective anti-tumor CTL response. Intact class I HLA/peptide complexes are shed by breast cancer cells and represent potentially relevant cancer biomarkers. In this work, a breakthrough biomarker screening system for cancer diagnostics incorporating T-cell receptor mimic monoclonal antibodies combined with a novel, label-free biosensor utilizing guided-mode resonance (GMR) sensor technology is presented. Detection of shed MIF/HLA-A*02:01 complexes in MDA-MB-231 cell supernatants, spiked human serum, and patient plasma is demonstrated. The impact of this work could revolutionize personalized medicine through development of companion disease diagnostics for targeted immunotherapies.


Subject(s)
Biomarkers, Tumor/analysis , Biosensing Techniques/methods , Breast Neoplasms/chemistry , HLA-A2 Antigen/analysis , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/immunology , Biomarkers, Tumor/immunology , Breast Neoplasms/immunology , Breast Neoplasms/pathology , Cell Line, Tumor , Female , HLA-A2 Antigen/chemistry , HLA-A2 Antigen/immunology , Humans , Intramolecular Oxidoreductases/chemistry , Intramolecular Oxidoreductases/immunology , Macrophage Migration-Inhibitory Factors/chemistry , Macrophage Migration-Inhibitory Factors/immunology , Peptide Fragments/chemistry , Peptide Fragments/immunology , T-Lymphocytes, Cytotoxic/immunology
7.
Eur J Immunol ; 44(7): 1936-46, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24723377

ABSTRACT

We used a newly generated T-cell receptor mimic monoclonal antibody (TCRm MAb) that recognizes a known nonself immunodominant peptide epitope from West Nile virus (WNV) NS4B protein to investigate epitope presentation after virus infection in C57BL/6 mice. Previous studies suggested that peptides of different length, either SSVWNATTAI (10-mer) or SSVWNATTA (9-mer) in complex with class I MHC antigen H-2D(b) , were immunodominant after WNV infection. Our data establish that both peptides are presented on the cell surface after WNV infection and that CD8(+) T cells can detect 10- and 9-mer length variants similarly. This result varies from the idea that a given T-cell receptor (TCR) prefers a single peptide length bound to its cognate class I MHC. In separate WNV infection studies with the TCRm MAb, we show that in vivo the 10-mer was presented on the surface of uninfected and infected CD8α(+) CD11c(+) dendritic cells, which suggests the use of direct and cross-presentation pathways. In contrast, CD11b(+) CD11c(-) cells bound the TCRm MAb only when they were infected. Our study demonstrates that TCR recognition of peptides is not limited to certain peptide lengths and that TCRm MAbs can be used to dissect the cell-type specific mechanisms of antigen presentation in vivo.


Subject(s)
Dendritic Cells/immunology , Immunodominant Epitopes , Receptors, Antigen, T-Cell/physiology , West Nile virus/immunology , Animals , CD11b Antigen/analysis , CD11c Antigen/analysis , CD8-Positive T-Lymphocytes/immunology , Mice , Mice, Inbred C57BL , Viral Nonstructural Proteins/immunology
8.
J Natl Cancer Inst ; 105(3): 202-18, 2013 Feb 06.
Article in English | MEDLINE | ID: mdl-23300219

ABSTRACT

BACKGROUND: Applications of trastuzumab are limited to breast cancer patients with high Her2-expressing tumors. We developed a T-cell receptor mimic (TCRm) monoclonal antibody (hereafter called RL1B) that targets the Her2-E75 peptide (residues 369-377)-HLA-A2 complex and examined its effects in Her2-expressing cancer cells. METHODS: RL1B binding affinity was determined by surface plasmon resonance and specificity was demonstrated using Her2 antigen-positive and negative tumor cell lines. Immunohistochemistry was used to assess binding to frozen sections of human carcinomas (n = 3). Antitumor activity mediated by RL1B and trastuzumab against Her2(+) tumor cell lines was evaluated using the WST-1 cell viability assay and caspase-3 and poly(ADP-ribose) polymerase cleavage assays. A xenograft mouse model (n = 6 per group) was used to assess RL1B antitumor activity. Mechanisms of RL1B-mediated cytotoxicity were evaluated with confocal microscopy, flow cytometry, and histology. All statistical tests were two-sided. RESULTS: RL1B bound with high specificity and affinity to the E75 peptide-HLA-A2 complex in all Her2(+) and HLA-A2(+) cancer cell lines and human carcinomas. Compared with control antibody, RL1B suppressed growth of low Her2-expressing breast tumors in mice (mean volume, RL1B vs control = 241 mm(3) vs 1531 mm(3); P = .0109) and statistically significantly increased mouse survival (P = .0098). It reduced viability compared to control monoclonal antibody-treated cells and statistically significantly increased caspase 3 activation of all Her2(+) carcinoma cell lines tested, whereas trastuzumab induced apoptosis only in high Her2-expressing cancer cells. Mechanisms of RL1B cytotoxicity were associated with antibody internalization and intracellular signaling. CONCLUSION: The TCRm RL1B could be a new approach to immunotherapy of Her2-expressing malignancies.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacology , Antibodies, Monoclonal/pharmacology , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , HLA-A2 Antigen/drug effects , Intracellular Signaling Peptides and Proteins/drug effects , Neoplasms/drug therapy , Oncogene Proteins, Fusion/antagonists & inhibitors , Receptor, ErbB-2/antagonists & inhibitors , Receptors, Antigen, T-Cell/agonists , Animals , Breast Neoplasms/drug therapy , Carcinoma/drug therapy , Caspase 3/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Colorectal Neoplasms/drug therapy , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Frozen Sections , Gene Expression Regulation, Neoplastic/drug effects , HLA-A2 Antigen/metabolism , Humans , Immunohistochemistry , Immunosorbent Techniques , Intracellular Signaling Peptides and Proteins/metabolism , Mice , Mice, Nude , Microscopy, Confocal , Microscopy, Fluorescence , Nanoparticles , Neoplasms/metabolism , Oncogene Proteins, Fusion/metabolism , Pancreatic Neoplasms/drug therapy , Peptide Fragments/drug effects , Peptide Fragments/metabolism , Poly(ADP-ribose) Polymerases/metabolism , Receptor, ErbB-2/metabolism , Signal Transduction/drug effects , Trastuzumab , Up-Regulation , Xenograft Model Antitumor Assays
9.
Int Rev Immunol ; 30(5-6): 328-40, 2011.
Article in English | MEDLINE | ID: mdl-22053972

ABSTRACT

The human leukocyte antigen (HLA; also called major histocompatibility, or MHC) class I system presents peptides that distinguish healthy from diseased cells. Therefore, the discovery of peptide/MHC class I markers can provide highly specific targets for immunotherapy. Over the course of almost two decades, various strategies have been used, with mixed success, to produce antibodies that have recognition specificity for unique peptide/MHC class I complexes that mark infected and cancerous cells. Using these antibody reagents, novel peptide/MHC class I targets have been directly validated on diseased cells and new insight has been gained into the mechanisms of antigen presentation. More recently, these antibodies have shown promise for clinical applications such as therapeutic targeting of cancerous and infected cells and diagnosis and imaging of diseased cells. In this review, the authors comprehensively describe the methods used to identify disease-specific peptide/MHC class I epitopes and generate antibodies to these markers. Finally, they offer several examples that illustrate the promise of using these antibodies as anti-cancer agents.


Subject(s)
Antibodies, Monoclonal/immunology , Epitopes, T-Lymphocyte/immunology , Histocompatibility Antigens Class I/immunology , Peptides/immunology , Receptors, Antigen, T-Cell/immunology , Animals , Cell Line , Cell Line, Tumor , Histocompatibility Antigens Class I/metabolism , Humans , Immunotherapy/methods , Peptides/metabolism , Receptors, Antigen, T-Cell/metabolism
10.
Int Rev Immunol ; 30(5-6): 312-27, 2011.
Article in English | MEDLINE | ID: mdl-22053971

ABSTRACT

The development of immunotherapies offers significant promise for clinical applications in cancer and infectious diseases. Here the authors describe a novel, integrated approach to immunotherapy that combines novel technologies to discover and target disease-specific peptide/HLA class I complexes. This unique class of markers makes the entire proteome accessible to antibody reagents and offers unsurpassed specificity for targeting cancerous and infected cells. Arm one of the three-armed approach uses an innovative technology for the efficient, direct discovery of new peptide/HLA class I markers. Arm two applies a powerful and inventive strategy to generate T-cell receptor mimics (TCRms), which are antibodies with exquisite binding specificity for peptide/HLA class I markers, and uses TCRms to validate the specific expression of markers on cancerous and infected cells. The third arm uses TCRms to target and kill diseased cells with high sensitivity and specificity. In summary, the combination of two pioneering technologies expands the repertoire of disease-specific markers that can be targeted by therapeutic antibodies and enables a powerful, integrated approach to HLA-based immunotherapy.


Subject(s)
Antibodies, Monoclonal/immunology , Histocompatibility Antigens Class I/metabolism , Immunotherapy/methods , Neoplasms/immunology , Neoplasms/therapy , Receptors, Antigen, T-Cell/metabolism , Antibodies, Monoclonal/therapeutic use , Antibody Specificity , Cell Line, Tumor , Humans , Molecular Mimicry , Peptides/metabolism
11.
J Immunol ; 186(5): 3265-76, 2011 Mar 01.
Article in English | MEDLINE | ID: mdl-21282517

ABSTRACT

mAbs that recognize peptides presented on the cell surface by MHC class I molecules are potential therapeutic agents for cancer therapy. We have previously demonstrated that these Abs, which we termed TCR mimic mAbs (TCRm), reduce tumor growth in models of breast carcinoma. However, mechanisms of TCRm-mediated tumor growth reduction remain largely unknown. In this study, we report that these Abs, in contrast to several mAbs used currently in the clinic, destroy tumor cells independently of immune effector mechanisms such as Ab-dependent cellular cytotoxicity (ADCC) and complement-dependent cytotoxicity (CDC). We found that TCRm-mediated apoptosis of tumor cells was associated with selective and specific binding of these Abs to peptide/HLA class I complexes, which triggered the activation of JNK and intrinsic caspase pathways. This signaling was accompanied by the release of mitochondrial cytochrome c and apoptosis-inducing factor. TCRm-induced apoptosis in tumor cells was completely inhibited by soluble MHC tetramers loaded with relevant peptide as well as with inhibitors for JNK and caspases. Furthermore, mAbs targeting MHC class I, independent of the peptide bound by HLA, did not stimulate apoptosis, suggesting that the Ab-binding site on the MHC/peptide complex determines cytotoxicity. This study suggests the existence of mechanisms, in addition to ADCC and CDC, through which these therapeutic Abs destroy tumor cells. These mechanisms would appear to be of particular importance in severely immunocompromised patients with advanced neoplastic disease, since immune cell-mediated killing of tumor cells through ADCC and CDC is substantially limited in these individuals.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antibodies, Neoplasm/therapeutic use , Apoptosis/immunology , Molecular Mimicry/immunology , Receptors, Antigen, T-Cell/therapeutic use , Xenograft Model Antitumor Assays/methods , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/metabolism , Antibodies, Neoplasm/administration & dosage , Antibodies, Neoplasm/metabolism , Cell Line, Tumor , Female , HLA-A2 Antigen/metabolism , Humans , Leukemia, Monocytic, Acute/immunology , Leukemia, Monocytic, Acute/pathology , Leukemia, Monocytic, Acute/therapy , Mammary Neoplasms, Experimental/immunology , Mammary Neoplasms, Experimental/pathology , Mammary Neoplasms, Experimental/therapy , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Melanoma, Experimental/therapy , Mice , Mice, Nude , Protein Binding/immunology , Receptors, Antigen, T-Cell/metabolism
12.
J Cell Physiol ; 225(3): 664-72, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20506235

ABSTRACT

Organ-specific vascular targeting, for example, to the blood-brain barrier, requires the identification of unique molecular addresses on a subset of endothelial cells. The present study describes a crucial step towards tapping the exquisite specificity of the peptide/HLA class I system for this goal. We utilized a novel T-cell receptor (TCR) mimic antibody of high affinity and specificity, which is restricted by HLA-A2 and has been generated to recognize a peptide epitope derived from p68 RNA helicase (YLLPAIVHI). The parent protein is highly expressed by brain endothelial cells. Flow cytometry and confocal imaging showed that the antibody binds to HLA-A2-positive human brain-derived endothelial cells, both immortalized hCMEC/D3 cells and primary cells. The TCR mimic antibody undergoes internalization into vesicles, where significant colocalization occurs with the early endosomal marker EEA-1, but barely with caveolin-1. To our knowledge internalization of neither MHC class I protein nor TCR mimics by brain endothelial cells has been previously observed. Knock down of p68 protein expression by siRNA reduced the presentation of YLLPAIVHI-peptide/HLA-A2 complexes on the cell membrane by half as measured by flow cytometry 48 h later. We also found that brain endothelial cells isolated from HLA-A2 transgenic mouse strains express the A2 transgene, and brain endothelial cells of one of these strains also present YLLPAIVHI-peptide/HLA-A2, making these mouse strains suitable models for studying TCR mimic antibodies in vivo. In conclusion, these data strongly support the notion that TCR mimic antibodies could be a new class of therapeutic targeting agents in a wide variety of diseases.


Subject(s)
Antibodies, Monoclonal , Blood-Brain Barrier/immunology , Brain/blood supply , DEAD-box RNA Helicases/immunology , Endothelial Cells/immunology , HLA-A2 Antigen/immunology , Immunomagnetic Separation , Receptors, Antigen, T-Cell/immunology , Animals , Antibodies, Monoclonal/metabolism , Antibody Affinity , Antibody Specificity , Blood-Brain Barrier/cytology , Blood-Brain Barrier/metabolism , Caveolin 1/metabolism , Cells, Cultured , DEAD-box RNA Helicases/genetics , DEAD-box RNA Helicases/metabolism , Endocytosis , Endosomes/immunology , Endosomes/metabolism , Endothelial Cells/metabolism , Flow Cytometry , HLA-A2 Antigen/genetics , Humans , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Transgenic , Microscopy, Confocal , Molecular Mimicry , Platelet Endothelial Cell Adhesion Molecule-1/immunology , RNA Interference , Receptors, Antigen, T-Cell/metabolism , Vesicular Transport Proteins/metabolism
13.
J Immunol ; 184(8): 4423-30, 2010 Apr 15.
Article in English | MEDLINE | ID: mdl-20212098

ABSTRACT

The generation of a robust CD8(+) T cell response is an ongoing challenge for the development of DNA vaccines. One problem encountered with classical DNA plasmid immunization is that peptides produced are noncovalently and transiently associated with MHC class I molecules and thus may not durably stimulate CD8(+) T cell responses. To address this and enhance the expression and presentation of the antigenic peptide/MHC complexes, we generated single-chain trimers (SCTs) composed of a single polypeptide chain with a linear composition of antigenic peptide, beta(2)-microglobulin, and H chain connected by flexible linkers. In this study, we test whether the preassembled nature of the SCT makes them effective for eliciting protective CD8(+) T cell responses against pathogens. A DNA plasmid was constructed encoding an SCT incorporating the human MHC class I molecule HLA-A2 and the immunodominant peptide SVG9 derived from the envelope protein of West Nile virus (WNV). HLA-A2 transgenic mice vaccinated with the DNA encoding the SVG9/HLA-A2 SCT generated a robust epitope-specific CD8(+) T cell response and showed enhanced survival rate and lower viral burden in the brain after lethal WNV challenge. Inclusion of a CD4(+) Th cell epitope within the SCT did not increase the frequency of SVG9-specific CD8(+) T cells, but did enhance protection against WNV challenge. Overall, these findings demonstrate that the SCT platform can induce protective CD8(+) T cell responses against lethal virus infection and may be paired with immunogens that elicit robust neutralizing Ab responses to generate vaccines that optimally activate all facets of adaptive immunity.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Epitopes, T-Lymphocyte/immunology , HLA-A2 Antigen/immunology , Immunodominant Epitopes/immunology , Vaccines, Subunit/immunology , West Nile Fever/prevention & control , West Nile Virus Vaccines/immunology , West Nile virus/immunology , Animals , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/virology , Cells, Cultured , Cytotoxicity, Immunologic/genetics , Disease Models, Animal , Epitopes, T-Lymphocyte/administration & dosage , Epitopes, T-Lymphocyte/genetics , HLA-A2 Antigen/administration & dosage , HLA-A2 Antigen/genetics , Humans , Immunodominant Epitopes/administration & dosage , Immunodominant Epitopes/genetics , Mice , Mice, Transgenic , Protein Binding/genetics , Protein Binding/immunology , Survival Analysis , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/metabolism , T-Lymphocytes, Helper-Inducer/virology , Vaccines, DNA/administration & dosage , Vaccines, DNA/genetics , Vaccines, DNA/immunology , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/genetics , Viral Load/genetics , Viral Load/immunology , West Nile Fever/immunology , West Nile Fever/mortality , West Nile Virus Vaccines/administration & dosage , West Nile Virus Vaccines/genetics , West Nile virus/pathogenicity
14.
J Immunol ; 184(4): 2156-65, 2010 Feb 15.
Article in English | MEDLINE | ID: mdl-20065111

ABSTRACT

Our laboratory has developed a process for generating mAbs with selectivity to unique peptides in the context of MHC molecules. Recently, we reported that RL4B, an mAb that we have called a TCR mimic (TCRm) because it recognizes peptide in the context of MHC, has cytotoxic activity in vitro and prevented growth of tumor cells in a prophylactic setting. When presented in the context of HLA-A2, RL4B TCRm recognizes the peptide GVLPALPQV derived from human chorionic gonadotropin (hCG)-beta. In this study, we show that RL4B TCRm has strong binding affinity for the GVLPALPQV peptide/HLA-A2 epitope and fine binding specificity for cells that express endogenous hCGbeta Ag and HLA-A2. In addition, suppression of tumor growth with RL4B TCRm was observed in orthotopic models for breast cancer. Using two aggressive human tumor cell lines, MDA-MB-231 and MCF-7, we provide evidence that RL4B TCRm significantly retards tumor growth, supporting a possible role for TCRm agents in therapeutic settings. Moreover, tumors in mice responded to RL4B TCRm therapy in a dose-dependent manner, eliminating tumors at the highest dose. RL4B TCRm strongly detects the hCGbeta peptide/HLA-A2 epitope in human primary breast tumor tissue, but does not react or reacts weakly with normal breast tissue from the same patient. These results further illustrate the selective nature of TCRm Abs and the clinical relevance of the GVLPALPQV peptide/HLA-A2 epitope expression in tumor cells, because they provide the first evidence that Abs that mimic the TCR can be used to markedly reduce and suppress tumor growth.


Subject(s)
Antibodies, Monoclonal/metabolism , Antibody Affinity , Antibody Specificity , Breast Neoplasms/therapy , Chorionic Gonadotropin, beta Subunit, Human/immunology , Growth Inhibitors/physiology , Molecular Mimicry/immunology , Peptide Fragments/metabolism , Animals , Antibodies, Monoclonal/physiology , Antibodies, Monoclonal/therapeutic use , Antigen Presentation/genetics , Antigen Presentation/immunology , Antineoplastic Agents/metabolism , Antineoplastic Agents/therapeutic use , Biomarkers, Tumor/immunology , Biomarkers, Tumor/metabolism , Breast Neoplasms/immunology , Breast Neoplasms/pathology , Cell Line, Transformed , Cell Line, Tumor , Chorionic Gonadotropin, beta Subunit, Human/metabolism , Cytotoxicity Tests, Immunologic , Epitopes, T-Lymphocyte/biosynthesis , Epitopes, T-Lymphocyte/genetics , Gene Targeting , Growth Inhibitors/therapeutic use , HLA-A Antigens/immunology , HLA-A Antigens/metabolism , HLA-A2 Antigen , Humans , Mice , Mice, Nude , Peptide Fragments/immunology
15.
Cancer Immunol Immunother ; 59(4): 563-73, 2010 Apr.
Article in English | MEDLINE | ID: mdl-19779714

ABSTRACT

The identification and validation of new cancer-specific T cell epitopes continues to be a major area of research interest. Nevertheless, challenges remain to develop strategies that can easily discover and validate epitopes expressed in primary cancer cells. Regarded as targets for T cells, peptides presented in the context of the major histocompatibility complex (MHC) are recognized by monoclonal antibodies (mAbs). These mAbs are of special importance as they lend themselves to the detection of epitopes expressed in primary tumor cells. Here, we use an approach that has been successfully utilized in two different infectious disease applications (WNV and influenza). A direct peptide-epitope discovery strategy involving mass spectrometric analysis led to the identification of peptide YLLPAIVHI in the context of MHC A*02 allele (YLL/A2) from human breast carcinoma cell lines. We then generated and characterized an anti-YLL/A2 mAb designated as RL6A TCRm. Subsequently, the TCRm mAb was used to directly validate YLL/A2 epitope expression in human breast cancer tissue, but not in normal control breast tissue. Moreover, mice implanted with human breast cancer cells grew tumors, yet when treated with RL6A TCRm showed a marked reduction in tumor size. These data demonstrate for the first time a coordinated direct discovery and validation strategy that identified a peptide/MHC complex on primary tumor cells for antibody targeting and provide a novel approach to cancer immunotherapy.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Breast Neoplasms/diagnosis , Breast Neoplasms/therapy , Epitopes, T-Lymphocyte/immunology , Peptide Fragments/therapeutic use , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes, Cytotoxic/immunology , Animals , Antibodies, Monoclonal/immunology , Antigens, Neoplasm/immunology , Blotting, Western , Breast/metabolism , Breast/pathology , Breast Neoplasms/immunology , Cancer Vaccines/therapeutic use , DEAD-box RNA Helicases/immunology , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Histocompatibility Antigens Class I/immunology , Humans , Immunoenzyme Techniques , Mice , Mice, Nude , Molecular Mimicry , Peptide Fragments/immunology , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Surface Plasmon Resonance
16.
Int J Oncol ; 34(3): 597-608, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19212664

ABSTRACT

The ability of aspirin to trigger apoptosis in cancer cells is well known and is consistent with the clinical and epidemiological evidence on its chemopreventive effects in curtailing epithelial cancers, including breast cancer. We hypothesized that the anticancer effects of aspirin may involve acetylation of the tumor suppressor protein p53, a known regulator of apoptosis. In the present study, we determined if aspirin at the physiologically achievable concentration of 100 microM acetylates p53 and modulates the expression of p21CIP1, a protein involved in cell cycle arrest, and Bax, a pro-apoptotic protein. Using MDA-MB-231 human breast cancer cells, we demonstrate that aspirin at 100 microM concentration markedly acetylated the p53 protein, which was primarily localized to the nucleus. Aspirin induced p21CIP1 protein levels in a transient fashion in contrast to the sustained induction of Bax. The induction of p21CIP1 protein levels began at 3 h and was maximal at 6-8 h; however, it decreased to control levels by 30 h. In contrast, the anticancer drug, camptothecin (CPT) induced a steady accumulation of p21CIP1 protein. Remarkably, when cells were co-treated with aspirin and CPT, p21CIP1 levels were drastically downregulated, and this phenomenon was observed in many cancer cell lines. Incubation of recombinant p21 with cytoplasmic extracts from aspirin-treated cells caused its degradation suggesting the involvement of proteases in the disappearance of p21CIP1. Consistent with this data, aspirin decreased the survival of CPT-treated cells and greatly increased the extent of apoptosis. Our observation that aspirin has the ability to inhibit p21CIP1 after its initial induction has important implications in chemotherapy, and suggests its potential use to increase the efficacy of anticancer agents.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Aspirin/pharmacology , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Camptothecin/pharmacology , Cyclin-Dependent Kinase Inhibitor p21/biosynthesis , Acetylation/drug effects , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Camptothecin/antagonists & inhibitors , Cell Nucleus/metabolism , Cyclin-Dependent Kinase Inhibitor p21/antagonists & inhibitors , Cyclin-Dependent Kinase Inhibitor p21/genetics , DNA, Neoplasm/metabolism , Drug Synergism , Humans , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Tumor Cells, Cultured , Tumor Suppressor Protein p53/biosynthesis , Tumor Suppressor Protein p53/metabolism , bcl-2-Associated X Protein/biosynthesis
17.
J Immunother ; 31(8): 693-703, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18779750

ABSTRACT

For ovarian cancer (OC) patients with advanced or metastatic disease, standard treatments (chemotherapy and radiotherapy) are not very effective and have undesirable side effects. Newer and more promising approaches in cancer treatment use components of the immune system. In this study, we applied an adoptive immunotherapy-based approach using a cancer testis antigen, sperm protein 17, as a target for the treatment of human metastatic OC in a NOD.CB17-PrkDCcid/J (nonobese, diabetic severe combined immunodeficient) mouse model. We used the human SK-OV-3A2.A3 OC cell line, endogenously expressing sperm protein 17, to induce tumor growth in mice. We provide direct evidence, for the first time, that in vitro cultured, monoclonal, cytotoxic T lymphocytes (derived either from advanced OC patients or from healthy donors), specific for sperm protein 17, can eradicate human metastatic OC cells. In addition, we observed no evidence of autoimmunity after histologic examination of the tissue sections adding to the safety profile of our approach.


Subject(s)
Antigens, Surface/immunology , Carrier Proteins/immunology , Immunotherapy, Adoptive/methods , Ovarian Neoplasms/immunology , Ovarian Neoplasms/therapy , T-Lymphocytes, Cytotoxic/transplantation , Animals , Antigens, Surface/metabolism , Calmodulin-Binding Proteins , Carrier Proteins/metabolism , Cell Line, Tumor , Female , Humans , Male , Membrane Proteins , Mice , Mice, Inbred NOD , Mice, SCID , Ovarian Neoplasms/pathology , T-Lymphocytes, Cytotoxic/immunology
18.
Vaccine ; 26(25): 3092-102, 2008 Jun 13.
Article in English | MEDLINE | ID: mdl-18353510

ABSTRACT

Dendritic cells (DCs) are highly specialized antigen-presenting cells of the immune system that are efficient at presenting peptide-antigen for the activation of T cells and are often the cell type of choice for vaccine targeting by virtue of high expression levels of MHC and costimulatory molecules. Since the level of peptide-MHC complex significantly influences stimulation of T cells, a proof-of-concept potency assay was developed to directly examine the presentation and density of MHC class I peptides derived from the processing of a model tumor antigen, (hCGbeta), on the surface of DCs. In this study we first generated antibodies (TCR mimics or TCRm) to two peptide-HLA-A*0201 epitopes derived from hCGbeta designated as TMT (40-48) and GVL (47-55). Characterization of each TCRm by ELISA and flow cytometric analysis, demonstrated specific binding to soluble recombinant HLA-A2 protein and HLA-A2.1+ T2 cells loaded with relevant peptide. TCRm reactive against the TMT and GVL epitopes blocked granzyme-B production by peptide-specific cytotoxic T lymphocytes (CTL) lines further supporting their recognition specificity. For the assessment of antigen presentation function, human immature monocyte-derived DCs (iDCs) were treated with the mannose receptor targeting vaccine, B11-hCGbeta and matured with Poly I:C. The TMT and GVL epitope reactive CTL lines responded to vaccine-treated but not vehicle-treated mature DCs (mDCs) with TMT and GVL TCRm specifically blocking IFN-gamma production. The TCRm were then used to directly confirm specific peptide-MHC complexes on mDCs. TCRm staining of vaccine-treated mDCs showed detection of the TMT and GVL peptide-HLA-A2 complexes. These findings demonstrate that TCRms may be important tools for determining the potency of DC-based vaccines.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Antigens, Neoplasm/immunology , Cancer Vaccines/administration & dosage , Epitopes/immunology , HLA-A2 Antigen/immunology , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes, Cytotoxic/immunology , Antibodies, Monoclonal/immunology , Antibody Specificity , Cancer Vaccines/immunology , Cell Line , Dendritic Cells/immunology , Epitopes/metabolism , HLA-A2 Antigen/genetics , Humans
19.
J Proteome Res ; 7(4): 1445-57, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18345606

ABSTRACT

Cellular immune mechanisms detect and destroy cancerous and infected cells via the human leukocyte antigen (HLA) class I molecules that present peptides of intracellular origin on the surface of all nucleated cells. The identification of novel, tumor-specific epitopes is a critical step in the development of immunotherapeutics for breast cancer. To directly identify peptide epitopes unique to cancerous cells, secreted human class I HLA molecules (sHLA) were constructed by deletion of the transmembrane and cytoplasmic domain of HLA A*0201. The resulting sHLA-A*0201 was transferred and expressed in breast cancer cell lines MCF-7, MDA-MB-231, and BT-20 as well as in the immortal, nontumorigenic cell line MCF10A. Stable transfectants were seeded into bioreactors for production of > 25 mg of sHLA-A*0201. Peptides eluted from affinity purified sHLA were analyzed by mass spectroscopy. Comparative analysis of HLA-A*0201 peptides revealed 5 previously uncharacterized epitopes uniquely presented on breast cancer cells. These peptides were derived from intracellular proteins with either well-defined or putative roles in breast cancer development and progression: Cyclin Dependent Kinase 2 (Cdk2), Ornithine Decarboxylase (ODC1), Kinetochore Associated 2 (KNTC2 or HEC1), Macrophage Migration Inhibitory Factor (MIF), and Exosome Component 6 (EXOSC6). Cellular recognition of the MIF, KNTC2, EXOSC6, and Cdk2 peptides by circulating CD8+ cells was demonstrated by tetramer staining and IFN-gamma ELISPOT. The identification and characterization of peptides unique to the class I of breast cancer cells provide putative targets for the development of immune diagnostic tools and therapeutics.


Subject(s)
Breast Neoplasms/immunology , Epitopes, T-Lymphocyte/analysis , HLA-A Antigens/immunology , Amino Acid Sequence , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Line, Tumor , Chromatography, High Pressure Liquid , Cyclin-Dependent Kinase 2/immunology , Cyclin-Dependent Kinase 2/metabolism , Cytoskeletal Proteins , Epitopes, T-Lymphocyte/immunology , Epitopes, T-Lymphocyte/isolation & purification , Exoribonucleases/immunology , Exoribonucleases/metabolism , Female , HLA-A Antigens/chemistry , HLA-A2 Antigen , Humans , Interferon-gamma/metabolism , Intramolecular Oxidoreductases/immunology , Intramolecular Oxidoreductases/metabolism , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Macrophage Migration-Inhibitory Factors/immunology , Macrophage Migration-Inhibitory Factors/metabolism , Mass Spectrometry , Nuclear Proteins/immunology , Nuclear Proteins/metabolism , Ornithine Decarboxylase/immunology , Ornithine Decarboxylase/metabolism
20.
Viral Immunol ; 21(4): 435-42, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19115932

ABSTRACT

Recent studies demonstrate that recombinant adeno-associated virus (rAAV)-based antigen loading of dendritic cells (DCs) generates, in vitro, significant and rapid cytotoxic T-lymphocyte (CTL) responses against viral antigens. We used the rAAV system to induce specific CTLs against tumor antigens for the development of ovarian cancer (OC) gene therapy. As an extension of the versatility of the rAAV system, we incorporated a self-antigen, Her-2/neu, which is expressed in many cancers, including breast and ovarian. We analyzed two different vectors containing a short (157-612) and long domain (1-1197). Our rAAV vector induced strong stimulation of CTLs directed against the self tumor antigen, Her-2/neu. We then investigated the efficiency of the CTLs in killing Her-2/neu-targeted cells. A significant MHC class I-restricted, anti-Her-2/neu-specific CTL killing was demonstrated against Her-2/neu-positive OC cells after one in vitro stimulation. In summary, single peripheral blood mononuclear cell (PBMC) stimulation with rAAV/157-612- or rAAV/1-1197-pulsed DCs induces strong antigen-specific CTL generation. The CTLs were capable of lysing low doses of peptides pulsed into target cells or OC Her-2/neu(+) tumors. These data suggest that AAV-based antigen loading of DCs is highly effective for generating human CTL responses against OC antigens.


Subject(s)
Dendritic Cells/immunology , Dependovirus/genetics , Genes, MHC Class I , Ovarian Neoplasms/immunology , Receptor, ErbB-2/immunology , Antigens, Neoplasm/genetics , Antigens, Neoplasm/immunology , Cell Line, Tumor , Cells, Cultured , Cytotoxicity, Immunologic , Dendritic Cells/virology , Dependovirus/immunology , Female , Gene Expression , Genetic Vectors/genetics , Genetic Vectors/immunology , Humans , Immunity, Cellular , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/virology , Ovarian Neoplasms/genetics , Ovarian Neoplasms/virology , Receptor, ErbB-2/genetics , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/virology
SELECTION OF CITATIONS
SEARCH DETAIL
...