Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
N Engl J Med ; 390(5): 432-441, 2024 Feb 01.
Article in English | MEDLINE | ID: mdl-38294975

ABSTRACT

BACKGROUND: Hereditary angioedema is a rare genetic disease that leads to severe and unpredictable swelling attacks. NTLA-2002 is an in vivo gene-editing therapy based on clustered regularly interspaced short palindromic repeats (CRISPR)-CRISPR-associated protein 9. NTLA-2002 targets the gene encoding kallikrein B1 (KLKB1), with the goal of lifelong control of angioedema attacks after a single dose. METHODS: In this phase 1 dose-escalation portion of a combined phase 1-2 trial of NTLA-2002 in adults with hereditary angioedema, we administered NTLA-2002 at a single dose of 25 mg, 50 mg, or 75 mg. The primary end points were the safety and side-effect profile of NTLA-2002 therapy. Secondary and exploratory end points included pharmacokinetics, pharmacodynamics, and clinical efficacy determined on the basis of investigator-confirmed angioedema attacks. RESULTS: Three patients received 25 mg of NTLA-2002, four received 50 mg, and three received 75 mg. At all dose levels, the most common adverse events were infusion-related reactions and fatigue. No dose-limiting toxic effects, serious adverse events, grade 3 or higher adverse events, or clinically important laboratory findings were observed after the administration of NTLA-2002. Dose-dependent reductions in the total plasma kallikrein protein level were observed between baseline and the latest assessment, with a mean percentage change of -67% in the 25-mg group, -84% in the 50-mg group, and -95% in the 75-mg group. The mean percentage change in the number of angioedema attacks per month between baseline and weeks 1 through 16 (primary observation period) was -91% in the 25-mg group, -97% in the 50-mg group, and -80% in the 75-mg group. Among all the patients, the mean percentage change in the number of angioedema attacks per month from baseline through the latest assessment was -95%. CONCLUSIONS: In this small study, a single dose of NTLA-2002 led to robust, dose-dependent, and durable reductions in total plasma kallikrein levels, and no severe adverse events were observed. In exploratory analyses, reductions in the number of angioedema attacks per month were observed at all dose levels. (Funded by Intellia Therapeutics; ClinicalTrials.gov number, NCT05120830.).


Subject(s)
Angioedemas, Hereditary , CRISPR-Cas Systems , Gene Editing , Adult , Humans , Angioedema , Angioedemas, Hereditary/blood , Angioedemas, Hereditary/drug therapy , Angioedemas, Hereditary/genetics , Complement C1 Inhibitor Protein/therapeutic use , Dose-Response Relationship, Drug , Gene Editing/methods , Plasma Kallikrein/genetics , Treatment Outcome
2.
Am J Hematol ; 96(2): 251-257, 2021 02 01.
Article in English | MEDLINE | ID: mdl-33180328

ABSTRACT

ß-thalassemias result from mutations in ß-globin, causing ineffective erythropoiesis and secondary iron overload due to inappropriately low levels of the iron regulatory hormone hepcidin. Mutations in transferrin receptor 2 (TFR2) lead to hereditary hemochromatosis (HH) as a result of inappropriately increased iron uptake from the diet, also due to improperly regulated hepcidin. TFR2 is also thought to be required for efficient erythropoiesis through its interaction with the erythropoietin receptor in erythroid progenitors. Transmembrane serine protease 6 (TMPRSS6), a membrane serine protease expressed selectively in the liver, participates in regulating hepcidin production in response to iron stores by cleaving hemojuvelin (HJV). We have previously demonstrated that inhibiting TMPRSS6 expression with a hepatocyte-specific siRNA formulation, induces hepcidin, mitigates anemia, and reduces iron overload in murine models of ß-thalassemia intermedia and HH. Here, we demonstrate that Tmprss6 siRNA treatment of double mutant Tfr2Y245X/Y245X HH Hbbth3/+ thalassemic mice induces hepcidin and diminishes tissue and serum iron levels. Importantly, treated double mutant animals produce more mature red blood cells and have a nearly 50% increase in hemoglobin compared to untreated ß-thalassemic mice. Furthermore, we also show that treatment of Tfr2Y245X/Y245X HH mice leads to increased hepcidin expression and reduced total body iron burden. These data indicate that siRNA suppression of Tmprss6, in conjunction with the targeting of TFR2, may be superior to inhibiting Tmprss6 alone in the treatment of the anemia and secondary iron loading in ß-thalassemia intermedia and may be useful as a method of suppressing the primary iron overload in TFR2-related (type 3) hereditary hemochromatosis.


Subject(s)
Hemochromatosis/metabolism , Iron Deficiencies , Receptors, Transferrin/deficiency , beta-Thalassemia/metabolism , Amino Acid Substitution , Animals , Disease Models, Animal , GPI-Linked Proteins/genetics , GPI-Linked Proteins/metabolism , Hemochromatosis/genetics , Hemochromatosis/pathology , Hemochromatosis Protein/genetics , Hemochromatosis Protein/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Transgenic , Mutation, Missense , Receptors, Transferrin/metabolism , Serine Endopeptidases/genetics , Serine Endopeptidases/metabolism , beta-Thalassemia/genetics , beta-Thalassemia/pathology
3.
Am J Hematol ; 95(5): 492-496, 2020 05.
Article in English | MEDLINE | ID: mdl-31990410

ABSTRACT

Reduced ferrochelatase activity in erythropoietic protoporphyria (EPP) causes the accumulation of protoporphyrin IX (PPIX) leading to acute cutaneous photosensitivity and liver injury. Many EPP patients also have a mild hypochromic, microcytic anemia and iron deficiency. Iron deficiency can lead to decreased PPIX accumulation in another erythropoietic porphyria, congenital erythropoietic porphyria (CEP). Expression of the iron regulatory peptide hepcidin is negatively regulated by the serine protease TMPRSS6. Hepcidin induction by siRNA-mediated inhibition of TMPRSS6 expression reduces iron availability and induces iron deficiency. To interrogate the therapeutic potential of iron deficiency to modify EPP, we treated an ethylnitrosourea-induced mouse model of EPP, Fech m1Pas , with a GalNAc-conjugated Tmprss6 siRNA and PPIX levels, anemia and iron parameters were monitored. The GalNAc-RNAi therapeutic reduces Tmprss6 expression and induces mild iron deficiency in Fech m1Pas animals. However, decreases in erythrocyte PPIX levels and liver PPIX accumulation were not seen. These results indicate short-term induction of iron deficiency, at least in a murine model of EPP, does not lead to decreased PPIX production.


Subject(s)
Anemia, Iron-Deficiency/etiology , Protoporphyria, Erythropoietic/complications , Animals , Disease Models, Animal , Female , Humans , Mice , Phenotype
4.
Am J Hematol ; 93(6): 745-750, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29498084

ABSTRACT

Diminished ß-globin synthesis in ß-thalassemia is associated with ineffective erythropoiesis, leading to secondary iron overload caused by inappropriately low levels of hepcidin and to splenomegaly in the symptomatic thalassemias. Splenectomy is often employed in patients with ß-thalassemia to reduce hemolysis. Expression of the iron regulatory peptide hormone hepcidin is repressed by the serine protease TMPRSS6. Hepcidin induction by RNAi-mediated inhibition of TMPRSS6 expression reduces iron overload and mitigates anemia in murine models of ß-thalassemia intermedia. To interrogate the efficacy of RNAi-mediated reduction of Tmprss6 in splenectomized ß-thalassemia, splenectomized ß-thalassemic Hbbth3/+ animals were treated with a GalNAc-conjugated siRNA targeting Tmprss6 (GalNAc-Tmprss6) and their hematological and iron parameters monitored. We demonstrate that treatment with GalNAc-Tmprss6 significantly diminishes Tmprss6 expression and appropriately elevates hepcidin expression in splenectomized Hbbth3/+ animals. Similar to unsplenectomized animals, treated animals have markedly improved anemia due to diminished ineffective erythropoiesis and reduced iron loading in both serum and tissue. These results suggest that RNAi-mediated reduction of Tmprss6 may have positive outcomes even in splenectomized ß-thalassemia patients.


Subject(s)
Anemia/prevention & control , Iron Overload/prevention & control , Membrane Proteins/antagonists & inhibitors , RNA Interference/physiology , Splenectomy , beta-Thalassemia/complications , Acetylgalactosamine/chemistry , Animals , Disease Models, Animal , Erythropoiesis , Hepcidins , Liver/metabolism , Membrane Proteins/chemistry , Mice , Serine Endopeptidases/chemistry
5.
Mol Ther ; 26(1): 105-114, 2018 01 03.
Article in English | MEDLINE | ID: mdl-28988716

ABSTRACT

The hepatocyte-specific asialoglycoprotein receptor (ASGPR) is an ideal candidate for targeted drug delivery to the liver due to its high capacity for substrate clearance from circulation together with its well-conserved expression and function across species. The development of GalNAc-siRNA conjugates, in which a synthetic triantennary N-acetylgalactosamine-based ligand is conjugated to chemically modified siRNA, has enabled efficient, ASGPR-mediated delivery to hepatocytes. To investigate the potential impact of variations in receptor expression on the efficiency of GalNAc-siRNA conjugate delivery, we evaluated the pharmacokinetics and pharmacodynamics of GalNAc-siRNA conjugates in multiple pre-clinical models with reduced receptor expression. Despite greater than 50% reduction in ASGPR levels, GalNAc conjugate activity was retained, suggesting that the remaining receptor capacity was sufficient to mediate efficient uptake of potent GalNAc-siRNAs at pharmacologically relevant dose levels. Collectively, our data support a broad application of the GalNAc-siRNA technology for hepatic targeting, including disease states where ASGPR expression may be reduced.


Subject(s)
Acetylgalactosamine , Asialoglycoprotein Receptor/genetics , Gene Expression Regulation , RNA Interference , RNA, Small Interfering/genetics , Acetylgalactosamine/chemistry , Animals , Asialoglycoprotein Receptor/chemistry , Asialoglycoprotein Receptor/metabolism , Disease Models, Animal , Drug Carriers , Drug Delivery Systems , Drug Evaluation, Preclinical , Female , Gene Silencing , Hepatocytes/metabolism , Humans , Liver Cirrhosis/genetics , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , Mice , Mice, Knockout , Protein Subunits/genetics , Protein Subunits/metabolism , RNA, Small Interfering/chemistry
6.
Amyloid ; 23(2): 109-18, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27033334

ABSTRACT

ATTR amyloidosis is a systemic, debilitating and fatal disease caused by transthyretin (TTR) amyloid accumulation. RNA interference (RNAi) is a clinically validated technology that may be a promising approach to the treatment of ATTR amyloidosis. The vast majority of TTR, the soluble precursor of TTR amyloid, is expressed and synthesized in the liver. RNAi technology enables robust hepatic gene silencing, the goal of which would be to reduce systemic levels of TTR and mitigate many of the clinical manifestations of ATTR that arise from hepatic TTR expression. To test this hypothesis, TTR-targeting siRNAs were evaluated in a murine model of hereditary ATTR amyloidosis. RNAi-mediated silencing of hepatic TTR expression inhibited TTR deposition and facilitated regression of existing TTR deposits in pathologically relevant tissues. Further, the extent of deposit regression correlated with the level of RNAi-mediated knockdown. In comparison to the TTR stabilizer, tafamidis, RNAi-mediated TTR knockdown led to greater regression of TTR deposits across a broader range of affected tissues. Together, the data presented herein support the therapeutic hypothesis behind TTR lowering and highlight the potential of RNAi in the treatment of patients afflicted with ATTR amyloidosis.


Subject(s)
Amyloid Neuropathies, Familial/therapy , Liver/metabolism , Prealbumin/antagonists & inhibitors , RNA, Messenger/antagonists & inhibitors , RNA, Small Interfering/administration & dosage , Amyloid Neuropathies, Familial/genetics , Amyloid Neuropathies, Familial/metabolism , Amyloid Neuropathies, Familial/pathology , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Benzoxazoles/pharmacology , Disease Models, Animal , Drug Evaluation, Preclinical , Female , Gene Expression , Humans , Liver/pathology , Macaca fascicularis , Male , Mice , Mice, Transgenic , Prealbumin/genetics , Prealbumin/metabolism , RNA Interference , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/pharmacokinetics
7.
Am J Hematol ; 90(4): 310-3, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25557851

ABSTRACT

ß-thalassemias result from diminished ß-globin synthesis and are associated with ineffective erythropoiesis and secondary iron overload caused by inappropriately low levels of the iron regulatory hormone hepcidin. The serine protease TMPRSS6 attenuates hepcidin production in response to iron stores. Hepcidin induction reduces iron overload and mitigates anemia in murine models of ß-thalassemia intermedia. To further interrogate the efficacy of an RNAi-therapeutic downregulating Tmprss6, ß-thalassemic Hbb(th3/+) animals on an iron replete, an iron deficient, or an iron replete diet also containing the iron chelator deferiprone were treated with Tmprss6 siRNA. We demonstrate that the total body iron burden is markedly improved in Hbb(th3/+) animals treated with siRNA and chelated with oral deferiprone, representing a significant improvement compared to either compound alone. These data indicate that siRNA suppression of Tmprss6, in conjunction with oral iron chelation therapy, may prove superior for treatment of anemia and secondary iron loading seen in ß-thalassemia intermedia.


Subject(s)
Iron Chelating Agents/therapeutic use , Iron/metabolism , Membrane Proteins/genetics , Pyridones/therapeutic use , RNA Interference , Serine Endopeptidases/genetics , beta-Thalassemia/drug therapy , Administration, Oral , Animals , Combined Modality Therapy , Deferiprone , Disease Models, Animal , Drug Carriers/chemistry , Female , Hepcidins/biosynthesis , Hepcidins/blood , Iron/blood , Iron Chelating Agents/administration & dosage , Mice , Nanoparticles/chemistry , Pyridones/administration & dosage , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/genetics , beta-Thalassemia/genetics , beta-Thalassemia/metabolism
8.
Haematologica ; 100(2): 167-77, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25425686

ABSTRACT

Mice have been essential for distinguishing the role of hepcidin in iron homeostasis. Currently, investigators monitor levels of murine hepatic hepcidin-1 mRNA as a surrogate marker for the bioactive hepcidin protein itself. Here, we describe and validate a competitive, enzyme-linked immunosorbent assay that quantifies hepcidin-1 in mouse serum and urine. The assay exhibits a biologically relevant lower limit of detection, high precision, and excellent linearity and recovery. We also demonstrate correlation between serum and urine hepcidin-1 values and validate the competitive enzyme-linked immunosorbent assay by analyzing plasma hepcidin response of mice to physiological challenges, including iron deficiency, iron overload, acute blood loss, and inflammation. Furthermore, we analyze multiple murine genetic models of iron dysregulation, including ß-thalassemia intermedia (Hbb(th3/+)), hereditary hemochromatosis (Hfe(-/-), Hjv(-/-), and Tfr2(Y245X/Y245X)), hypotransferrinemia (Trf(hpx/hpx)), heterozygous transferrin receptor 1 deficiency (Tfrc(+/-)) and iron refractory iron deficiency anemia (Tmprss6(-/-) and Tmprss6(hem8/hem8)). Novel compound iron metabolism mutants were also phenotypically characterized here for the first time. We demonstrate that serum hepcidin concentrations correlate with liver hepcidin mRNA expression, transferrin saturation and non-heme liver iron. In some circumstances, serum hepcidin-1 more accurately predicts iron parameters than hepcidin mRNA, and distinguishes smaller, statistically significant differences between experimental groups.


Subject(s)
Disease Models, Animal , Enzyme-Linked Immunosorbent Assay/methods , Hepcidins/metabolism , Homeostasis/physiology , Iron/administration & dosage , Liver/metabolism , Anemia, Iron-Deficiency/genetics , Anemia, Iron-Deficiency/metabolism , Anemia, Iron-Deficiency/pathology , Animals , Cells, Cultured , Female , Hemochromatosis/genetics , Hemochromatosis/metabolism , Hemochromatosis/pathology , Hepcidins/genetics , Homeostasis/drug effects , Humans , Inflammation/chemically induced , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Iron/metabolism , Iron Overload/genetics , Iron Overload/metabolism , Iron Overload/pathology , Lipopolysaccharides/toxicity , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , beta-Thalassemia/genetics , beta-Thalassemia/metabolism , beta-Thalassemia/pathology
9.
N Engl J Med ; 369(9): 819-29, 2013 Aug 29.
Article in English | MEDLINE | ID: mdl-23984729

ABSTRACT

BACKGROUND: Transthyretin amyloidosis is caused by the deposition of hepatocyte-derived transthyretin amyloid in peripheral nerves and the heart. A therapeutic approach mediated by RNA interference (RNAi) could reduce the production of transthyretin. METHODS: We identified a potent antitransthyretin small interfering RNA, which was encapsulated in two distinct first- and second-generation formulations of lipid nanoparticles, generating ALN-TTR01 and ALN-TTR02, respectively. Each formulation was studied in a single-dose, placebo-controlled phase 1 trial to assess safety and effect on transthyretin levels. We first evaluated ALN-TTR01 (at doses of 0.01 to 1.0 mg per kilogram of body weight) in 32 patients with transthyretin amyloidosis and then evaluated ALN-TTR02 (at doses of 0.01 to 0.5 mg per kilogram) in 17 healthy volunteers. RESULTS: Rapid, dose-dependent, and durable lowering of transthyretin levels was observed in the two trials. At a dose of 1.0 mg per kilogram, ALN-TTR01 suppressed transthyretin, with a mean reduction at day 7 of 38%, as compared with placebo (P=0.01); levels of mutant and nonmutant forms of transthyretin were lowered to a similar extent. For ALN-TTR02, the mean reductions in transthyretin levels at doses of 0.15 to 0.3 mg per kilogram ranged from 82.3 to 86.8%, with reductions of 56.6 to 67.1% at 28 days (P<0.001 for all comparisons). These reductions were shown to be RNAi-mediated. Mild-to-moderate infusion-related reactions occurred in 20.8% and 7.7% of participants receiving ALN-TTR01 and ALN-TTR02, respectively. CONCLUSIONS: ALN-TTR01 and ALN-TTR02 suppressed the production of both mutant and nonmutant forms of transthyretin, establishing proof of concept for RNAi therapy targeting messenger RNA transcribed from a disease-causing gene. (Funded by Alnylam Pharmaceuticals; ClinicalTrials.gov numbers, NCT01148953 and NCT01559077.).


Subject(s)
Amyloid Neuropathies, Familial/therapy , Prealbumin/genetics , RNA, Small Interfering/therapeutic use , Adolescent , Adult , Amyloid Neuropathies, Familial/genetics , Animals , Dose-Response Relationship, Drug , Female , Humans , Liposomes , Macaca fascicularis , Male , Nanocapsules , Prealbumin/metabolism , RNA, Small Interfering/administration & dosage , Young Adult
10.
Mol Ther ; 20(8): 1582-9, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22850721

ABSTRACT

Therapeutics based on RNA interference (RNAi) have emerged as a potential new class of drugs for treating human disease by silencing the target messenger RNA (mRNA), thereby reducing levels of the corresponding pathogenic protein. The major challenge for RNAi therapeutics is the development of safe delivery vehicles for small interfering RNAs (siRNAs). We previously showed that cholesterol-conjugated siRNAs (chol-siRNA) associate with plasma lipoprotein particles and distribute primarily to the liver after systemic administration to mice. We further demonstrated enhancement of silencing by administration of chol-siRNA pre-associated with isolated high-density lipoprotein (HDL) or low-density lipoprotein (LDL). In this study, we investigated mimetic lipoprotein particle prepared from recombinant apolipoprotein A1 (apoA) and apolipoprotein E3 (apoE) as a delivery vehicle for chol-siRNAs. We show that apoE-containing particle (E-lip) is highly effective in functional delivery of chol-siRNA to mouse liver. E-lip delivery was found to be considerably more potent than apoA-containing particle (A-lip). Furthermore, E-lip-mediated delivery was not significantly affected by high endogenous levels of plasma LDL. These results demonstrate that E-lip has substantial potential as delivery vehicles for lipophilic conjugates of siRNAs.


Subject(s)
Lipoproteins/administration & dosage , Lipoproteins/chemistry , RNA, Small Interfering/administration & dosage , Animals , Apolipoprotein A-I/administration & dosage , Apolipoprotein A-I/chemistry , Apolipoproteins E/administration & dosage , Apolipoproteins E/chemistry , Lipoproteins, HDL/administration & dosage , Lipoproteins, HDL/chemistry , Lipoproteins, LDL/administration & dosage , Lipoproteins, LDL/chemistry , Male , Mice , Mice, Inbred C57BL , RNA Interference/physiology , RNA, Small Interfering/genetics
11.
Mol Ther ; 18(7): 1357-64, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20461061

ABSTRACT

Lipid nanoparticles (LNPs) have proven to be highly efficient carriers of short-interfering RNAs (siRNAs) to hepatocytes in vivo; however, the precise mechanism by which this efficient delivery occurs has yet to be elucidated. We found that apolipoprotein E (apoE), which plays a major role in the clearance and hepatocellular uptake of physiological lipoproteins, also acts as an endogenous targeting ligand for ionizable LNPs (iLNPs), but not cationic LNPs (cLNPs). The role of apoE was investigated using both in vitro studies employing recombinant apoE and in vivo studies in wild-type and apoE(-/-) mice. Receptor dependence was explored in vitro and in vivo using low-density lipoprotein receptor (LDLR(-/-))-deficient mice. As an alternative to endogenous apoE-based targeting, we developed a targeting approach using an exogenous ligand containing a multivalent N-acetylgalactosamine (GalNAc)-cluster, which binds with high affinity to the asialoglycoprotein receptor (ASGPR) expressed on hepatocytes. Both apoE-based endogenous and GalNAc-based exogenous targeting appear to be highly effective strategies for the delivery of iLNPs to liver.


Subject(s)
RNA Interference/physiology , Animals , Apolipoproteins E/genetics , Apolipoproteins E/metabolism , Asialoglycoprotein Receptor/metabolism , Female , HeLa Cells , Hepatocytes/metabolism , Humans , Ligands , Mice , Mice, Inbred C57BL , Nanoparticles/chemistry , Receptors, LDL/genetics , Receptors, LDL/metabolism
12.
Lung Cancer ; 70(2): 174-9, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20207442

ABSTRACT

BACKGROUND: Thirty percent of newly diagnosed NSCLC patients present with synchronous brain metastases, most of whom are treated with whole brain radiation. Systemic chemotherapy is usually avoided during WBRT due to concerns regarding toxicity. However, concurrent administration of targeted agents, such as Erlotinib, during WBRT may address systemic disease without causing toxicity. We report our institutional data on outcomes and toxicities with this treatment approach. MATERIALS AND METHODS: Medical records of patients with newly diagnosed NSCLC and brain metastases receiving concurrent WBRT and Erlotinib treatment were reviewed. Radiographic response to therapy and toxicities were analyzed. RESULT: Eight patients were identified and 7 were evaluable for response. All patients had intracranial disease control. In the extracranial sites, 3 (37.5%, intent-to-treat) showed partial response (PR), 2 (25%) had stable disease (SD), 1 (12.5%) had progression (PD) and 1 (12.5%) had new air space disease obscuring tumor response assessment. Among the three responders, two were female never smokers, while one was a female current smoker. Unanticipated grade 3 hepatotoxitity, hyponatremia, mental status changes, grade 3 and 4 thrombocytopenia, and grade 4 neutropenia with sepsis were observed. Three deaths occurred without clear signs of disease progression: one from neutropenic sepsis, one from wide spread air space disease, and one from neurologic deterioration. CONCLUSION: Our data demonstrates a high percentage of extracranial tumor response rates with first line Erlotinib in selected NSCLC patients. We observed unexpected serious complications and postulate possible mechanisms. We recommend caution to be exercised when considering Erlotinib treatment during WBRT, particularly in regard to drug-drug interactions and infection control. Data from prospective trials are needed to determine the benefits and toxicities of Erlotinib during WBRT.


Subject(s)
Brain Neoplasms/drug therapy , Carcinoma, Non-Small-Cell Lung/drug therapy , Lung Neoplasms/drug therapy , Lung Neoplasms/radiotherapy , Quinazolines/therapeutic use , Adult , Aged , Aged, 80 and over , Brain Neoplasms/physiopathology , Brain Neoplasms/radiotherapy , Brain Neoplasms/secondary , Carcinoma, Non-Small-Cell Lung/physiopathology , Carcinoma, Non-Small-Cell Lung/radiotherapy , Carcinoma, Non-Small-Cell Lung/secondary , Combined Modality Therapy/adverse effects , Disease Progression , Drug Interactions , Erlotinib Hydrochloride , Female , Humans , Hyponatremia/etiology , Lung Neoplasms/pathology , Lung Neoplasms/physiopathology , Male , Middle Aged , Neutropenia/etiology , Treatment Outcome , Whole-Body Irradiation
13.
J Mol Biol ; 395(4): 705-16, 2010 Jan 29.
Article in English | MEDLINE | ID: mdl-19913028

ABSTRACT

The physiologically active form of p53 consists of a tetramer of four identical 393-amino-acid subunits associated via their tetramerization domains (TDs; residues 325-355). One in two human tumors contains a point mutation in the DNA binding domain (DBD) of p53 (residues 94-312). Most existing studies on the effects of these mutations on p53 structure and function have been carried out on the isolated DBD fragment, which is monomeric. Recent structural evidence, however, suggests that DBDs may interact with each other in full-length tetrameric forms of p53. Here, we investigate the effects of tumorigenic DBD mutations on the folding of p53 in its tetrameric form. We employ the construct consisting of DBD and TD (amino acids 94-360). We characterize the stability and conformational state of the tumorigenic DBD mutants R248Q, R249S, and R282Q using equilibrium denaturation and functional assays. Destabilizing mutations cause DBD to misfold when it is part of the p53 tetramer, but not when it is monomeric. This conformation is populated under moderately destabilizing conditions (10 degrees C in 2 M urea, and at physiological temperature in the absence of denaturant). Under those same conditions, it is not present in the isolated DBD fragment or in the presence of the TD mutation L344P, which abolishes tetramerization. Misfolding appears to involve intramolecular DBD-DBD association within a single tetrameric molecule. This association is promoted by destabilization of DBD (caused by mutation or elevated temperature) and by the high local DBD concentration enforced by tetramerization of TD. Disrupting the nonnative DBD-DBD interaction or transiently inhibiting tetramerization and allowing p53 to fold as a monomer may be potential strategies for pharmacological intervention in cancer.


Subject(s)
Neoplasms/chemistry , Neoplasms/genetics , Point Mutation , Tumor Suppressor Protein p53/chemistry , Tumor Suppressor Protein p53/genetics , Amino Acid Substitution , Base Sequence , DNA/genetics , DNA/metabolism , Humans , In Vitro Techniques , Models, Molecular , Neoplasms/physiopathology , Protein Denaturation , Protein Folding , Protein Interaction Domains and Motifs/genetics , Protein Multimerization , Protein Stability , Protein Structure, Quaternary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Thermodynamics , Tumor Suppressor Protein p53/physiology
14.
Biochemistry ; 48(15): 3497-507, 2009 Apr 21.
Article in English | MEDLINE | ID: mdl-19260676

ABSTRACT

Circular permutation of a protein covalently links its original termini and creates new ends at another location. To maintain the stability of the permuted structure, the termini are typically bridged by a peptide long enough to span the original distance between them. Here, we take the opposite approach and employ a very short linker to introduce conformational strain into a protein by forcing its termini together. We join the N- and C-termini of the small ribonuclease barnase (normally 27.2 A distant) with a single Cys residue and introduce new termini at a surface loop, to create pBn. Compared to a similar variant permuted with an 18-residue linker, permutation with a single amino acid dramatically destabilizes barnase. Surprisingly, pBn is folded at 10 degrees C and possesses near wild-type ribonuclease activity. The 2.25 A X-ray crystal structure of pBn reveals how the barnase fold is able to adapt to permutation, partially defuse conformational strain, and preserve enzymatic function. We demonstrate that strain in pBn can be relieved by cleaving the linker with a chemical reagent. Catalytic activity of both uncleaved (strained) pBn and cleaved (relaxed) pBn is proportional to their thermodynamic stabilities, i.e., the fraction of folded molecules. The stability and activity of cleaved pBn are dependent on protein concentration. At concentrations above approximately 2 microM, cleaving pBn is predicted to increase the fraction of folded molecules and thus enhance ribonuclease activity at 37 degrees C. This study suggests that introducing conformational strain by permutation, and releasing strain by cleavage, is a potential mechanism for engineering an artificial zymogen.


Subject(s)
Protein Conformation , Ribonucleases/chemistry , Thermodynamics , Amino Acid Sequence , Bacillus/enzymology , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Crystallography, X-Ray , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/metabolism , Molecular Sequence Data , Protein Folding , Protein Structure, Secondary , Protein Structure, Tertiary , Ribonucleases/antagonists & inhibitors , Ribonucleases/genetics , Stress, Physiological/genetics
15.
Proc Natl Acad Sci U S A ; 104(25): 10459-64, 2007 Jun 19.
Article in English | MEDLINE | ID: mdl-17556539

ABSTRACT

The spectra of equilibrium chain conformation fluctuations of apomyoglobin (apoMb) as a function of folding, from the acid-denatured state at pH 2.6 through the stable molten globule state pH approximately 4.1 to the folded state at pH 6.3, are reported, as measured by fluorescence correlation spectroscopy. The conformational fluctuations, which are detected by quenching of an N-terminal fluorescent label by contact with various amino acids, can be represented by superpositions of decaying exponentials with time scales ranging from approximately 3 to approximately 200 micros. Both the time scales and amplitudes of the fluctuations increase with the degree of acid denaturation, with principal shifts associated with the transition across the molten globule state. Measurements of the diffusion of apoMb confirm theoretical values showing a approximately 40% increase in the hydrodynamic radius upon acid denaturation. This study uses the model protein apoMb to illustrate the complex scope of folding associated structural dynamics.


Subject(s)
Apoproteins/chemistry , Myoglobin/chemistry , Protein Conformation , Protein Denaturation , Protein Folding , Spectrometry, Fluorescence , Amino Acid Sequence , Apoproteins/genetics , Apoproteins/isolation & purification , Cloning, Molecular , Diffusion , Gene Expression , Hydrogen-Ion Concentration , Kinetics , Models, Molecular , Mutation , Myoglobin/genetics , Myoglobin/isolation & purification , Protein Structure, Secondary , Time Factors
16.
Biochemistry ; 46(10): 2630-9, 2007 Mar 13.
Article in English | MEDLINE | ID: mdl-17297920

ABSTRACT

The DNA binding domain (DBD) of p53 folds by a complex mechanism that involves parallel pathways and multiple intermediates, both on- and off-pathway. This heterogeneity renders DBD particularly susceptible to misfolding and aggregation. The origins of parallel folding mechanisms are not well understood. DBD folding heterogeneity may be caused by the presence of the single bound Zn2+. To test that hypothesis, we carried out kinetic folding studies of DBD in its Zn2+-free form (apoDBD) and in the presence of various concentrations of free Zn2+ and the Zn2+-nitrilotriacetate (NTA) complex. Folding kinetics of apoDBD and DBD are similar, although apoDBD folds faster than DBD at some urea concentrations. The principle consequence of Zn2+ removal is to accelerate unfolding and simplify it from two exponential phases to one. Metal binding interactions are therefore not responsible for the observed complexity of the folding reaction. A slight stoichiometric excess of free Zn2+ arrests folding and traps the protein in a misfolded state in which Zn2+ is bound to nonphysiological ligands. Folding can be rescued by providing metal ions in the form of the NTA-Zn2+ complex, which simultaneously protects against misligation and provides a source of Zn2+ for regenerating the functional protein. This chemical metallochaperone strategy may be an effective means for improving folding efficiency of other metal binding proteins. The findings suggest that, in vivo, DBD must fold in an environment where free Zn2+ concentration is low and its bioavailability is carefully regulated by cellular metallochaperones.


Subject(s)
Protein Folding , Protein Structure, Tertiary/physiology , Tumor Suppressor Protein p53/chemistry , Zinc/pharmacology , Biological Availability , DNA/metabolism , DNA-Binding Proteins/chemistry , Models, Molecular , Protein Binding
17.
Protein Sci ; 15(11): 2457-65, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17001034

ABSTRACT

p53 modulates a large number of cellular response pathways and is critical for the prevention of cancer. Wild-type p53, as well as tumorigenic mutants, exhibits the singular property of spontaneously losing DNA binding activity at 37 degrees C. To understand the molecular basis for this effect, we examine the folding mechanism of the p53 DNA binding domain (DBD) at elevated temperatures. Folding kinetics do not change appreciably from 5 degrees C to 35 degrees C. DBD therefore folds by the same two-channel mechanism at physiological temperature as it does at 10 degrees C. Unfolding rates, however, accelerate by 10,000-fold. Elevated temperatures thus dramatically increase the frequency of cycling between folded and unfolded states. The results suggest that function is lost because a fraction of molecules become trapped in misfolded conformations with each folding-unfolding cycle. In addition, at 37 degrees C, the equilibrium stabilities of the off-pathway species are predicted to rival that of the native state, particularly in the case of destabilized mutants. We propose that it is the presence of these misfolded species, which can aggregate in vitro and may be degraded in the cell, that leads to p53 inactivation.


Subject(s)
Protein Folding , Temperature , Tumor Suppressor Protein p53/chemistry , Binding Sites , Body Temperature , DNA-Binding Proteins/chemistry , Humans , Kinetics , Models, Chemical , Mutant Proteins/chemistry , Protein Denaturation , Protein Structure, Tertiary
18.
J Mol Biol ; 357(4): 1058-62, 2006 Apr 07.
Article in English | MEDLINE | ID: mdl-16483603

ABSTRACT

A regulatory mechanism is introduced whereupon the catalytic activity of a given enzyme is controlled by ligand binding to a receptor domain of choice. A small enzyme (barnase) and a ligand-binding polypeptide (GCN4) are fused so that a simple topological constraint prevents them from existing simultaneously in their folded states. The two domains consequently engage in a thermodynamic tug-of-war in which the more stable domain forces the less stable domain to unfold. In the absence of ligand, the barnase domain is more stable and is therefore folded and active; the GCN4 domain is substantially unstructured. DNA binding induces folding of GCN4, forcibly unfolding and inactivating the barnase domain. Barnase-GCN4 is thus a "natively unfolded" protein that uses ligand binding to switch between partially folded forms. The key characteristics of each parent protein (catalytic efficiency of barnase, DNA binding affinity and sequence specificity of GCN4) are retained in the chimera. Barnase-GCN4 thus defines a modular approach for assembling enzymes with novel sensor capabilities from a variety of catalytic and ligand binding domains.


Subject(s)
DNA-Binding Proteins/chemistry , Protein Conformation , Protein Folding , Recombinant Fusion Proteins/chemistry , Ribonucleases/chemistry , Saccharomyces cerevisiae Proteins/chemistry , Transcription Factors/chemistry , Animals , Bacterial Proteins , Basic-Leucine Zipper Transcription Factors , Catalytic Domain , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Ligands , Models, Molecular , Peptides/chemistry , Peptides/genetics , Peptides/metabolism , Protein Binding , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Ribonucleases/genetics , Ribonucleases/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Thermodynamics , Transcription Factors/genetics , Transcription Factors/metabolism
19.
J Mol Biol ; 350(5): 906-18, 2005 Jul 29.
Article in English | MEDLINE | ID: mdl-15982667

ABSTRACT

The DNA-binding domain (DBD) of wild-type p53 loses DNA binding activity spontaneously at 37 degrees C in vitro, despite being thermodynamically stable at this temperature. We test the hypothesis that this property is due to kinetic misfolding of DBD. Interrupted folding experiments and chevron analysis show that native molecules are formed via four tracks (a-d) under strongly native conditions. Folding half-lives of tracks a-d are 7.8 seconds, 50 seconds, 5.3 minutes and more than five hours, respectively, in 0.3M urea (10 degrees C). Approximately equal fractions of molecules fold through each track in zero denaturant, but above 2.0M urea approximately 90% fold via track c. A kinetic mechanism consisting of two parallel folding channels (fast and slow) is proposed. Each channel populates an on-pathway intermediate that can misfold to form an aggregation-prone, dead-end species. Track a represents direct folding through the fast channel. Track b proceeds through the fast channel but via the off-pathway state. Track c corresponds to folding via the slow channel, primarily through the off-pathway state. Track d proceeds by way of an even slower, uncharacterized route. We postulate that activity loss is caused by partitioning to the slower tracks, and that structural unfolding limits this process. In support of this view, tumorigenic hot-spot mutants G245S, R249S and R282Q accelerate unfolding rates but have no affect on folding kinetics. We suggest that these and other destabilizing mutants facilitate loss of p53 function by causing DBD to cycle unusually rapidly between folded and unfolded states. A significant fraction of DBD molecules become effectively trapped in a non-functional state with each unfolding-folding cycle.


Subject(s)
Protein Folding , Tumor Suppressor Protein p53/chemistry , Binding Sites , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Kinetics , Mutation, Missense , Protein Denaturation , Spectrometry, Fluorescence , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Urea
20.
J Biol Chem ; 280(7): 5929-33, 2005 Feb 18.
Article in English | MEDLINE | ID: mdl-15591072

ABSTRACT

P22 is a well characterized tailed bacteriophage that infects Salmonella enterica serovar Typhimurium. It is characterized by a "short" tail, which is formed by five proteins: the dodecameric portal protein (gp1), three tail accessory factors (gp4, gp10, gp26), and six trimeric copies of the tail-spike protein (gp9). We have isolated the gene encoding tail accessory factor gp26, which is responsible for stabilization of viral DNA within the mature phage, and using a variety of biochemical and biophysical techniques we show that gp26 is very likely a triple stranded coiled-coil protein. Electron microscopic examination of purified gp26 indicates that the protein adopts a rod-like structure approximately 210 angstroms in length. This trimeric rod displays an exceedingly high intrinsic thermostability (T(m) approximately 85 degrees C), which suggests a potentially important structural role within the phage tail apparatus. We propose that gp26 forms the thin needle-like fiber emanating from the base of the P22 neck that has been observed by electron microscopy of negatively stained P22 virions. By analogy with viral trimeric coiled-coil class I membrane fusion proteins, gp26 may represent the membrane-penetrating device used by the phage to pierce the host outer membrane.


Subject(s)
Bacteriophage P22/chemistry , Viral Tail Proteins/chemistry , Amino Acid Sequence , Bacteriophage P22/ultrastructure , Computational Biology , Microscopy, Electron , Molecular Sequence Data , Protein Structure, Secondary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/ultrastructure , Viral Tail Proteins/genetics , Viral Tail Proteins/isolation & purification , Viral Tail Proteins/ultrastructure
SELECTION OF CITATIONS
SEARCH DETAIL
...