Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
J Clin Pharmacol ; 59(7): 915-922, 2019 07.
Article in English | MEDLINE | ID: mdl-30768795

ABSTRACT

Although the medical profession strives for safe prescribing, most medications are unique challenges even when prescribed by an experienced provider. In this article we discuss the pitfalls associated with drug interactions between commonly used antibiotics and anticoagulants, the complexities associated with the administration of novel reversible anticoagulants, the often-overlooked severe adverse drug reactions from commonly used classes of medications such as corticosteroids, the nuances of managing an acetaminophen overdose, and uncommon yet serious adverse events associated with the use of contraceptive hormone drugs.


Subject(s)
Acetaminophen/adverse effects , Adrenal Cortex Hormones/adverse effects , Anti-Bacterial Agents/adverse effects , Anticoagulants/adverse effects , Contraceptives, Oral, Hormonal/adverse effects , Adolescent , Aged , Drug Interactions , Drug Overdose , Female , Humans , Internship and Residency , Male , Middle Aged , Young Adult
2.
J Clin Pharmacol ; 57(8): 947-955, 2017 08.
Article in English | MEDLINE | ID: mdl-28614591

ABSTRACT

Biologicals are a rapidly expanding class of medications used in the treatment of many different conditions. This article reviews the common characteristics of this class and the requirements for safe and effective use in patients. Several vignettes are included to illustrate common challenges.


Subject(s)
Biological Products/therapeutic use , Antibodies, Monoclonal/therapeutic use , Clinical Competence , Humans , Internship and Residency , Pharmacology, Clinical
3.
J Clin Pharmacol ; 56(12): 1461-1473, 2016 12.
Article in English | MEDLINE | ID: mdl-27539787

ABSTRACT

Differences in physiology related to young or old age and/or organ system impairment alter the absorption, distribution, metabolism, and excretion of many medications and consequently their effectiveness and toxicity. This module discusses common alterations in medication use and dosage that are required in the pediatric age group, in the elderly, and in patients with renal or hepatic disease.


Subject(s)
Drug Dosage Calculations , Kidney/drug effects , Liver/drug effects , Pharmaceutical Preparations/administration & dosage , Adolescent , Aged , Aged, 80 and over , Child , Child, Preschool , Dosage Forms/standards , Drug-Related Side Effects and Adverse Reactions/metabolism , Drug-Related Side Effects and Adverse Reactions/prevention & control , Humans , Infant , Infant, Newborn , Kidney/metabolism , Liver/metabolism , Pediatrics/methods , Pharmaceutical Preparations/metabolism
4.
PLoS One ; 9(11): e112664, 2014.
Article in English | MEDLINE | ID: mdl-25401701

ABSTRACT

Glucose metabolism is under the cooperative regulation of both insulin receptor (IR) and ß(2)-adrenergic receptor (ß(2)AR), which represent the receptor tyrosine kinases (RTKs) and seven transmembrane receptors (7TMRs), respectively. Studies demonstrating cross-talk between these two receptors and their endogenous coexpression have suggested their possible interactions. To evaluate the effect of IR and prospective heteromerization on ß(2)AR properties, we showed that IR coexpression had no effect on the ligand binding properties of ß(2)AR; however, IR reduced ß(2)AR surface expression and accelerated its internalization. Additionally, both receptors displayed a similar distribution pattern with a high degree of colocalization. To test the possible direct interaction between ß(2)AR and IR, we employed quantitative BRET(2) saturation and competition assays. Saturation assay data suggested constitutive ß(2)AR and IR homo- and heteromerization. Calculated acceptor/donor (AD50) values as a measure of the relative affinity for homo- and heteromer formation differed among the heteromers that could not be explained by a simple dimer model. In heterologous competition assays, a transient increase in the BRET(2) signal with a subsequent hyperbolical decrease was observed, suggesting higher-order heteromer formation. To complement the BRET(2) data, we employed the informational spectrum method (ISM), a virtual spectroscopy method to investigate protein-protein interactions. Computational peptide scanning of ß(2)AR and IR identified intracellular domains encompassing residues at the end of the 7th TM domain and C-terminal tail of ß(2)AR and a cytoplasmic part of the IR ß chain as prospective interaction domains. ISM further suggested a high probability of heteromer formation and homodimers as basic units engaged in heteromerization. In summary, our data suggest direct interaction and higher-order ß(2)AR:IR oligomer formation, likely comprising heteromers of homodimers.


Subject(s)
Computational Biology , Models, Biological , Receptor, Insulin/metabolism , Receptors, Adrenergic, beta-2/metabolism , Algorithms , Cell Line , Cell Membrane/metabolism , Computational Biology/methods , Gene Expression , Humans , Protein Binding , Protein Interaction Domains and Motifs , Protein Multimerization , Protein Transport , Receptor, Insulin/chemistry , Receptor, Insulin/genetics , Receptors, Adrenergic, beta-2/chemistry , Receptors, Adrenergic, beta-2/genetics , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
5.
Front Oncol ; 4: 266, 2014.
Article in English | MEDLINE | ID: mdl-25325015

ABSTRACT

Cancer patients frequently exhibit a deficiency in Type-1 (but not Type-2 or regulatory) CD4(+) T cell responses against tumor-associated antigens (TAA), which may limit protection against disease progression or responsiveness to immunotherapy in these individuals. Since such deficiency was acutely evident in patients with active disease (AD), where chronic stimulation of anti-tumor CD4(+) T cells would be expected and activation-induced cell death may be prevalent, we employed MHC Class II-peptide tetramers to characterize the frequency and apoptotic status of TAA- vs. influenza (FluM1) virus-specific CD4(+) T cells in the peripheral blood of HLA-DR*0401(+) patients with melanoma or renal cell carcinoma. We observed that Flu-specific CD4(+) T cells ranged from 0.17 to 3.89%, while up to approximately 1% of CD4(+) T cells reacted against individual TAA epitopes derived from the EphA2 or MAGE-6 proteins. The frequencies of EphA2 and MAGE-6-specific CD4(+) T cells in patients were significantly correlated with AD and gender of the patient (i.e., females > males), while frequencies of Flu-specific CD4(+) T cells were distributed within a normal range in all patients. Notably, patient CD4(+) T cells reactive with MHC class II-TAA (but not MHC class II-Flu) tetramers were significantly enriched for a pro-apoptotic (Annexin-V(+)) phenotype, particularly amongst the Th1 (T-bet(+)) subset. These results suggest that the preferential sensitivity of TAA (but not viral)-specific CD4(+) Th1 cells to apoptosis in melanoma patients with AD will need to be overcome for optimal clinical benefit of immunotherapeutic approaches to be realized.

6.
J Immunother ; 33(3): 316-25, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20445352

ABSTRACT

AlphaVbeta3 (alphavbeta3) is an important molecule for tumor-induced angiogenesis and is upregulated in metastatic melanoma (MM). We proposed to study the mechanism of action of etaracizumab, a monoclonal antibody targeting alphavbeta3, in MM. Patients with MM and biopsiable tumor were treated with etaracizumab in 3 dose cohorts starting from 8 mg/kg. Tumor saturation by etaracizumab using LM609 immunohistochemical staining of tumor sections was the primary endpoint. Subsequent dose cohorts were defined based on the tumor saturation by etaracizumab. Secondary end points were analysis of clinical benefit and changes from baseline of several tumor and peripheral blood biomarkers. Eighteen patients were enrolled at 3 dose levels. Etaracizumab showed better melanoma cell saturation at the 8mg/kg and 1 mg/kg dose compared with the 4 mg/kg dose and better vascular endothelial cell saturation at 8 mg/kg compared with lower dose groups. Etaracizumab demonstrated an acceptable safety profile. The optimal biologic dose out of those selected for investigation was 8 mg/kg. Patients treated at the highest dose may have had better clinical benefit secondary to suppression of the activated immediate downstream effector of alphavbeta3 signaling, FAK, in melanoma cells, but this alone did not ultimately affect melanoma cell proliferation or apoptosis. No apparent antiangiogenic or immunomodulatory effects of etaracizumab were noted.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Melanoma/drug therapy , Adult , Aged , Aged, 80 and over , Antibodies, Monoclonal/adverse effects , Antibodies, Monoclonal/pharmacokinetics , Antibodies, Monoclonal, Humanized , Biomarkers, Tumor/blood , Cohort Studies , Dose-Response Relationship, Drug , Female , Follow-Up Studies , Humans , Immunohistochemistry , Integrin alphaVbeta3/immunology , Ki-67 Antigen/blood , Male , Melanoma/immunology , Melanoma/pathology , Middle Aged , Neoplasm Metastasis , Survival Analysis , Tachycardia/chemically induced , Time Factors , Treatment Outcome , Vascular Endothelial Growth Factor A/blood
7.
Cancer Res ; 69(17): 6995-7003, 2009 Sep 01.
Article in English | MEDLINE | ID: mdl-19690146

ABSTRACT

EphA2, a member of the receptor tyrosine kinase family, is commonly expressed by a broad range of cancer types, where its level of (over)expression correlates with poor clinical outcome. Because tumor cell expressed EphA2 is a nonmutated "self" protein, specific CD8(+) T cells are subject to self-tolerance mechanisms and typically exhibit only moderate-to-low functional avidity, rendering them marginally competent to recognize EphA2(+) tumor cells in vitro or in vivo. We have recently reported that the ability of specific CD8(+) T cells to recognize EphA2(+) tumor cells can be augmented after the cancer cells are pretreated with EphA2 agonists that promote proteasomal degradation and up-regulated expression of EphA2/class I complexes on the tumor cell membrane. In the current study, we show that treatment of EphA2(+) tumor cells with the irreversible heat shock protein 90 inhibitor, 17-dimethylaminoethylamino-17-demethoxygeldanamycin (17-DMAG), similarly enhances their recognition by EphA2-specific CD8(+) T-cell lines and clones in vitro via a mechanism that is dependent on proteasome and transporter-associated protein function as well as the retrotranslocation of EphA2 into the tumor cytoplasm. When 17-DMAG and agonist anti-EphA2 monoclonal antibodies are coapplied, T-cell recognition of tumor cells is further increased over that observed for either agent alone. These studies suggest that EphA2 represents a novel heat shock protein 90 client protein and that the treatment of cancer patients with 17-DMAG-based "pulse" therapy may improve the antitumor efficacy of CD8(+) T effector cells reactive against EphA2-derived epitopes.


Subject(s)
Benzoquinones/pharmacology , CD8-Positive T-Lymphocytes/immunology , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Lactams, Macrocyclic/pharmacology , Neoplasms/immunology , Neoplasms/metabolism , Receptor, EphA2/immunology , Antibodies, Monoclonal , Antigens, Neoplasm/immunology , Benzoquinones/immunology , Benzoquinones/therapeutic use , Cell Line, Tumor , Epitopes , HSP90 Heat-Shock Proteins/immunology , HSP90 Heat-Shock Proteins/metabolism , Humans , Immunotherapy , Lactams, Macrocyclic/immunology , Lactams, Macrocyclic/therapeutic use , Lymphocyte Activation/drug effects , Neoplasms/therapy , Proteasome Endopeptidase Complex , Protein Transport , Receptor, EphA2/metabolism
8.
J Immunol ; 181(11): 7721-7, 2008 Dec 01.
Article in English | MEDLINE | ID: mdl-19017961

ABSTRACT

The EphA2 receptor tyrosine kinase is an attractive therapeutic target that is commonly overexpressed on solid tumors, with the degree of overexpression associated with disease progression, metastatic potential, and poor prognosis. Agonistic mAbs or ligand (ephrinA1)-Fc fusion protein are capable of inducing EphA2 internalization and degradation, thereby (at least transiently) eliminating the influence of this oncoprotein. We and others have also shown that EphA2 contains multiple peptide epitopes that can be recognized by effector CD4(+) and CD8(+) T cells isolated from tumor-bearing patients. Herein, we show that "agonist" reagents that trigger the proteasome-dependent degradation of tumor cell EphA2 result in the improved presentation of peptides derived from (both the extracellular and intracellular domains of) EphA2 in MHC class I complexes expressed on the tumor cell membrane for at least 48 h, as manifested by increased recognition by EphA2-specific CD8(+) T cells in vitro. We also observed that while delivery of ephrinA1-Fc fusion protein or agonist mAb into EphA2(+) tumor lesions promotes EphA2 degradation in situ, this single administration of agent does not dramatically alter tumor progression in a humanized SCID model. However, when combined with the adoptive transfer of normally nontherapeutic (human) anti-EphA2 CD8(+) CTL, this dual-agent regimen results in complete tumor eradication. These results suggest that strategies targeting the conditional proteasome-mediated destruction of tumor cell EphA2 may enable EphA2-specific CD8(+) T cells (of modest functional avidity) to realize improved therapeutic potential.


Subject(s)
Antigen Presentation/immunology , CD8-Positive T-Lymphocytes/immunology , Epitopes, T-Lymphocyte/immunology , Neoplasm Proteins/immunology , Peptides/immunology , Prostatic Neoplasms/immunology , Receptor, EphA2/immunology , Adoptive Transfer , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Antibodies, Neoplasm/pharmacology , Antigen Presentation/drug effects , Cell Line, Tumor , Disease Models, Animal , Female , Humans , Male , Mice , Mice, SCID , Neoplasm Metastasis , Neoplasm Transplantation , Prostatic Neoplasms/therapy , Proteasome Endopeptidase Complex/immunology , Protein Structure, Tertiary , Receptor, EphA2/antagonists & inhibitors
9.
Clin Immunol ; 127(2): 123-9, 2008 May.
Article in English | MEDLINE | ID: mdl-18405863

ABSTRACT

A patient from the University of Pittsburgh is presented who developed autoimmunity during IL-2 based combination therapy. IL-2 was originally described as a "T cell growth factor" capable of expanding previously activated T cells, enhancing the cytotoxicity of antigen-specific cytotoxic T cells and NK cells. High dose Interleukin 2 (HDIL2) is now FDA-approved for therapy of patients with metastatic melanoma and renal cell carcinoma, based on its ability to induce durable responses in 5-10% of patients. The antitumor effect of HDIL2 is incompletely understood, but it appears that this regimen alters the balance of immigrant T effector cells in relation to T suppressor cells. It promotes a less immunosuppressive tumor microenvironment, inducing tumor regression in a subset of patients that is yet to be defined. The antitumor activity of IL-2, as for other agents that promote durable antitumor activity against melanoma such as interferon alpha and anti-CTLA4 antibody, is frequently associated with development of autoimmunity as observed in this patient. We present studies investigating the mechanisms for the therapeutic benefit of HDIL2 in melanoma.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/adverse effects , Interleukin-2/adverse effects , Melanoma/therapy , Antineoplastic Combined Chemotherapy Protocols/immunology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Autoimmunity/drug effects , Autoimmunity/immunology , CD4-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Dacarbazine/administration & dosage , Dacarbazine/adverse effects , Dacarbazine/analogs & derivatives , Humans , Immunotherapy/adverse effects , Immunotherapy/methods , Interleukin-2/administration & dosage , Interleukin-2/immunology , Male , Melanoma/immunology , Middle Aged , Temozolomide , Vitiligo/chemically induced , Vitiligo/immunology
10.
J Immunother ; 31(3): 254-62, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18317362

ABSTRACT

Dendritic cells (DCs) are potent antigen presenting cells and represent attractive candidates for use in novel immunotherapies for patients with renal cell carcinoma (RCC), a disease that has proven refractory to conventional treatment modalities, such as chemotherapy and radiotherapy. Given the perceived need to augment antitumor type-1 immunity (TC1 and Th1) as a therapeutic end point, and the known functional plasticity of DC populations that may display heterogeneous capacity to promote T-cell responses, we sought to identify a preferred DC preparation with this capacity. We compared 2 different preparations of monocyte-derived DC using interferon-alpha (IFN-alpha) (IFN-DC and alphaDC1) with classic DCs "matured" (mDCs) using interleukin-1beta/interleukin-6/tumor necrosis factor-alpha/prostaglandin E2, for their ability to promote autologous TC1 antitumor responses from RCC patients in vitro. IFN-alpha-conditioned DC promoted significantly higher numbers of RCC-specific CD8+ T cells exhibiting a cytotoxic phenotype after in vitro stimulation (IVS) than cytokine cocktail-mDCs. Furthermore, IVS using IFN-DCs was able to diminish regulatory-type T cells among CD4+ T-cell responder populations versus IVS using conventional mDC-based vaccines. These data emphasize an important role for IFN-alpha in modulating the immunologic functions of DCs toward a polarized DC1-type capable of coordinately promoting TH1-type and TC1-type T-cell mediated immunity and supports the translational development of patient-derived IFN-alpha-conditioned DC for use in novel immunotherapies for patients with RCC, and in whom, endogenous tumor-specific TC1 effector cells may be dysfunctional, anergic, or prone to undergo apoptosis.


Subject(s)
Carcinoma, Renal Cell/immunology , Dendritic Cells/immunology , Interferon-alpha/immunology , Kidney Neoplasms/immunology , Lymphocyte Activation , T-Lymphocytes, Regulatory/immunology , Th1 Cells/immunology , Carcinoma, Renal Cell/blood , Carcinoma, Renal Cell/pathology , Cell Communication , Cell Differentiation/drug effects , Cell Differentiation/immunology , Cells, Cultured , Cytokines/immunology , Cytotoxicity, Immunologic , Dendritic Cells/pathology , Humans , Immunologic Memory , Immunotherapy, Active , Interferon Type I/immunology , Interferon Type I/pharmacology , Interferon-alpha/metabolism , Interferon-alpha/pharmacology , Interferon-gamma/immunology , Kidney Neoplasms/blood , Kidney Neoplasms/pathology , Lymphocyte Activation/drug effects , Recombinant Proteins
11.
Clin Cancer Res ; 13(22 Pt 1): 6796-806, 2007 Nov 15.
Article in English | MEDLINE | ID: mdl-18006782

ABSTRACT

PURPOSE: Although T-helper (Th) epitopes have been previously reported for many tumor antigens, including MAGE-A6, the relevant HLA-DR alleles that present these peptides are expressed by only a minority of patients. The identification of tumor antigenic epitopes presented promiscuously by many HLA-DR alleles would extend the clinical utility of these peptides in vaccines and for the immunomonitoring of cancer patients. EXPERIMENTAL DESIGN: A neural network algorithm and in vitro sensitization assays were employed to screen candidate peptides for their immunogenicity. RESULTS: The MAGE-A6(140-170), MAGE-A6(172-187), and MAGE-A6(280-302) epitopes were recognized by CD4+ T cells isolated from the majority of normal donors and melanoma patients evaluated. Peptide-specific CD4+ T cells also recognized autologous antigen-presenting cell pulsed with recombinant MAGE-A6 (rMAGE) protein, supporting the natural processing and MHC presentation of these epitopes. Given the strong primary in vitro sensitization of normal donor CD4+ T cells by the MAGEA6(172-187) epitope, suggestive of potential cross-reactivity against an environmental stimulus, we identified a highly homologous peptide within the Mycoplasma penetrans HF-2 permease (MPHF2) protein. MPHF2 peptide-primed CD4+ T cells cross-reacted against autologous APC pulsed with the MAGE-A6(172-187) peptide or rMAGE protein and recognized HLA-matched MAGE-A6+ melanoma cell lines. These responses seemed heteroclitic in nature because the functional avidity of MPHF2 peptide-primed CD4+ T cells for the MAGE-A6(172-187) peptide was approximately 1,000 times greater than that of CD4+ T cells primed with the corresponding MAGE-A6 peptide. CONCLUSIONS: We believe that these novel "promiscuous" MAGE-A6/MPHF2 Th epitopes may prove clinically useful in the treatment and/or monitoring of a high proportion of cancer patients.


Subject(s)
Antigens, Neoplasm/pharmacology , Bacterial Proteins/pharmacology , CD4-Positive T-Lymphocytes/drug effects , Epitopes, T-Lymphocyte/pharmacology , Mycoplasma penetrans/immunology , Neoplasm Proteins/pharmacology , Peptides/pharmacology , Adult , Aged , Amino Acid Sequence , Antigens, Neoplasm/chemistry , Antigens, Neoplasm/genetics , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , CD4-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Epitopes, T-Lymphocyte/chemistry , Epitopes, T-Lymphocyte/genetics , Female , Humans , Male , Middle Aged , Molecular Sequence Data , Neoplasm Proteins/chemistry , Neoplasm Proteins/genetics , Peptides/chemistry , Peptides/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/pharmacology
12.
Cancer Res ; 67(12): 5949-56, 2007 Jun 15.
Article in English | MEDLINE | ID: mdl-17575165

ABSTRACT

The goal of this study was to investigate the effects of adenosine and its stable analogue 2-chloroadenosine (CADO) on the cytotoxic activity and cytokine production by human antimelanoma specific CD8+ and CD4+ T-helper type 1 (Th1) clones. The cytotoxic activity of CD8+ T cells was inhibited by adenosine and CADO. Using Lab MAP multiplex technology, we found that adenosine inhibits production of various cytokines and chemokines by CD8+ and CD4+ T cells. Studies with CGS21680, a specific agonist of adenosine A2A receptor (AdoRA2A), and ZM241385, an AdoRA2-selective antagonist, indicate that the inhibitory effects of adenosine are mediated via cyclic AMP (cAMP)-elevating AdoRA2A, leading to protein kinase A (PKA) activation. Using cAMP analogues with different affinities for the A and B sites of the regulatory subunits of PKAI and PKAII, we found that activation of PKAI, but not of PKAII, mimicked the inhibitory effects of adenosine on T-cell cytotoxic activity and cytokine production. Inhibitors of the PKA catalytic subunits (H89 and PKA inhibitor peptide 14-22) failed to abrogate the inhibitory effects of CADO. In contrast, Rp-8-Br-cAMPS that antagonizes binding of cAMP to the regulatory I subunit and PKA activation was efficient in blocking the inhibitory effect of adenosine on the functional activity of T cells. Our findings on the ability of adenosine to inhibit the effector function of antimelanoma specific T cells suggest that intratumor-produced adenosine could impair the function of tumor-infiltrating T lymphocytes. Thus, blocking the inhibitory activity of tumor-produced adenosine might represent a new strategy for improvement of cancer immunotherapy.


Subject(s)
Adenosine/metabolism , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cyclic AMP-Dependent Protein Kinases/metabolism , Cytokines/biosynthesis , Melanoma/immunology , 2-Chloroadenosine/metabolism , Adenosine/analogs & derivatives , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , Cytotoxicity, Immunologic , Humans , Receptor, Adenosine A2A/metabolism , Signal Transduction/immunology
13.
J Immunol ; 174(3): 1751-9, 2005 Feb 01.
Article in English | MEDLINE | ID: mdl-15661941

ABSTRACT

NY-ESO-1 is expressed by a broad range of human tumors and is often recognized by Abs in the sera of cancer patients with NY-ESO-1-expressing tumors. The NY-ESO-1 gene also encodes several MHC class I- and class II-restricted tumor epitopes recognized by T lymphocytes. In this study we report one novel pan-MHC class II-restricted peptide sequence, NY-ESO-1 87-111, that is capable of binding to multiple HLA-DR and HLA-DP4 molecules, including HLA-DRB1*0101, 0401, 0701, and 1101 and HLA-DPB1*0401 and 0402 molecules. We also demonstrate that peptide NY-ESO-1 87-111 stimulates Th1-type and Th-2/Th0-type CD4(+) T cells and clones when presented in the context of these HLA-DR and HLA-DP4 molecules. Both bulk CD4(+) T cells and CD4(+) T cell clones were capable of recognizing not only peptide-pulsed APCs, but also autologous dendritic cells, either loaded with the NY-ESO-1 protein or transfected with NY-ESO-1 cDNAs. Using IFN-gamma and IL-5 ELISPOT assays and PBL from patients with NY-ESO-1-expressing tumors, we observed the existence of Th1-type circulating CD4(+) T cells recognizing peptide NY-ESO-1 87-111 in the context of HLA-DP4 molecules. Taken together, these data represent the first report of an HLA-DR- and HLA-DP-restricted epitope from a tumor Ag. They also support the relevance of cancer vaccine trials with peptides NY-ESO-1 87-111 in the large number of cancer patients with NY-ESO-1-expressing tumors.


Subject(s)
Antigens, Neoplasm/metabolism , CD4-Positive T-Lymphocytes/immunology , Epitopes, T-Lymphocyte/metabolism , HLA-DP Antigens/metabolism , HLA-DR Antigens/metabolism , Immunodominant Epitopes/metabolism , Melanoma/immunology , Membrane Proteins/metabolism , Peptide Fragments/metabolism , Alleles , Amino Acid Sequence , Antibodies, Neoplasm/biosynthesis , Antigen Presentation , Antigens, Neoplasm/biosynthesis , Antigens, Neoplasm/immunology , CD4-Positive T-Lymphocytes/metabolism , Clone Cells , Dendritic Cells/immunology , Dendritic Cells/metabolism , Epitopes, T-Lymphocyte/blood , Epitopes, T-Lymphocyte/immunology , Female , HLA-DP Antigens/immunology , HLA-DP beta-Chains , HLA-DR Antigens/immunology , Humans , Immunodominant Epitopes/blood , Immunodominant Epitopes/immunology , Lymphocyte Activation/immunology , Male , Melanoma/metabolism , Membrane Proteins/biosynthesis , Membrane Proteins/immunology , Molecular Sequence Data , Peptide Fragments/immunology , Protein Binding/immunology , Th2 Cells/immunology , Th2 Cells/metabolism
14.
Srp Arh Celok Lek ; 132 Suppl 1: 45-8, 2004 Oct.
Article in Serbian | MEDLINE | ID: mdl-15615465

ABSTRACT

A small group of three patients presented in our study represents extraordinary and very complicated problem of congenital hiatus hernia in infant period from 6th to 9th month of life, associated with gastroesophageal reflux and consecutive esophageal stenosis. There are two very rare and delicate entities within differential diagnosis, in the domain of the same pathology: congenitally short esophagus and congenital esophageal stenosis; with completely different surgical options for their treatment. That is why an optimal diagnostics and an adequate operative technique are extremely important for the treatment of hiatus hernia. The uppergastrointestinal barium radiography is definitely the method of diagnosing hiatus hernia, which provides typical, almost pathognomonic image of hiatus hernia accompanied by the esophageal stenosis. Nissen fundoplication is the technique of choice for its surgical treatment, with gastrostomy for probable postoperative esophageal dilatation. The results are more than satisfying: early peroral feeding, with spontaneous resolving of esophageal stenosis, which significantly diminishes the need for esophageal bougienage.


Subject(s)
Esophageal Stenosis/etiology , Gastroesophageal Reflux/complications , Hernia, Hiatal/congenital , Hernia, Hiatal/surgery , Esophageal Stenosis/congenital , Esophageal Stenosis/diagnosis , Esophagus/abnormalities , Gastroesophageal Reflux/diagnosis , Gastroesophageal Reflux/surgery , Hernia, Hiatal/complications , Hernia, Hiatal/diagnosis , Humans , Infant
15.
Cancer Res ; 63(19): 6506-15, 2003 Oct 01.
Article in English | MEDLINE | ID: mdl-14559844

ABSTRACT

The NY-ESO-1 and LAGE-1 genes are expressed by many human cancers, but not by normal tissues, with the exception of testis and placenta. The NY-ESO-1 and LAGE-1 genes give rise to multiple MHC class I and class II-presented epitopes derived from the open reading frames (ORF) 1 and 2. Here, we have investigated whether NY-ESO-1/LAGE-1 ORF2 encodes promiscuous MHC class II-restricted epitopes. Using a set of overlapping peptides from the ORF2 protein sequence and autologous dendritic cells (DCs) from normal donors and melanoma patients, we have identified three HLA-DRB1*0401-restricted peptide sequences from the LAGE-1 ORF2 that are capable of stimulating T-helper 1-type melanoma-reactive CD4+ T cells. From these bulk CD4+ T cells, we have generated CD4+ T-cell clones able to recognize not only peptide-pulsed DCs but also autologous DCs loaded with the LAGE-1 ORF2 protein. We have demonstrated that these peptides not only bind to multiple HLA-DR molecules apart from HLA-DRB1*0401 but also stimulate CD4+ T cells when presented in the context of these HLA-DR molecules. Furthermore, our binding data have delineated two additional sequences capable of broadly binding to multiple HLA-DR molecules. Altogether, these data support the immunogenicity of NY-ESO-1/LAGE-1 ORF2 gene products and clearly demonstrate their capability to stimulate T-helper 1 type CD4+ T cells. Because of the role of these cells in promoting long-lasting antitumor CTL responses, our data provide a rationale for cancer vaccine trials with peptides derived from the NY-ESO-1/LAGE-1 ORF2 for a large fraction of patients with NY-ESO-1/LAGE-1(+) tumors.


Subject(s)
Epitopes, T-Lymphocyte/immunology , HLA-DR Antigens/immunology , Membrane Proteins , Proteins/genetics , Th1 Cells/immunology , Amino Acid Sequence , Antigens, Neoplasm/genetics , Antigens, Neoplasm/immunology , Antigens, Surface , Binding Sites , Cell Line, Tumor , Dendritic Cells/immunology , Epitopes, T-Lymphocyte/genetics , HLA-DR Antigens/genetics , HLA-DRB1 Chains , Humans , Melanoma/genetics , Melanoma/immunology , Molecular Sequence Data , Open Reading Frames/immunology , Proteins/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...