Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
Breast Cancer Res Treat ; 169(2): 359-369, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29388015

ABSTRACT

PURPOSE: Better tools are needed to estimate local recurrence (LR) risk after breast-conserving surgery (BCS) for DCIS. The DCIS score (DS) was validated as a predictor of LR in E5194 and Ontario DCIS cohort (ODC) after BCS. We combined data from E5194 and ODC adjusting for clinicopathological factors to provide refined estimates of the 10-year risk of LR after treatment by BCS alone. METHODS: Data from E5194 and ODC were combined. Patients with positive margins or multifocality were excluded. Identical Cox regression models were fit for each study. Patient-specific meta-analysis was used to calculate precision-weighted estimates of 10-year LR risk by DS, age, tumor size and year of diagnosis. RESULTS: The combined cohort includes 773 patients. The DS and age at diagnosis, tumor size and year of diagnosis provided independent prognostic information on the 10-year LR risk (p ≤ 0.009). Hazard ratios from E5194 and ODC cohorts were similar for the DS (2.48, 1.95 per 50 units), tumor size ≤ 1 versus  > 1-2.5 cm (1.45, 1.47), age ≥ 50 versus < 50 year (0.61, 0.84) and year ≥ 2000 (0.67, 0.49). Utilization of DS combined with tumor size and age at diagnosis predicted more women with very low (≤ 8%) or higher (> 15%) 10-year LR risk after BCS alone compared to utilization of DS alone or clinicopathological factors alone. CONCLUSIONS: The combined analysis provides refined estimates of 10-year LR risk after BCS for DCIS. Adding information on tumor size and age at diagnosis to the DS adjusting for year of diagnosis provides improved LR risk estimates to guide treatment decision making.


Subject(s)
Breast Neoplasms/surgery , Carcinoma, Intraductal, Noninfiltrating/surgery , Mastectomy, Segmental/adverse effects , Neoplasm Recurrence, Local/physiopathology , Aged , Breast Neoplasms/epidemiology , Breast Neoplasms/physiopathology , Carcinoma, Intraductal, Noninfiltrating/epidemiology , Carcinoma, Intraductal, Noninfiltrating/physiopathology , Female , Humans , Middle Aged , Neoplasm Recurrence, Local/epidemiology , Prognosis , Risk Assessment
2.
Br J Cancer ; 104(3): 488-95, 2011 Feb 01.
Article in English | MEDLINE | ID: mdl-21206494

ABSTRACT

BACKGROUND: Although it is accepted that metastatic colorectal cancers (mCRCs) that carry activating mutations in KRAS are unresponsive to anti-epidermal growth factor receptor (EGFR) monoclonal antibodies, a significant fraction of KRAS wild-type (wt) mCRCs are also unresponsive to anti-EGFR therapy. Genes encoding EGFR ligands amphiregulin (AREG) and epiregulin (EREG) are promising gene expression-based markers but have not been incorporated into a test to dichotomise KRAS wt mCRC patients with respect to sensitivity to anti-EGFR treatment. METHODS: We used RT-PCR to test 110 candidate gene expression markers in primary tumours from 144 KRAS wt mCRC patients who received monotherapy with the anti-EGFR antibody cetuximab. Results were correlated with multiple clinical endpoints: disease control, objective response, and progression-free survival (PFS). RESULTS: Expression of many of the tested candidate genes, including EREG and AREG, strongly associate with all clinical endpoints. Using multivariate analysis with two-layer five-fold cross-validation, we constructed a four-gene predictive classifier. Strikingly, patients below the classifier cutpoint had PFS and disease control rates similar to those of patients with KRAS mutant mCRC. CONCLUSION: Gene expression appears to identify KRAS wt mCRC patients who receive little benefit from cetuximab. It will be important to test this model in an independent validation study.


Subject(s)
Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Proto-Oncogene Proteins/genetics , ras Proteins/genetics , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal, Humanized , Antineoplastic Agents/therapeutic use , Cetuximab , Colorectal Neoplasms/secondary , Drug Resistance, Neoplasm/genetics , Gene Expression , Humans , Proto-Oncogene Proteins p21(ras)
3.
Oncology ; 61 Suppl 2: 37-42, 2001.
Article in English | MEDLINE | ID: mdl-11694786

ABSTRACT

The pivotal phase II and III Herceptin trials proved the efficacy and safety of second- or third-line single-agent Herceptin and first-line Herceptin in combination with chemotherapy, respectively. In the current trial, 114 patients were randomized to one of two dose groups of first-line Herceptin monotherapy: standard dose of 4 mg/ kg initial dose followed by 2 mg/kg intravenous (i.v.) weekly; or high dose of 8 mg/kg initial dose followed by 4 mg/kg i.v. weekly. The regimen was generally well tolerated. A similar incidence of adverse events was demonstrated in the two dose groups with the possible exception of acute infusion-related events such as fever and chills as well as rash and dyspnea, which appear to be more prevalent in the higher dose group. The overall response rate was 26% and response rates were similar between the two dose groups (24% for the standard Herceptin dose group and 28% for the high Herceptin dose group). Subgroup analysis determined a higher response rate in IHC 3+ patients (35%) and FISH-positive patients (41%). When women with stable disease for > or =6 months were included with responders, the clinical benefit rate in IHC 3+ patients was 47%. Median survival was 24.4 months, which is comparable with the survival rate seen in the pivotal phase III combination trial (25 months). Therefore, single-agent Herceptin is an important new option for the first-line treatment of HER2-positive metastatic breast cancer patients.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/therapeutic use , Breast Neoplasms/drug therapy , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/adverse effects , Antibodies, Monoclonal, Humanized , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Biomarkers, Tumor/analysis , Breast Neoplasms/chemistry , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Clinical Trials, Phase II as Topic , Clinical Trials, Phase III as Topic , Combined Modality Therapy , Disease Progression , Disease-Free Survival , Female , Fever/chemically induced , Heart Diseases/chemically induced , Humans , Neoplasm Metastasis , Neoplasm Proteins/analysis , Pain/virology , Palliative Care , Randomized Controlled Trials as Topic , Receptor, ErbB-2/analysis , Receptors, Estrogen/analysis , Safety , Salvage Therapy , Survival Analysis , Trastuzumab , Treatment Outcome
4.
Eur J Cancer ; 37 Suppl 1: 25-29, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11342197

ABSTRACT

Following confirmation of the appropriate dosage, safety and potential efficacy of Herceptin(R) (trastuzumab) in small-scale phase I and II trials involving patients with refractory disease, a large trial was conducted in 222 patients with breast cancer who had relapsed after one or two chemotherapy regimens for their metastatic disease. The results showed a positive and durable overall response rate (15% according to a response evaluation committee (REC) assessment) using trastuzumab monotherapy (initial dose 4 mg/kg intravenously (i.v.) followed by 2 mg/kg i.v. weekly). In another recently completed phase II trial, 113 patients were randomised to two dose levels (initial dose of 4 mg/kg i.v. dose followed by 2 mg/kg i.v. weekly, or initial dose of 8 mg/kg followed by 4 mg/kg i.v. weekly) of single-agent trastuzumab as first-line therapy for metastatic disease. The preliminary overall response rate was 23% based on investigator assessment, and tolerability was excellent as in previous trials; efficacy was similar in both dose groups, but the side-effects tended to be more frequent in the higher dose group. The preferred dosage is therefore the same as that currently recommended, i.e. an initial dose of 4 mg/kg i.v. followed by 2 mg/kg weekly i.v. until disease progression.

5.
N Engl J Med ; 344(11): 783-92, 2001 Mar 15.
Article in English | MEDLINE | ID: mdl-11248153

ABSTRACT

BACKGROUND: The HER2 gene, which encodes the growth factor receptor HER2, is amplified and HER2 is overexpressed in 25 to 30 percent of breast cancers, increasing the aggressiveness of the tumor. METHODS: We evaluated the efficacy and safety of trastuzumab, a recombinant monoclonal antibody against HER2, in women with metastatic breast cancer that overexpressed HER2. We randomly assigned 234 patients to receive standard chemotherapy alone and 235 patients to receive standard chemotherapy plus trastuzumab. Patients who had not previously received adjuvant (postoperative) therapy with an anthracycline were treated with doxorubicin (or epirubicin in the case of 36 women) and cyclophosphamide alone (138 women) or with trastuzumab (143 women). Patients who had previously received adjuvant anthracycline were treated with paclitaxel alone (96 women) or paclitaxel with trastuzumab (92 women). RESULTS: The addition of trastuzumab to chemotherapy was associated with a longer time to disease progression (median, 7.4 vs. 4.6 months; P<0.001), a higher rate of objective response (50 percent vs. 32 percent, P<0.001), a longer duration of response (median, 9.1 vs. 6.1 months; P<0.001), a lower rate of death at 1 year (22 percent vs. 33 percent, P=0.008), longer survival (median survival, 25.1 vs. 20.3 months; P=0.01), and a 20 percent reduction in the risk of death. The most important adverse event was cardiac dysfunction of New York Heart Association class III or IV, which occurred in 27 percent of the group given an anthracycline, cyclophosphamide, and trastuzumab; 8 percent of the group given an anthracycline and cyclophosphamide alone; 13 percent of the group given paclitaxel and trastuzumab; and 1 percent of the group given paclitaxel alone. Although the cardiotoxicity was potentially severe and, in some cases, life-threatening, the symptoms generally improved with standard medical management. CONCLUSIONS: Trastuzumab increases the clinical benefit of first-line chemotherapy in metastatic breast cancer that overexpresses HER2.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast Neoplasms/drug therapy , Receptor, ErbB-2/immunology , Adult , Aged , Anthracyclines/adverse effects , Anthracyclines/therapeutic use , Antibodies, Monoclonal/adverse effects , Antibodies, Monoclonal, Humanized , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Breast Neoplasms/metabolism , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Cyclophosphamide/adverse effects , Cyclophosphamide/therapeutic use , Disease Progression , Doxorubicin/adverse effects , Doxorubicin/therapeutic use , Epirubicin/therapeutic use , Female , Heart Diseases/chemically induced , Humans , Middle Aged , Neoplasm Metastasis , Paclitaxel/adverse effects , Paclitaxel/therapeutic use , Receptor, ErbB-2/metabolism , Survival Analysis , Trastuzumab
6.
J Clin Oncol ; 19(3): 843-50, 2001 Feb 01.
Article in English | MEDLINE | ID: mdl-11157038

ABSTRACT

PURPOSE: We investigated the safety and pharmacokinetics of a recombinant human monoclonal antibody to vascular endothelial growth factor (rhuMAb VEGF) in patients with cancer. PATIENTS AND METHODS: Cohorts of patients with metastatic cancer having failed prior therapy entered a phase I trial of rhuMAb VEGF administered by a 90-minute intravenous infusion at doses from 0.1 to 10.0 mg/kg on days 0, 28, 35, and 42. Patients underwent pharmacokinetic sampling on day 0 and had serum samples obtained during the subsequent 28 days. Response assessment was carried out on days 49 and 72. RESULTS: Twenty-five patients with a median Eastern Cooperative Oncology Group performance status of 0 were accrued. There were no grade III or IV adverse events definitely related to the antibody. There were three episodes of tumor-related bleeding. Infusions of rhuMAb VEGF were well tolerated without significant toxicity. Grades I and II adverse events possibly or probably related to study drug included asthenia, headache, and nausea. Pharmacokinetics revealed a linear profile with a half-life of 21 days. There were no objective responses, though 12 patients experienced stable disease over the duration of the study. CONCLUSION: rhuMAb VEGF was safely administered without dose-limiting toxicity at doses ranging up to 10 mg/kg. Multiple doses of rhuMAb VEGF were well tolerated, and pharmacokinetic studies indicate that doses of > or = 0.3 mg/kg have a half-life similar to that of other humanized antibodies. Subsequent trials will explore rhuMAb VEGF alone and in combination chemotherapy.


Subject(s)
Antibodies, Monoclonal/adverse effects , Antibodies, Monoclonal/pharmacokinetics , Endothelial Growth Factors/immunology , Lymphokines/immunology , Neoplasms/therapy , Adult , Aged , Antibodies, Monoclonal/therapeutic use , Dose-Response Relationship, Immunologic , Drug Administration Schedule , Endothelial Growth Factors/antagonists & inhibitors , Endothelial Growth Factors/blood , Female , Humans , Infusions, Intravenous , Lymphokines/antagonists & inhibitors , Lymphokines/blood , Male , Middle Aged , Neoplasms/blood supply , Neoplasms/metabolism , Neovascularization, Pathologic/prevention & control , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
7.
Eur J Cancer ; 37 Suppl 1: S25-9, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11167088

ABSTRACT

Following confirmation of the appropriate dosage, safety and potential efficacy of Herceptin(trastuzumab) in small-scale phase I and II trials involving patients with refractory disease, a large trial was conducted in 222 patients with breast cancer who had relapsed after one or two chemotherapy regimens for their metastatic disease. The results showed a positive and durable overall response rate (15% according to a response evaluation committee (REC) assessment) using trastuzumab monotherapy (initial dose 4 mg/kg intravenously (i.v.) followed by 2 mg/kg i.v. weekly). In another recently completed phase II trial, 113 patients were randomised to two dose levels (initial dose of 4 mg/kg i.v. dose followed by 2 mg/kg i.v. weekly, or initial dose of 8 mg/kg followed by 4 mg/kg i.v. weekly) of single-agent trastuzumab as first-line therapy for metastatic disease. The preliminary overall response rate was 23% based on investigator assessment, and tolerability was excellent as in previous trials; efficacy was similar in both dose groups, but the side-effects tended to be more frequent in the higher dose group. The preferred dosage is therefore the same as that currently recommended, i.e. an initial dose of 4 mg/kg i.v. followed by 2 mg/kg weekly i.v. until disease progression.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/therapeutic use , Breast Neoplasms/drug therapy , Adult , Aged , Aged, 80 and over , Antibodies, Monoclonal, Humanized , Female , Humans , Middle Aged , Multicenter Studies as Topic , Randomized Controlled Trials as Topic , Trastuzumab , Treatment Outcome
8.
Hum Gene Ther ; 11(2): 361-71, 2000 Jan 20.
Article in English | MEDLINE | ID: mdl-10680848

ABSTRACT

Targeting adenoviral vectors for cystic fibrosis gene therapy to the human airways with minimal exposure to alveoli would avoid adverse reactions and maximize response. At present, to deliver gene therapy vectors, large volumes of fluid are instilled or nebulized as aerosols. Either approach would likely cause alveolar exposure and increases the potential for side effects. We describe a coarse spray delivery device that precisely and reproducibly delivers the viral vector to the human airways to treat a small region of the airways for clinical trials. An endoscopic washing pipe (Olympus) that can be inserted into the channel of a bronchoscope was used. To minimize the escape of the therapeutic material downstream from the site of administration, we restricted the volume delivered to <150 microl (to prevent bulk flow), and used large droplets. Their high velocity further enhanced the probability of impaction in the vicinity of the nozzle. A pneumatic dosing system (Kahnetics) was used to reproducibly deliver the spray. The droplet size distribution was determined by laser diffraction and confirmed by cascade impaction: 190-microm volume median diameter with 1% mass <10 microm. The localization of the spray was studied in hollow cast models of human airways. 99mTc-sulfur colloid was used as a radiolabeled marker for these studies. Localization of the deposited spray was determined by scintigraphy and by measuring the radioactivity exiting the terminal airways. In the lung casts the spray was localized to one or two generations over an approximately 2-cm2 area. We conclude that delivery of large droplet sprays limits exposure to a few generations and may be useful in topical gene delivery clinical trials.


Subject(s)
Adenoviridae/genetics , Gene Transfer Techniques , Genetic Therapy/methods , Genetic Vectors , Lung , Administration, Inhalation , Aerosols , Cell Line , Cystic Fibrosis/therapy , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Dose-Response Relationship, Drug , Humans , Lung/anatomy & histology , Models, Anatomic , Particle Size , Technetium Tc 99m Sulfur Colloid/pharmacology
9.
J Clin Invest ; 104(9): 1245-55, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10545523

ABSTRACT

We sought to evaluate the ability of an E1(-), E3(-) adenovirus (Ad) vector (Ad(GV)CFTR.10) to transfer the normal human cystic fibrosis transmembrane conductance regulator (CFTR) cDNA to the airway epithelium of individuals with cystic fibrosis (CF). We administered Ad(GV)CFTR.10 at doses of 3 x 10(6) to 2 x 10(9) plaque-forming units over 9 months by endobronchial spray to 7 pairs of individuals with CF. Each 3-month cycle, we measured vector-derived versus endogenous CFTR mRNA in airway epithelial cells prior to therapy, as well as 3 and 30 days after therapy. The data demonstrate that (a) this strategy appears to be safe; (b) after the first administration, vector-derived CFTR cDNA expression in the CF airway epithelium is dose-dependent, with greater than 5% endogenous CFTR mRNA levels at the higher vector doses; (c) expression is transient, lasting less than 30 days; (d) expression can be achieved with a second administration, but only at intermediate doses, and no expression is observed with the third administration; and (e) the progressive lack of expression with repetitive administration does not closely correlate with induction of systemic anti-Ad neutralizing antibodies. The major advantage of an Ad vector is that it can deliver sufficient levels of CFTR cDNA to the airway epithelium so that CFTR expression protects the lungs from the respiratory manifestations of CF. However, this impressive level of expression is linked to the challenging fact that expression is limited in time. Although this can be initially overcome by repetitive administration, unknown mechanisms eventually limit this strategy, and further repetitive administration does not lead to repetitive expression.


Subject(s)
Bronchi/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cystic Fibrosis/metabolism , Cystic Fibrosis/therapy , Genetic Therapy/methods , Trachea/metabolism , Adenoviridae/genetics , Adolescent , Adult , Cohort Studies , Cystic Fibrosis Transmembrane Conductance Regulator/administration & dosage , Cystic Fibrosis Transmembrane Conductance Regulator/immunology , DNA, Complementary/genetics , Dose-Response Relationship, Drug , Epithelium/metabolism , Female , Genetic Vectors/metabolism , Humans , Male , Middle Aged , Models, Genetic , Polymerase Chain Reaction , RNA, Messenger/metabolism , Recombination, Genetic , Time Factors
10.
J Clin Oncol ; 17(9): 2639-48, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10561337

ABSTRACT

PURPOSE: Overexpression of the HER2 protein occurs in 25% to 30% of human breast cancers and leads to a particularly aggressive form of the disease. Efficacy and safety of recombinant humanized anti-HER2 monoclonal antibody as a single agent was evaluated in women with HER2-overexpressing metastatic breast cancer that had progressed after chemotherapy for metastatic disease. PATIENTS AND METHODS: Two hundred twenty-two women, with HER2-overexpressing metastatic breast cancer that had progressed after one or two chemotherapy regimens, were enrolled. Patients received a loading dose of 4 mg/kg intravenously, followed by a 2-mg/kg maintenance dose at weekly intervals. RESULTS: Study patients had advanced metastatic disease and had received extensive prior therapy. A blinded, independent response evaluation committee identified eight complete and 26 partial responses, for an objective response rate of 15% in the intent-to-treat population (95% confidence interval, 11% to 21%). The median duration of response was 9.1 months; the median duration of survival was 13 months. The most common adverse events, which occurred in approximately 40% of patients, were infusion-associated fever and/or chills that usually occurred only during the first infusion, and were of mild to moderate severity. These symptoms were treated successfully with acetaminophen and/or diphenhydramine. The most clinically significant adverse event was cardiac dysfunction, which occurred in 4.7% of patients. Only 1% of patients discontinued the study because of treatment-related adverse events. CONCLUSION: Recombinant humanized anti-HER2 monoclonal antibody, administered as a single agent, produces durable objective responses and is well tolerated by women with HER2-overexpressing metastatic breast cancer that has progressed after chemotherapy for metastatic disease. Side effects that are commonly observed with chemotherapy, such as alopecia, mucositis, and neutropenia, are rarely seen.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Breast Neoplasms/therapy , Receptor, ErbB-2/immunology , Adult , Aged , Aged, 80 and over , Antibodies, Monoclonal/adverse effects , Antibodies, Monoclonal/pharmacokinetics , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Confidence Intervals , Disease Progression , Disease-Free Survival , Female , Heart Diseases/etiology , Humans , Middle Aged , Multivariate Analysis , Quality of Life , Receptor, ErbB-2/metabolism , Time Factors
11.
Semin Oncol ; 26(4 Suppl 12): 71-7, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10482196

ABSTRACT

The recombinant humanized anti-HER2 monoclonal antibody trastuzumab (Herceptin; Genentech, San Francisco, CA) was evaluated in human clinical trials for treatment of women with metastatic breast cancer who have tumors that overexpress HER2. The trastuzumab clinical program consisted of a series of phase I, phase II, and phase III clinical trials. Clinical experience with this novel biologic has been obtained in more than 1,000 women with HER2-overexpressing metastatic breast cancer. Two pivotal trials were performed to evaluate trastuzumab efficacy and safety: (1) trastuzumab in combination with chemotherapy as first-line therapy and (2) trastuzumab as a single agent in second- and third-line chemotherapy. Preliminary results of the pivotal clinical trials that have been presented at national meetings are summarized below. The data suggest that trastuzumab will be an important new treatment option for women with HER2-overexpressing metastatic breast cancer.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/therapeutic use , Breast Neoplasms/drug therapy , Receptor, ErbB-2/immunology , Antibodies, Monoclonal, Humanized , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Clinical Trials as Topic , Female , Humans , Neoplasm Metastasis , Trastuzumab
12.
Breast Dis ; 10(5-6): 61-4, 1998 Dec.
Article in English | MEDLINE | ID: mdl-15687586
13.
Clin Exp Immunol ; 106(2): 243-52, 1996 Nov.
Article in English | MEDLINE | ID: mdl-8918569

ABSTRACT

The effects of recombinant mouse DNase on a well established murine model of spontaneous SLE have been evaluated. Daily intraperitoneal injections of DNase were given to female NZB/NZW F1 mice during the period of disease development from 4 to 7 months of age or at the height of disease activity from the age of 7 months for 3 weeks. This treatment was compared with the injections of diluent and with an immunosuppressive dose of dexamethasone. The effects of treatment were evaluated using the immunological parameters of disease activity (antinucleoprotein antibody, immune complexes, serum immunoglobulins, anti-cardiolipin antibodies), protein-uria, serum creatinine and renal histopathology (light microscopy, immunofluorescence and electron microscopy). The dose of dexamethasone used (1 mg/kg per day from the age of 4 months) was sufficient to suppress the development of lupus entirely. Treatment with DNase starting at the age of 4 months postponed the development of the disease by about 1 months and extended the period from the onset of disease to death by about 30%. Mice treated for 3 weeks during the most active phase of the disease at 7 months of age showed more dramatic effects. Proteinuria and serum creatinine were significantly reduced and renal histopathology was strikingly less severe than in the control group. Immune complexes involving DNA-containing antigens are believed to play a crucial role in the pathogenesis of SLE. DNA-nucleoprotein, even in immune complexes, can be destroyed by DNase. This enzyme therefore provides a rational way to interfere with the disease process. The results reported here encourage a trial of recombinant human DNase in human SLE and lupus nephritis.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Deoxyribonuclease I/therapeutic use , Dexamethasone/therapeutic use , Lupus Erythematosus, Systemic/therapy , Recombinant Proteins/therapeutic use , Animals , Antibodies, Anticardiolipin/analysis , Antibodies, Antinuclear/analysis , Antigen-Antibody Complex/analysis , Autoantigens/drug effects , Creatinine/blood , Disease Models, Animal , Female , Immunoglobulin G/analysis , Injections, Intraperitoneal , Kidney/pathology , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/mortality , Lupus Erythematosus, Systemic/pathology , Male , Mice , Mice, Inbred NZB
14.
Proc Natl Acad Sci U S A ; 93(16): 8225-9, 1996 Aug 06.
Article in English | MEDLINE | ID: mdl-8710851

ABSTRACT

Human deoxyribonuclease I (DNase I), an enzyme recently approved for treatment of cystic fibrosis (CF), has been engineered to create two classes of mutants: actin-resistant variants, which still catalyze DNA hydrolysis but are no longer inhibited by globular actin (G-actin) and active site variants, which no longer catalyze DNA hydrolysis but still bind G-actin. Actin-resistant variants with the least affinity for actin, as measured by an actin binding ELISA and actin inhibition of [33P] DNA hydrolysis, resulted from the introduction of charged, aliphatic, or aromatic residues at Ala-114 or charged residues on the central hydrophobic actin binding interface at Tyr-65 or Val-67. In CF sputum, the actin-resistant variants D53R, Y65A, Y65R, or V67K were 10-to 50-fold more potent than wild type in reducing viscoelasticity as determined in sputum compaction assays. The reduced viscoelasticity correlated with reduced DNA length as measured by pulsed-field gel electrophoresis. In contrast, the active site variants H252A or H134A had no effect on altering either viscoelasticity or DNA length in CF sputum. The data from both the active site and actin-resistant variants demonstrate that the reduction of viscoelasticity by DNase I results from DNA hydrolysis and not from depolymerization of filamentous actin (F-actin). The increased potency of the actin-resistant variants indicates that G-actin is a significant inhibitor of DNase I in CF sputum. These results further suggest that actin-resistant DNase I variants may have improved efficacy in CF patients.


Subject(s)
Actins/pharmacology , Cystic Fibrosis/drug therapy , Deoxyribonuclease I/antagonists & inhibitors , Actins/ultrastructure , Cystic Fibrosis/enzymology , DNA/metabolism , Deoxyribonuclease I/chemistry , Deoxyribonuclease I/ultrastructure , Enzyme Inhibitors , Humans , Macromolecular Substances , Models, Molecular , Mutagenesis, Site-Directed , Protein Engineering , Sputum/enzymology , Structure-Activity Relationship , Viscosity
15.
Eur Respir J ; 9(4): 765-9, 1996 Apr.
Article in English | MEDLINE | ID: mdl-8726943

ABSTRACT

Recombinant human deoxyribonuclease (rhDNase) has been demonstrated to reduce in vitro the viscosity and to improve the transport capacity of purulent respiratory mucus in cystic fibrosis. During episodes of exacerbation of chronic bronchitis, the patients generally expectorate purulent mucus. Purulence of mucus is associated with an increased deoxyriboneucleic acid (DNA) concentration. We analyzed in vitro the potential effect of rhDNase on chronic bronchitis mucus transport by the ciliary activity (frog palate model) and by simulated cough (cough machine model), as well as the effect on mucus viscosity (controlled stress rheometer) and surface properties (contact angle). Purulent sputa collected from patients with chronic bronchitis (n = 15) during an episode of exacerbation were incubated for 30 min at 37 degrees C with either rhDNase at two different concentrations (final concentration 2 or 4 micrograms.mL-1) or placebo. The median mucociliary transport rate was significantly improved by rhDNase from 0.68 with placebo to 0.79 and 0.83 with 2 and 4 micrograms.mL-1 of rhDNase, respectively. A significant improvement in mucus cough transport was also induced by rhDNase from 25.5 mm with placebo to 27.0 mm with either 2 or 4 micrograms.mL-1 rhDNase. These improvements in mucus transport capacity were associated with alterations in the physical properties of the mucus. The mucus median control viscosity (511.4 Pa.s) and median contact angle (0.85 rd) significantly decreased to 112.5 Pa.s and 0.74 rd, respectively, in the presence of 4 micrograms.mL-1 of rhDNase. These findings demonstrate that recombinant deoxyribonuclease may exert a beneficial effect on mucus clearance in vitro by altering the viscosity and surface properties of the purulent chronic bronchitic sputum samples.


Subject(s)
Bronchitis/metabolism , Deoxyribonucleases/metabolism , Mucus/chemistry , Mucus/drug effects , Chronic Disease , Cough/metabolism , DNA/analysis , Deoxyribonucleases/genetics , Female , Humans , Male , Mucociliary Clearance/physiology , Mucus/physiology , Recombinant Proteins/metabolism , Surface Properties , Viscosity
16.
Arch Pediatr ; 2(7): 666-9, 1995 Jul.
Article in French | MEDLINE | ID: mdl-7663657

ABSTRACT

Despite the hopes raised by the first attempts in gene therapy, direct correction of the defect in CFTR protein associated with cystic fibrosis is still beyond clinical reach. Therefore we have to set upon the consequences of the defect. Respiratory distress and progressive lung destruction in cystic fibrosis can be accounted for by infectious exacerabations and the accumulation of viscous purulent secretions in the airways. For a long time we have known that purulent secretions that accumulate in the airways of patients with cystic fibrosis contain large amounts of DNA, a complex macromolecule that contributes mostly to the viscosity and hinders the mucociliary function. Hence we hypothesized that enzymatic cleaving of DNA molecules by desoxyribonuclease (DNase) should reduce the viscosity of sputum, and slow or prevent the deterioration of pulmonary function. Using the techniques of molecular biology and genetic engineering, we identified the gene of human DNase I, which was cloned in mammalian cells to produce large amounts of a glycosylated protein for therapeutic use. Catalytic amounts of rhDNase greatly reduce the viscosity of purulent cystic fibrosis sputum, transforming it within minutes from a nonflowing viscous gel to a flowing liquid. This effect was associated with a decrease in size of DNA fragments in the sputum. Our studies suggested that inhalation of a rhDNase aerosol might be a simple direct approach to reduce the viscosity of purulent secretions and thereby help patients with cystic fibrosis clear their airways and breathe more easily.


Subject(s)
Cystic Fibrosis/enzymology , Deoxyribonuclease I/metabolism , Cloning, Molecular , Cystic Fibrosis/drug therapy , Deoxyribonuclease I/therapeutic use , Expectorants/therapeutic use , Humans , In Vitro Techniques , Recombinant Proteins/metabolism , Recombinant Proteins/therapeutic use
17.
Eur Respir J ; 8(3): 381-6, 1995 Mar.
Article in English | MEDLINE | ID: mdl-7789481

ABSTRACT

Recombinant human deoxyribonuclease (rhDNase) has been demonstrated to reduce the viscosity of purulent cystic fibrosis (CF) respiratory mucus, to improve pulmonary function and to reduce the risk of respiratory tract infectious exacerbations, but its effect on mucus transportability has not so far been investigated. The dose-dependent effect of rhDNase was analysed in vitro on mucus transport rate (tr) by ciliary activity and by simulated cough (cough transport (ct)), as well as on mucus viscosity and surface properties. Purulent CF sputa (n = 15) were incubated for 30 min at 37 degrees C with either rhDNase at three different concentrations (final concentrations 0.2, 2 or 20 micrograms.ml-1 of mucus) or placebo. No significant dose-dependent effect of rhDNase on the mucociliary transport rate was observed when the samples wer statistically analysed together. However, in the larger group of mucus samples (n = 11) with a low initial mucociliary transport rate, the latter was improved at each rhDNase concentration (tr0.2 = 0.69, tr2 = 0.88 and tr20 = 0.87) as compared to placebo (trp = 0.58). In the smaller group of mucus samples (n = 4) with high initial transport rate, a decrease in mucociliary transport rate was observed, particularly at the highest concentration rhDNase assayed, i.e. 20 micrograms.ml-1 of mucus (tr20 = 0.58) as compared to placebo (trp = 0.86). The mucus cough transport was increased by rhDNase (ct0.2 = 25 mm, ct2 = 27.5 mm and ct20 = 31 mm) as compared to placebo (ctp = 23.5 mm).(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Cystic Fibrosis/physiopathology , Deoxyribonuclease I/pharmacology , Mucus/drug effects , Administration, Inhalation , Adult , Cough/physiopathology , Cystic Fibrosis/drug therapy , Deoxyribonuclease I/administration & dosage , Dose-Response Relationship, Drug , Humans , In Vitro Techniques , Mucociliary Clearance/drug effects , Recombinant Proteins/administration & dosage , Recombinant Proteins/pharmacology , Rheology , Viscosity
18.
Chest ; 107(2 Suppl): 65S-70S, 1995 Feb.
Article in English | MEDLINE | ID: mdl-7842816

ABSTRACT

Respiratory symptoms, recurrent infectious exacerbations, and progressive lung destruction in cystic fibrosis can be attributed to bacterial persistence and the accumulation of viscous purulent secretions in the airways. Purulent secretions contain high concentrations of extracellular DNA, a viscous material released by leukocytes. To evaluate the potential clinical utility of recombinant human DNase I (rhDNase or Pulmozyme), the human enzyme was cloned, sequenced, and expressed. In in vitro studies, rhDNase has been shown to reduce the viscoelasticity, reduce the adhesiveness, and improve the mucociliary transportability of cystic fibrosis sputum. In short-term phase 1 and phase 2 clinical trials, rhDNase has been shown to be safely tolerated and to improve the FEV1, FVC, and symptoms of dyspnea. A long-term placebo-controlled phase 3 study was performed in 968 adults and children (> or = 5 years) with cystic fibrosis to determine the effect of rhDNase on the risk of respiratory exacerbations requiring parenteral antibiotics and on the FEV1. Compared with placebo-treated patients, patients treated with rhDNase once daily or twice daily experienced a reduced risk of respiratory exacerbations by 28% (p = 0.04) and 37% (p = 0.01), respectively, and had a mean improvement in FEV1 of 5.8% (p < 0.01) and 5.6% (p < 0.01), respectively. Compared with placebo-treated patients, patients treated with rhDNase spent 2.7 fewer days receiving parenteral antibiotics (p = 0.04) and spent 1.3 fewer days in the hospital (p = 0.06) over the 6-month treatment period. Inhalation of rhDNase did not cause anaphylaxis but was associated with upper airway symptoms (ie, voice alteration, hoarseness, pharyngitis) that were generally mild and transient. In conclusion, aerosol administration of rhDNase was safely tolerated, reduced the risk of infectious exacerbations requiring parenteral antibiotics, and improved pulmonary function and patient well-being.


Subject(s)
Cystic Fibrosis/drug therapy , Deoxyribonuclease I/therapeutic use , Expectorants/therapeutic use , Administration, Inhalation , Aerosols , Clinical Trials as Topic , Cystic Fibrosis/physiopathology , Deoxyribonuclease I/administration & dosage , Expectorants/administration & dosage , Forced Expiratory Volume , Humans , Randomized Controlled Trials as Topic , Recombinant Proteins/administration & dosage , Recombinant Proteins/therapeutic use
19.
Anal Biochem ; 222(2): 351-8, 1994 Nov 01.
Article in English | MEDLINE | ID: mdl-7864358

ABSTRACT

A simple, high throughput, and precise assay was developed for quantification of deoxyribonuclease I (DNase; IUB 3.1.21.1) activity. The method was adapted from the procedure devised by Kurnick which employs a substrate comprised of highly polymerized native DNA complexed with methyl green. Hydrolysis of the DNA produced unbound methyl green and a decrease in the absorbance of the solution at 620 nm. By adjusting the time and temperature of the reaction, the assay permits quantification of DNase activity over a wide concentration range (0.4 to 8900 ng/ml). Samples and standards were added to the substrate in microtiter plates and were incubated for 1-24 h at 25-37 degrees C to achieve the desired assay range. The DNase activity of the samples was interpolated from a standard curve generated with Pulmozyme recombinant human deoxyribonuclease I (rhDNase). Interassay precision was less than 12% CV and recovery was within 100 +/- 11%. Activity determination by the DNA-methyl green method correlated well with that determined by the widely used "hyperchromicity" method originated by Kunitz, which is based on the increase in absorbance at 260 nm upon hydrolysis of DNA. The DNA-methyl green assay was simpler and more versatile than the hyperchromicity method and was used to characterize the activity of rhDNase and DNase isolated from human urine.


Subject(s)
Colorimetry/methods , DNA/chemistry , Deoxyribonuclease I/analysis , Methyl Green/chemistry , Animals , Cations, Divalent , Cattle , Deoxyribonuclease I/urine , Drug Stability , Humans , Hydrogen-Ion Concentration , Nucleic Acid Denaturation , Pancreas/enzymology , Reproducibility of Results , Sensitivity and Specificity , Spectrophotometry, Ultraviolet , Substrate Specificity
20.
Bioconjug Chem ; 5(2): 133-40, 1994.
Article in English | MEDLINE | ID: mdl-8031876

ABSTRACT

CD4 immunoadhesin (CD4-IgG) is a chimeric glycoprotein molecule comprised of the gp120-binding portion of human CD4 fused to the hinge and Fc portions of human IgG. As a candidate for human therapeutic use, CD4-IgG represents an important advance over soluble CD4, insofar as the systemic clearance in humans of CD4-IgG is significantly slower. In an effort to prolong its in vivo residence time even further, we have modified CD4-IgG chemically by attaching monomethoxypoly(ethylene glycol) (MePEG) moieties to lysine residues via reductive alkylation. We synthesized MePEG aldehyde and investigated reaction conditions for adding a range of MePEG moieties per protein molecule. At neutral pH in the presence of sodium cyanoborohydride, the reaction was sufficiently slow to allow for significant control over the extent of MePEGylation. Addition of 7.7 or 14.4 MePEG moieties to CD4-IgG resulted in an approximately 4- or 5-fold increase, respectively, in the persistence of the protein in rats, as compared with unmodified CD4-IgG. These results suggest that the therapeutic utility of a human receptor IgG chimera can be improved by MePEGylation technology, provided that the modified immunoadhesin retains its biological activity in vivo. Such modification can lead to a significant additional increase in the in vivo residence time of the protein.


Subject(s)
CD4 Immunoadhesins/chemistry , Polyethylene Glycols/chemistry , Alkylation , Animals , Electrophoresis, Polyacrylamide Gel , HIV Envelope Protein gp120/metabolism , Humans , Immunoglobulin G/chemistry , Magnetic Resonance Spectroscopy , Male , Oxidation-Reduction , Peptide Mapping , Polyethylene Glycols/chemical synthesis , Rats , Rats, Sprague-Dawley , Recombinant Proteins/chemistry , Sequence Analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...