Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 53
Filter
Add more filters

Publication year range
1.
Gastroenterology ; 161(5): 1601-1614.e23, 2021 11.
Article in English | MEDLINE | ID: mdl-34303658

ABSTRACT

BACKGROUND & AIMS: Promoted by pancreatitis, oncogenic KrasG12D triggers acinar cells' neoplastic transformation through acinar-to-ductal metaplasia (ADM) and pancreatic intraepithelial neoplasia. Anterior gradient 2 (Agr2), a known inhibitor of p53, is detected at early stage of pancreatic ductal adenocarcinoma (PDAC) development. RNA polymerase II (RNAPII) is a key nuclear enzyme; regulation of its nuclear localization in mammalian cells represents a potential therapeutic target. METHODS: A mouse model of inflammation-accelerated KrasG12D-driven ADM and pancreatic intraepithelial neoplasia development was used. Pancreas-specific Agr2 ablation was performed to access its role in pancreatic carcinogenesis. Hydrophobic hexapeptides loaded in liposomes were developed to disrupt Agr2-RNAPII complex. RESULTS: We found that Agr2 is up-regulated in ADM-to-pancreatic intraepithelial neoplasia transition in inflammation and KrasG12D-driven early pancreatic carcinogenesis. Genetic ablation of Agr2 specifically blocks this metaplastic-to-neoplastic process. Mechanistically, Agr2 directs the nuclear import of RNAPII via its C-terminal nuclear localization signal, undermining the ATR-dependent p53 activation in ADM lesions. Because Agr2 binds to the largest subunit of RNAPII in a peptide motif-dependent manner, we developed a hexapeptide to interfere with the nuclear import of RNAPII by competitively disrupting the Agr2-RNAPII complex. This novel hexapeptide leads to dysfunction of RNAPII with concomitant activation of DNA damage response in early neoplastic lesions; hence, it dramatically compromises PDAC initiation in vivo. Moreover, the hexapeptide sensitizes PDAC cells and patient-derived organoids harboring wild-type p53 to RNAPII inhibitors and first-line chemotherapeutic agents in vivo. Of note, this therapeutic effect is efficient across various cancer types. CONCLUSIONS: Agr2 is identified as a novel adaptor protein for nuclear import of RNAPII in mammalian cells. Also, we provide genetic evidence defining Agr2-dependent nuclear import of RNAPII as a pharmaceutically accessible target for prevention and treatment in PDAC in the context of wild-type p53.


Subject(s)
Carcinoma in Situ/enzymology , Carcinoma, Pancreatic Ductal/enzymology , Mucoproteins/metabolism , Oncogene Proteins/metabolism , Pancreatic Neoplasms/enzymology , RNA Polymerase II/metabolism , Tumor Suppressor Protein p53/metabolism , Active Transport, Cell Nucleus , Animals , Antineoplastic Agents/pharmacology , Carcinoma in Situ/drug therapy , Carcinoma in Situ/genetics , Carcinoma in Situ/pathology , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Gene Expression Regulation, Neoplastic , Metaplasia , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Mucoproteins/genetics , Mutation , Oligopeptides/pharmacology , Oncogene Proteins/genetics , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Proto-Oncogene Proteins p21(ras)/genetics , RNA Polymerase II/genetics , Tumor Suppressor Protein p53/genetics
3.
J Am Chem Soc ; 143(35): 14322-14331, 2021 09 08.
Article in English | MEDLINE | ID: mdl-34459587

ABSTRACT

Synthetic methods on the macrocyclization of peptides are of high interest since they facilitate the synthesis of various types of potentially bioactive compounds, e.g. addressing targets like protein-protein-interactions. Herein, we report on an efficient method to construct tryptathionine-cross-links in peptides between the amino acids Trp and Cys. This reaction not only is the basis for the total synthesis of the death cap toxin α-amanitin but also provides rapid access to various new amanitin analogues. This study for the first time presents a systematic compilation of structure-activity relations (SAR) of amatoxins with regard to RNA polymerase II inhibition and cytotoxicity with one amanitin derivative of superior RNAP II inhibition. The present approach paves the way for the synthesis of structurally diverse amatoxins as future payloads for antibody-toxin conjugates in cancer therapy.

4.
Chemistry ; 27(40): 10282-10292, 2021 Jul 16.
Article in English | MEDLINE | ID: mdl-34058032

ABSTRACT

Alpha-amanitin, an extremely toxic bicyclic octapeptide extracted from the death-cap mushroom, Amanita phalloides, is a highly selective allosteric inhibitor of RNA polymerase II. Following on growing interest in using this toxin as a payload in antibody-drug conjugates, herein we report the synthesis and biochemical evaluation of several new derivatives of this toxin to probe the role of the trans-hydroxyproline (Hyp), which is known to be critical for toxicity. This structure activity relationship (SAR) study represents the first of its kind to use various Hyp-analogs to alter the conformational and H-bonding properties of Hyp in amanitin.


Subject(s)
Alpha-Amanitin , Immunoconjugates , Amanita , Hydroxyproline
5.
Nature ; 520(7549): 697-701, 2015 Apr 30.
Article in English | MEDLINE | ID: mdl-25901683

ABSTRACT

TP53, a well-known tumour suppressor gene that encodes p53, is frequently inactivated by mutation or deletion in most human tumours. A tremendous effort has been made to restore p53 activity in cancer therapies. However, no effective p53-based therapy has been successfully translated into clinical cancer treatment owing to the complexity of p53 signalling. Here we demonstrate that genomic deletion of TP53 frequently encompasses essential neighbouring genes, rendering cancer cells with hemizygous TP53 deletion vulnerable to further suppression of such genes. POLR2A is identified as such a gene that is almost always co-deleted with TP53 in human cancers. It encodes the largest and catalytic subunit of the RNA polymerase II complex, which is specifically inhibited by α-amanitin. Our analysis of The Cancer Genome Atlas (TCGA) and Cancer Cell Line Encyclopedia (CCLE) databases reveals that POLR2A expression levels are tightly correlated with its gene copy numbers in human colorectal cancer. Suppression of POLR2A with α-amanitin or small interfering RNAs selectively inhibits the proliferation, survival and tumorigenic potential of colorectal cancer cells with hemizygous TP53 loss in a p53-independent manner. Previous clinical applications of α-amanitin have been limited owing to its liver toxicity. However, we found that α-amanitin-based antibody-drug conjugates are highly effective therapeutic agents with reduced toxicity. Here we show that low doses of α-amanitin-conjugated anti-epithelial cell adhesion molecule (EpCAM) antibody lead to complete tumour regression in mouse models of human colorectal cancer with hemizygous deletion of POLR2A. We anticipate that inhibiting POLR2A will be a new therapeutic approach for human cancers containing such common genomic alterations.


Subject(s)
Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Genes, p53/genetics , Tumor Suppressor Protein p53/deficiency , Alpha-Amanitin/adverse effects , Alpha-Amanitin/chemistry , Alpha-Amanitin/pharmacology , Alpha-Amanitin/therapeutic use , Animals , Antibodies/chemistry , Antibodies/immunology , Antigens, Neoplasm/immunology , Catalytic Domain , Cell Adhesion Molecules/immunology , Cell Line, Tumor , Cell Proliferation/drug effects , Colorectal Neoplasms/enzymology , Colorectal Neoplasms/pathology , Databases, Genetic , Disease Models, Animal , Epithelial Cell Adhesion Molecule , Female , Gene Deletion , Gene Dosage/genetics , Humans , Immunoconjugates/adverse effects , Immunoconjugates/chemistry , Immunoconjugates/immunology , Immunoconjugates/therapeutic use , Mice , Protein Subunits/chemistry , Protein Subunits/deficiency , Protein Subunits/genetics , RNA Polymerase II/antagonists & inhibitors , RNA Polymerase II/chemistry , RNA Polymerase II/deficiency , RNA Polymerase II/genetics , Tumor Suppressor Protein p53/biosynthesis , Tumor Suppressor Protein p53/genetics , Xenograft Model Antitumor Assays
6.
Angew Chem Int Ed Engl ; 59(28): 11390-11393, 2020 07 06.
Article in English | MEDLINE | ID: mdl-32091645

ABSTRACT

α-Amanitin and related amatoxins have been studied for more than six decades mostly by isolation from death cap mushrooms. The total synthesis, however, remained challenging due to unique structural features. α-Amanitin is a potent inhibitor of RNA polymerase II. Interrupting the basic transcription processes of eukaryotes leads to apoptosis of the cell. This unique mechanism makes the toxin an ideal payload for antibody-drug conjugates (ADCs). Only microgram quantities of toxins, when delivered selectively to tumor sites through conjugation to antibodies, are sufficient to eliminate malignant tumor cells of almost every origin. By solving the stereoselective access to dihydroxyisoleucine, a photochemical synthesis of the tryptathion precursor, solid-phase peptide synthesis, and macrolactamization we obtained a scalable synthetic route towards synthetic α-amanitin. This makes α-amanitin and derivatives now accessible for the development of new ADCs.


Subject(s)
Alpha-Amanitin/chemical synthesis , Amanitins/chemical synthesis , Agaricales/chemistry , Alpha-Amanitin/chemistry , Amanitins/chemistry , Chromatography, High Pressure Liquid , Circular Dichroism , Cyclization , Immunoconjugates , Proton Magnetic Resonance Spectroscopy
9.
Drug Discov Today Technol ; 30: 85-89, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30553524

ABSTRACT

Amanitin-based ADCs represent a new class of ADCs using a novel mode of action. This payload introduces a novel mode of action into oncology therapy, the inhibition of RNA Polymerase II. The high potency of the toxin leads to highly efficacious ADCs. The development of the technology around this toxin will be described. These developments support the clinical development of amanitin-based ADCs by using a toxin with a new mode of action and with a favorable therapeutic index. HDP-101 is an Amanitin based ADC directed against BCMA and will be advancing to the clinical phase in 2019.


Subject(s)
Amanitins/chemistry , Antineoplastic Agents/chemistry , Immunoconjugates/chemistry , Humans , Structure-Activity Relationship
10.
Beilstein J Org Chem ; 14: 407-415, 2018.
Article in English | MEDLINE | ID: mdl-29520305

ABSTRACT

RGD-α-amanitin and isoDGR-α-amanitin conjugates were synthesized by joining integrin ligands to α-amanitin via various linkers and spacers. The conjugates were evaluated for their ability to inhibit biotinylated vitronectin binding to the purified αVß3 receptor, retaining good binding affinity, in the same nanomolar range as the free ligands. The antiproliferative activity of the conjugates was evaluated in three cell lines possessing different levels of αVß3 integrin expression: human glioblastoma U87 (αVß3+), human lung carcinoma A549 (αVß3-) and breast adenocarcinoma MDA-MB-468 (αVß3-). In the U87, in the MDA-MB-468, and partly in the A549 cancer cell lines, the cyclo[DKP-isoDGR]-α-amanitin conjugates bearing the lysosomally cleavable Val-Ala linker were found to be slightly more potent than α-amanitin. Apparently, for all these α-amanitin conjugates there is no correlation between the cytotoxicity and the expression of αVß3 integrin. To determine whether the increased cytotoxicity of the cyclo[DKP-isoDGR]-α-amanitin conjugates is governed by an integrin-mediated binding and internalization process, competition experiments were carried out in which the conjugates were tested with U87 (αVß3+, αVß5+, αVß6-, α5ß1+) and MDA-MB-468 (αVß3-, αVß5+, αVß6+, α5ß1-) cells in the presence of excess cilengitide, with the aim of blocking integrins on the cell surface. Using the MDA-MB-468 cell line, a fivefold increase of the IC50 was observed for the conjugates in the presence of excess cilengitide, which is known to strongly bind not only αVß3, but also αVß5, αVß6, and α5ß1. These data indicate that in this case the cyclo[DKP-isoDGR]-α-amanitin conjugates are possibly internalized by a process mediated by integrins different from αVß3 (e.g., αVß5).

11.
Anal Chem ; 86(10): 4642-7, 2014 May 20.
Article in English | MEDLINE | ID: mdl-24559101

ABSTRACT

Measurements of target activation in cells or tissues are key indicators of efficacy during drug development. In contrast to established methods that require reagents and multiple preprocessing steps, reagent-free in situ analysis of engaged drug targets or target-proximal pharmacodynamic signatures in solid tumors remains challenging. Here, we demonstrate that label-free quantification of histone acetylation-specific mass shifts by matrix-assisted laser desorption ionization (MALDI) mass spectrometry biotyping can be used for measurement of cellular potency of histone deacetylase inhibitors in intact cells. Furthermore, we employ MALDI mass spectrometry imaging of these mass shifts to visualize the spatiotemporal distribution of acetylated histones and thus the tumor-selective pharmacodynamic responses in a mouse model of gastrointestinal cancer. Taken together, our study suggests that the monitoring of drug-induced mass shifts in protein ion intensity fingerprints or images may be a powerful analytical tool in pharmacology and drug discovery.


Subject(s)
Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/drug effects , Animals , Cells/enzymology , Drug Discovery/methods , Gastrointestinal Neoplasms/drug therapy , Histone Deacetylase Inhibitors/therapeutic use , Mice , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
12.
Res Sq ; 2024 Jan 11.
Article in English | MEDLINE | ID: mdl-38260385

ABSTRACT

B-cell maturation antigen (BCMA) plays a pathobiologic role in myeloma and is a validated target with five BCMA-specific therapeutics having been approved for relapsed/refractory disease. However, these drugs are not curative, and responses are inferior in patients with molecularly-defined high-risk disease, including those with deletion 17p (del17p) involving the tumor suppressor TP53, supporting the need for further drug development. Del17p has been associated with reduced copy number and gene expression of RNA polymerase II subunit alpha (POLR2A) in other tumor types. We therefore studied the possibility that HDP-101, an anti-BCMA antibody drug conjugate (ADC) with the POLR2A poison α-amanitin could be an attractive agent in myeloma, especially with del17p. HDP-101 reduced viability in myeloma cell lines representing different molecular disease subtypes, and overcame adhesion-mediated and both conventional and novel drug resistance. After confirming that del17p is associated with reduced POLR2A levels in publicly available myeloma patient databases, we engineered TP53 wild-type cells with a TP53 knockout (KO), POLR2A knockdown (KD), or both, the latter to mimic del17p. HDP-101 showed potent anti-myeloma activity against all tested cell lines, and exerted enhanced efficacy against POLR2A KD and dual TP53 KO/POLR2A KD cells. Mechanistic studies showed HDP-101 up-regulated the unfolded protein response, activated apoptosis, and induced immunogenic cell death. Notably, HDP-101 impacted CD138-positive but not-negative primary cells, showed potent efficacy against aldehyde dehydrogenase-positive clonogenic cells, and eradicated myeloma in an in vivo cell line-derived xenograft (CDX). Interestingly, in the CDX model, prior treatment with HDP-101 precluded subsequent engraftment on tumor cell line rechallenge in a manner that appeared to be dependent in part on natural killer cells and macrophages. Finally, HDP-101 was superior to the BCMA-targeted ADC belantamab mafodotin against cell lines and primary myeloma cells in vitro, and in an in vivo CDX. Together, the data support the rationale for translation of HDP-101 to the clinic, where it is now undergoing Phase I trials, and suggest that it could emerge as a more potent ADC for myeloma with especially interesting activity against the high-risk del17p myeloma subtype.

13.
Eur J Immunol ; 42(8): 2100-8, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22585398

ABSTRACT

The fast production of effector cytokines, such as IL-2, is essential for the autocrine function in the rapid activation of naive CD4(+) T cells. Here, we show that the microRNA (miRNA) pathway plays an important role in the posttranscriptional regulation of proinflammatory cytokines in human CD4(+) T cells. miRNAs are small noncoding molecules that act as posttranscriptional regulators of gene expression by binding to the 3' untranslated region of target mRNAs. Using microarray and deep sequencing approaches, we detected an increase in the abundance of miR-9 in activated human CD4(+) T cells. To determine the impact of miR-9 on immune responses, we analyzed its effect on two putative target genes, PRDM1, which encodes for the transcription factor Blimp-1 (B lymphocyte-induced maturation protein-1), and Bcl-6 (B cell lymphoma-6 protein). Suppression of miR-9 led to increased expression of PRDM1 and Bcl-6, which subsequently resulted in diminished secretion of IL-2 and IFN-γ. Our data provide evidence that the abundance of Blimp-1, and consequently the secretion of proinflammatory cytokines, is regulated in two ways: (i) transcriptional regulation by activation of CD4(+) T cells and (ii) posttranscriptional regulation by enhanced miR-9 expression.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , DNA-Binding Proteins/biosynthesis , Interleukin-2/biosynthesis , Lymphocyte Activation , MicroRNAs/metabolism , Repressor Proteins/biosynthesis , CD4-Positive T-Lymphocytes/metabolism , Gene Expression Regulation , Humans , Interferon-gamma/biosynthesis , Positive Regulatory Domain I-Binding Factor 1 , Proto-Oncogene Proteins c-bcl-6
14.
J Med Chem ; 64(7): 4117-4129, 2021 04 08.
Article in English | MEDLINE | ID: mdl-33755471

ABSTRACT

Herein we describe the design and biological evaluation of a novel antitumor therapeutic platform that combines the most favorable properties of small-molecule drug conjugates (SMDCs) and antibody drug conjugates (ADCs). Although the small size of SMDCs, compared to ADCs, is an appealing feature for their application in the treatment of solid tumors, SMDCs usually suffer from poor pharmacokinetics, which severely limits their therapeutic efficacy. To overcome this limitation, in this proof-of-concept study we grafted an α-amanitin-based SMDC that targets prostate cancer cells onto an immunoglobulin Fc domain via a two-step "program and arm" chemoenzymatic strategy. We demonstrated the superior pharmacokinetic properties and therapeutic efficacy of the resulting Fc-SMDC over the SMDC in a prostate cancer xenograft mouse model. This approach may provide a general strategy toward effective antitumor therapeutics combining small size with pharmacokinetic properties close to those of an ADC.


Subject(s)
Alpha-Amanitin/therapeutic use , Antineoplastic Agents/therapeutic use , Immunoconjugates/therapeutic use , Immunoglobulin Fc Fragments/therapeutic use , Prostatic Neoplasms/drug therapy , Alpha-Amanitin/chemistry , Alpha-Amanitin/pharmacokinetics , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacokinetics , Cell Line, Tumor , Humans , Immunoconjugates/immunology , Immunoconjugates/pharmacokinetics , Immunoglobulin Fc Fragments/immunology , Immunoglobulin G/immunology , Immunoglobulin G/therapeutic use , Male , Mice, SCID , Prostatic Neoplasms/immunology , Xenograft Model Antitumor Assays
15.
Sci Transl Med ; 13(580)2021 02 10.
Article in English | MEDLINE | ID: mdl-33568521

ABSTRACT

The clinical challenge for treating HER2 (human epidermal growth factor receptor 2)-low breast cancer is the paucity of actionable drug targets. HER2-targeted therapy often has poor clinical efficacy for this disease due to the low level of HER2 protein on the cancer cell surface. We analyzed breast cancer genomics in the search for potential drug targets. Heterozygous loss of chromosome 17p is one of the most frequent genomic events in breast cancer, and 17p loss involves a massive deletion of genes including the tumor suppressor TP53 Our analyses revealed that 17p loss leads to global gene expression changes and reduced tumor infiltration and cytotoxicity of T cells, resulting in immune evasion during breast tumor progression. The 17p deletion region also includes POLR2A, a gene encoding the catalytic subunit of RNA polymerase II that is essential for cell survival. Therefore, breast cancer cells with heterozygous loss of 17p are extremely sensitive to the inhibition of POLR2A via a specific small-molecule inhibitor, α-amanitin. Here, we demonstrate that α-amanitin-conjugated trastuzumab (T-Ama) potentiated the HER2-targeted therapy and exhibited superior efficacy in treating HER2-low breast cancer with 17p loss. Moreover, treatment with T-Ama induced immunogenic cell death in breast cancer cells and, thereby, delivered greater efficacy in combination with immune checkpoint blockade therapy in preclinical HER2-low breast cancer models. Collectively, 17p loss not only drives breast tumorigenesis but also confers therapeutic vulnerabilities that may be used to develop targeted precision immunotherapy.


Subject(s)
Breast Neoplasms , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Cell Line, Tumor , Female , Humans , Immunotherapy , Receptor, ErbB-2/genetics , Trastuzumab
16.
Mol Cancer Ther ; 20(2): 367-378, 2021 02.
Article in English | MEDLINE | ID: mdl-33298585

ABSTRACT

Despite major treatment advances in recent years, patients with multiple myeloma inevitably relapse. The RNA polymerase II complex has been identified as a promising therapeutic target in both proliferating and dormant cancer cells. Alpha-amanitin, a toxin so far without clinical application due to high liver toxicity, specifically inhibits this complex. Here, we describe the development of HDP-101, an anti-B-cell maturation antigen (BCMA) antibody conjugated with an amanitin derivative. HDP-101 displayed high efficacy against both proliferating and resting myeloma cells in vitro, sparing BCMA-negative cells. In subcutaneous and disseminated murine xenograft models, HDP-101 induced tumor regression at low doses, including durable complete remissions after a single intravenous dose. In cynomolgus monkeys, HDP-101 was well tolerated with a promising therapeutic index. In conclusion, HDP-101 safely and selectively delivers amanitin to myeloma cells and provides a novel therapeutic approach to overcome drug resistance in this disease.


Subject(s)
Amanitins/therapeutic use , Cell Death/drug effects , Enzyme Inhibitors/therapeutic use , Immunoconjugates/therapeutic use , Multiple Myeloma/drug therapy , Amanitins/pharmacology , Animals , Cell Proliferation , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Female , Humans , Immunoconjugates/pharmacology , Mice , Mice, SCID
17.
Blood Adv ; 4(13): 2899-2911, 2020 07 14.
Article in English | MEDLINE | ID: mdl-32589729

ABSTRACT

Cancer cells commonly develop resistance to immunotherapy by loss of antigen expression. Combinatorial treatments that increase levels of the target antigen on the surface of cancer cells have the potential to restore efficacy to immunotherapy. Here, we use our CRISPR interference- and CRISPR activation-based functional genomics platform to systematically identify pathways controlling cell surface expression of the multiple myeloma immunotherapy antigen B-cell maturation antigen (BCMA). We discovered that pharmacologic inhibition of HDAC7 and the Sec61 complex increased cell surface BCMA, including in primary patient cells. Pharmacologic Sec61 inhibition enhanced the antimyeloma efficacy of a BCMA-targeted antibody-drug conjugate. A CRISPR interference chimeric antigen receptor T cells (CAR-T cells) coculture screen enabled us to identify both antigen-dependent and antigen-independent mechanisms controlling response of myeloma cells to BCMA-targeted CAR-T cells. Thus, our study shows the potential of CRISPR screens to uncover mechanisms controlling response of cancer cells to immunotherapy and to suggest potential combination therapies.


Subject(s)
B-Cell Maturation Antigen , Multiple Myeloma , B-Cell Maturation Antigen/genetics , Clustered Regularly Interspaced Short Palindromic Repeats , Humans , Immunotherapy , Immunotherapy, Adoptive , Multiple Myeloma/genetics , Multiple Myeloma/therapy , T-Lymphocytes
18.
Immunology ; 127(1): 40-9, 2009 May.
Article in English | MEDLINE | ID: mdl-18800988

ABSTRACT

Pemphigus vulgaris (PV) is an autoimmune bullous skin disorder associated with autoantibodies against desmoglein (Dsg) 3. An imbalance of type 1 regulatory T (Tr1) cells and T helper type 2 (Th2) cells specific for Dsg3 may be critical for the loss of tolerance against Dsg3 in PV. Within the population of Dsg3-responsive, interleukin (IL)-10-secreting Tr1 cell clones, two major subpopulations were identified and sorted by fluorescence-activated cell sorting (FACS) based on their size and granularity. Upon in vitro culture, the larger subpopulation differentiated back into the two former subpopulations of the Tr1 cell clones, while the smaller subpopulation died within 2 weeks. The smaller subpopulation of the Tr1 cell clones was characterized by the expression of Foxp3, the secretion of IL-10, transforming growth factor (TGF)-beta and IL-5 upon stimulation with Dsg3, a proliferative response to IL-2 but not to Dsg3 or mitogenic stimuli, and an inhibitory effect on the proliferative response of Dsg3-responsive Th clones in a Dsg3-specific manner. In contrast, the larger subpopulation showed a Th-like phenotype, lacking Foxp3, cytotoxic T-lymphocyte antigen 4 (CTLA4) and glucocorticoid-induced tumour necrosis factor receptor (GITR) expression and IL-2 secretion, and did not mount a proliferative response to Dsg3 and mitogenic stimuli. The two Tr1 subpopulations showed expression of identical T-cell receptor (TCR) V beta chains which varied among the PV patients studied. Upon inhibition of Foxp3, the smaller Tr1 subpopulation developed a proliferate response to Dsg3 and mitogenic stimuli, no longer suppressed Dsg3-specific Th cells, lost expression of GITR and CTLA4 and secreted IL-2. Thus, our observations suggest a distinct relationship between Dsg3-specific Tr1 and Th-like cells which may be critical for the continuous generation and survival of Dsg3-specific Tr1 cells.


Subject(s)
Desmoglein 3/immunology , Forkhead Transcription Factors/metabolism , Pemphigus/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Regulatory/immunology , Autoimmunity/immunology , Cell Proliferation , Cells, Cultured , Cytokines/biosynthesis , Forkhead Transcription Factors/genetics , Humans , Immune Tolerance/immunology , Immunophenotyping , Lymphocyte Activation/immunology , Oligonucleotides, Antisense/immunology , Reverse Transcriptase Polymerase Chain Reaction/methods
19.
Eur J Pediatr ; 168(7): 819-24, 2009 Jul.
Article in English | MEDLINE | ID: mdl-18936971

ABSTRACT

UNLABELLED: In spite of the lack of evidence for its efficacy, and of sporadic reports of severe adverse events, codeine is still widely used as an antitussive agent in children. A 3-year-old boy (twin 1) was found lying in vomit and apnoeic at night; he was resuscitated and immediately transferred to our paediatric intensive care unit (PICU). Two and a half hours later, his twin brother (twin 2) was found dead in his bed at home. Twin 1 required mechanical ventilation for 3 days, but he eventually made a full recovery; autopsy in twin 2 showed massive aspiration of gastric content. History revealed that the monozygotic twins had an upper respiratory tract infection for several days and had both been given codeine at a dose of "10 drops per day" by their mother. The blood of both twins was found to contain high levels of codeine and its metabolites. The weight of "10 drops" was determined experimentally and was found to range from 494 to 940 mg. Thus, the highest possible dose given by mother was 23.5 mg of codeine instead of the recommended 10 mg. The twins had identical CYP2D6 gene polymorphisms corresponding to the "extensive metaboliser" type. CONCLUSIONS: Because of the variability of drop size drug dosage, dosage "by drops" is unprecise and may result in accidental overdose. The combination of repeated overdosing and extensive metabolism to morphine is likely to have caused apnoea in these twins. These cases illustrate the danger of codeine as an antitussive in young children.


Subject(s)
Antitussive Agents/administration & dosage , Antitussive Agents/adverse effects , Codeine/administration & dosage , Codeine/adverse effects , Respiratory Aspiration/therapy , Twins , Vomiting/complications , Administration, Oral , Child, Preschool , Cytochrome P-450 CYP2D6/genetics , Drug Overdose/complications , Fatal Outcome , Genotype , Humans , Male , Polymorphism, Genetic , Respiration, Artificial/methods , Respiratory Aspiration/etiology , Respiratory Aspiration/physiopathology , Treatment Outcome , Vomiting/chemically induced
20.
J Clin Invest ; 115(3): 673-9, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15719070

ABSTRACT

Blockade of prostaglandin (PG) production by COX inhibitors is the treatment of choice for inflammatory pain but is also prone to severe side effects. Identification of signaling elements downstream of COX inhibition, particularly of PG receptor subtypes responsible for pain sensitization (hyperalgesia), provides a strategy for better-tolerated analgesics. Here, we have identified PGE2 receptors of the EP2 receptor subtype as key signaling elements in spinal inflammatory hyperalgesia. Mice deficient in EP2 receptors (EP2-/- mice) completely lack spinal PGE2-evoked hyperalgesia. After a peripheral inflammatory stimulus, EP2-/- mice exhibit only short-lasting peripheral hyperalgesia but lack a second sustained hyperalgesic phase of spinal origin. Electrophysiological recordings identify diminished synaptic inhibition of excitatory dorsal horn neurons as the dominant source of EP2 receptor-dependent hyperalgesia. Our results thus demonstrate that inflammatory hyperalgesia can be treated by targeting of a single PG receptor subtype and provide a rational basis for new analgesic strategies going beyond COX inhibition.


Subject(s)
Hyperalgesia/immunology , Hyperalgesia/metabolism , Receptors, Prostaglandin E/metabolism , Spinal Cord/physiology , Animals , Behavior, Animal/physiology , Cyclooxygenase 2 , Cyclooxygenase 2 Inhibitors , Cyclooxygenase Inhibitors , Female , Glycine/metabolism , Hot Temperature , In Vitro Techniques , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/metabolism , Pain/metabolism , Pain Measurement , Patch-Clamp Techniques , Prostaglandin-Endoperoxide Synthases/metabolism , Prostaglandins/administration & dosage , Prostaglandins/metabolism , Receptors, Prostaglandin E/genetics , Receptors, Prostaglandin E, EP2 Subtype , Signal Transduction/physiology , Spinal Cord/cytology , Synaptic Transmission
SELECTION OF CITATIONS
SEARCH DETAIL