Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
Ophthalmic Res ; 66(1): 1030-1043, 2023.
Article in English | MEDLINE | ID: mdl-37257422

ABSTRACT

INTRODUCTION: The Understanding New Interventions with GBM ThErapy (UNITE) study was designed to assess the effect of prophylaxis for ocular side effects (OSEs) in patients with glioblastoma receiving the antibody-drug conjugate (ADC) depatuxizumab mafodotin. UNITE (NCT03419403) was a phase 3b, open-label, randomized, exploratory study performed at 18 research sites in 5 countries. METHODS: The study enrolled adult patients with epidermal growth factor receptor-amplified, histologically confirmed, newly diagnosed supratentorial glioblastoma or grade IV gliosarcoma, and a Karnofsky Performance Status ≥70, receiving depatuxizumab mafodotin. All patients were administered depatuxizumab mafodotin during concurrent radiotherapy and temozolomide and with adjuvant temozolomide. Ninety patients were to be randomized (1:1:1) to OSE prophylactic treatments with each depatuxizumab mafodotin infusion: (a) standard steroid eye drops, (b) standard steroid eye drops plus vasoconstrictor eye drops and cold compress, or (c) enhanced steroids plus vasoconstrictor eye drops and cold compress. A Corneal Epitheliopathy Adverse Event (CEAE) scale was devised to capture symptoms, grade OSEs (scale of 0-5), and inform ADC dose modifications. The primary endpoint was the frequency of a required change in OSE management due to inadequate control of OSEs, defined as decline from baseline in visual acuity (using logarithm of the minimum angle of resolution [LogMAR] scale) or a Grade ≥3 CEAE event, in the worst eye in the first 8 weeks of treatment; unless otherwise specified, the treatment period refers to both the chemoradiation and adjuvant phases. RESULTS: The UNITE study was stopped early after interim analysis of separate phase III trial showed no difference in survival from depatuxizumab mafodotin. Forty patients were randomized (38 received depatuxizumab mafodotin). Overall, 23 patients experienced inadequate control of OSEs that required change in OSE management within 8 weeks of treatment, with 21 (70.0%) experiencing ≥+0.3 change on LogMAR scale in baseline-adjusted visual acuity and 12 reporting a grade ≥3 CEAE. There were no definitive differences among prophylactic treatments. CONCLUSIONS: The premature cessation of the study precludes definitive conclusions regarding the OSE prophylaxis strategies. No new clinically significant safety findings were noted. Despite these limitations, this study highlights the need for novel assessment tools to better understand and mitigate OSEs associated with ADCs.


Subject(s)
Glioblastoma , Adult , Humans , ErbB Receptors/metabolism , Glioblastoma/drug therapy , Glioblastoma/metabolism , Glioblastoma/pathology , Ophthalmic Solutions/therapeutic use , Steroids/therapeutic use , Temozolomide/therapeutic use , Vasoconstrictor Agents/therapeutic use
2.
Neuro Oncol ; 25(2): 339-350, 2023 02 14.
Article in English | MEDLINE | ID: mdl-35849035

ABSTRACT

BACKGROUND: Approximately 50% of newly diagnosed glioblastomas (GBMs) harbor epidermal growth factor receptor gene amplification (EGFR-amp). Preclinical and early-phase clinical data suggested efficacy of depatuxizumab mafodotin (depatux-m), an antibody-drug conjugate comprised of a monoclonal antibody that binds activated EGFR (overexpressed wild-type and EGFRvIII-mutant) linked to a microtubule-inhibitor toxin in EGFR-amp GBMs. METHODS: In this phase III trial, adults with centrally confirmed, EGFR-amp newly diagnosed GBM were randomized 1:1 to radiotherapy, temozolomide, and depatux-m/placebo. Corneal epitheliopathy was treated with a combination of protocol-specified prophylactic and supportive measures. There was 85% power to detect a hazard ratio (HR) ≤0.75 for overall survival (OS) at a 2.5% 1-sided significance level (ie traditional two-sided p ≤ 0.05) by log-rank testing. RESULTS: There were 639 randomized patients (median age 60, range 22-84; 62% men). Prespecified interim analysis found no improvement in OS for depatux-m over placebo (median 18.9 vs. 18.7 months, HR 1.02, 95% CI 0.82-1.26, 1-sided p = 0.63). Progression-free survival was longer for depatux-m than placebo (median 8.0 vs. 6.3 months; HR 0.84, 95% confidence interval [CI] 0.70-1.01, p = 0.029), particularly among those with EGFRvIII-mutant (median 8.3 vs. 5.9 months, HR 0.72, 95% CI 0.56-0.93, 1-sided p = 0.002) or MGMT unmethylated (HR 0.77, 95% CI 0.61-0.97; 1-sided p = 0.012) tumors but without an OS improvement. Corneal epitheliopathy occurred in 94% of depatux-m-treated patients (61% grade 3-4), causing 12% to discontinue. CONCLUSIONS: Interim analysis demonstrated no OS benefit for depatux-m in treating EGFR-amp newly diagnosed GBM. No new important safety risks were identified.


Subject(s)
Brain Neoplasms , Glioblastoma , Adult , Male , Humans , Middle Aged , Female , Glioblastoma/drug therapy , Glioblastoma/genetics , Glioblastoma/metabolism , Antibodies, Monoclonal, Humanized , Temozolomide/therapeutic use , ErbB Receptors , Brain Neoplasms/drug therapy , Brain Neoplasms/genetics , Brain Neoplasms/pathology
3.
Neurooncol Adv ; 2(1): vdz051, 2020.
Article in English | MEDLINE | ID: mdl-32642719

ABSTRACT

BACKGROUND: The randomized phase II INTELLANCE-2/EORTC_1410 trial on EGFR-amplified recurrent glioblastomas showed a trend towards improved overall survival when patients were treated with depatux-m plus temozolomide compared with the control arm of alkylating chemotherapy only. We here performed translational research on material derived from this clinical trial to identify patients that benefit from this treatment. METHODS: Targeted DNA-sequencing and whole transcriptome analysis was performed on clinical trial samples. High-throughput, high-content imaging analysis was done to understand the molecular mechanism underlying the survival benefit. RESULTS: We first define the tumor genomic landscape in this well-annotated patient population. We find that tumors harboring EGFR single-nucleotide variations (SNVs) have improved outcome in the depatux-m + TMZ combination arm. Such SNVs are common to the extracellular domain of the receptor and functionally result in a receptor that is hypersensitive to low-affinity EGFR ligands. These hypersensitizing SNVs and the ligand-independent EGFRvIII variant are inversely correlated, indicating two distinct modes of evolution to increase EGFR signaling in glioblastomas. Ligand hypersensitivity can explain the therapeutic efficacy of depatux-m as increased ligand-induced activation will result in increased exposure of the epitope to the antibody-drug conjugate. We also identified tumors harboring mutations sensitive to "classical" EGFR tyrosine-kinase inhibitors, providing a potential alternative treatment strategy. CONCLUSIONS: These data can help guide treatment for recurrent glioblastoma patients and increase our understanding into the molecular mechanisms underlying EGFR signaling in these tumors.

4.
J Neurooncol ; 144(1): 205-210, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31273577

ABSTRACT

PURPOSE: Epidermal growth factor receptor (EGFR) amplification has been reported to occur in ~ 50% of glioblastomas (GBMs). We are conducting several global studies that require central testing for EGFR amplification during screening, representing an opportunity to confirm the frequency of amplification in GBM in a large cohort and to evaluate whether EGFR amplification differs by region of the world. METHODS: EGFR amplification was measured by fluorescence in situ hybridization during screening for therapeutic trials of an EGFR antibody-drug conjugate: two Phase 2/3 global trials (INTELLANCE-1, INTELLANCE-2), and a Japanese Phase 1/2 trial (INTELLANCE-J). We evaluated the proportion of tumor tissue samples harboring EGFR amplification among those tested and differences in amplification frequency by geography. RESULTS: EGFR was amplified in 54% of 3150 informative cases screened for INTELLANCE-1 and -2, consistent with historic controls, but was significantly lower in patients from Asia versus the rest of the world (35% vs. 56%, P < 0.0030). The independent INTELLANCE-J trial validated this finding (33% amplified of 153 informative cases). CONCLUSIONS: EGFR amplification occurs less frequently in patients from Asia than elsewhere. Further study is required to understand biological differences to optimize treatment in glioblastoma.


Subject(s)
Biomarkers, Tumor/genetics , Brain Neoplasms/genetics , Gene Amplification , Glioblastoma/genetics , Mass Screening/standards , Patient Selection , Brain Neoplasms/drug therapy , Brain Neoplasms/pathology , Double-Blind Method , ErbB Receptors/genetics , Glioblastoma/drug therapy , Glioblastoma/pathology , Humans , Prognosis
5.
Neuro Oncol ; 21(10): 1263-1272, 2019 10 09.
Article in English | MEDLINE | ID: mdl-31125418

ABSTRACT

BACKGROUND: Precision medicine trials targeting the epidermal growth factor receptor (EGFR) in glioblastoma patients require selection for EGFR-amplified tumors. However, there is currently no gold standard in determining the amplification status of EGFR or variant III (EGFRvIII) expression. Here, we aimed to determine which technique and which cutoffs are suitable to determine EGFR amplification status. METHODS: We compared fluorescence in-situ hybridization (FISH) and real-time quantitative (RT-q)PCR data from patients screened for trial inclusion into the Intellance 2 clinical trial, with data from a panel-based next generation sequencing (NGS) platform (both DNA and RNA). RESULTS: By using data from >1000 samples, we show that at least 50% of EGFR amplified nuclei should be present to define EGFR gene amplification by FISH. Gene amplification (as determined by FISH) correlates with EGFR expression levels (as determined by RT-qPCR) with receiver operating characteristics analysis showing an area under the curve of up to 0.902. EGFR expression as assessed by RT-qPCR therefore may function as a surrogate marker for EGFR amplification. Our NGS data show that EGFR copy numbers can strongly vary between tumors, with levels ranging from 2 to more than 100 copies per cell. Levels exceeding 5 gene copies can be used to define EGFR-amplification by NGS; below this level, FISH detects very few (if any) EGFR amplified nuclei and none of the samples express EGFRvIII. CONCLUSION: Our data from central laboratories and diagnostic sequencing facilities, using material from patients eligible for clinical trial inclusion, help define the optimal cutoff for various techniques to determine EGFR amplification for diagnostic purposes.


Subject(s)
Glioblastoma/genetics , Nucleic Acid Amplification Techniques/standards , Patient Selection , Clinical Trials as Topic/standards , ErbB Receptors/analysis , ErbB Receptors/genetics , Gene Amplification , Gene Dosage , Glioblastoma/drug therapy , High-Throughput Nucleotide Sequencing/standards , Humans , In Situ Hybridization, Fluorescence/standards , Real-Time Polymerase Chain Reaction/standards , Reference Values
6.
J Clin Pharmacol ; 59(9): 1225-1235, 2019 09.
Article in English | MEDLINE | ID: mdl-30990907

ABSTRACT

Depatuxizumab mafodotin (depatux-m) is an antibody-drug conjugate (ADC) designed for the treatment of tumors expressing epidermal growth factor receptor (EGFR), consisting of a veneered "humanized" recombinant IgG1κ antibody that has binding properties specific to a unique epitope of human EGFR with noncleavable maleimido-caproyl linkers each attached to a potent antimitotic cytotoxin, monomethyl auristatin F. We aimed to describe the development and comparison of 2 population pharmacokinetic modeling approaches. Data from 2 phase 1 studies enrolling patients with glioblastoma multiforme or advanced solid tumors were included in the analysis. Patients in these studies received doses of depatux-m ranging from 0.5 to 4.0 mg/kg as monotherapy, in combination with temozolomide, or radiation plus temozolomide depending on the study and/or arm. First, an integrated ADC model to simultaneously describe the concentration-time data for ADC, total antibody, and cys-mafodotin was built using a 2-compartment model for ADC for each drug-to-antibody ratio. Then, 3 individual models were developed for ADC, total antibody, and cys-mafodotin separately using 2-compartment models for ADC and total antibody and a 1-compartment model for cys-mafodotin. Visual predictive checks suggested accurate model fitting across a range of concentrations. The analysis showed that both an integrated complex ADC model and the individual models that have shorter computational time would result in similar outcomes.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacokinetics , Glioblastoma/metabolism , Adult , Aged , Aged, 80 and over , Antibodies, Monoclonal, Humanized/therapeutic use , ErbB Receptors/metabolism , Female , Glioblastoma/drug therapy , Humans , Immunoconjugates/pharmacokinetics , Immunoconjugates/therapeutic use , Male , Middle Aged , Temozolomide/pharmacokinetics , Temozolomide/therapeutic use , Young Adult
7.
Neuro Oncol ; 21(1): 106-114, 2019 01 01.
Article in English | MEDLINE | ID: mdl-29982805

ABSTRACT

Background: Patients with glioblastoma (GBM) have a dismal prognosis. Nearly all will relapse with no clear standard of care for recurrent disease (rGBM). Approximately 50% of patients have tumors harboring epidermal growth factor receptor (EGFR) amplification. The antibody-drug conjugate depatuxizumab mafodotin (depatux-m) binds cells with EGFR amplification, is internalized, and releases a microtubule toxin, killing the cell. Here we report efficacy, safety and pharmacokinetics (PK) of depatux-m + temozolomide (TMZ) in patients with EGFR-amplified rGBM. Methods: M12-356 (NCT01800695) was an open-label study encompassing patients with newly diagnosed or rGBM across 3 treatment arms. Results are reported for adults with EGFR-amplified, measurable rGBM who received depatux-m (0.5-1.5 mg/kg) on days 1 and 15, and TMZ (150-200 mg/m2) on days 1-5 in a 28-day cycle. Patients were bevacizumab and nitrosourea naïve. Results: There were 60 patients, median age 56 years (range, 20-79). Fifty-nine patients previously received TMZ. Common adverse events (AEs) were blurred vision (63%), fatigue (38%), and photophobia (35%). Grades 3/4 AEs were split between ocular and non-ocular AEs, occurring in 22% of patients each. Systemic PK exposure of depatux-m was dose proportional. The objective response rate was 14.3%, the 6-month progression-free survival rate was 25.2%, and the 6-month overall survival rate was 69.1%. Conclusions: Depatux-m + TMZ displayed an AE profile similar to what was described previously. Antitumor activity in this TMZ-refractory population was encouraging. Continued study of depatux-m in patients with EGFR-amplified, newly diagnosed, or recurrent GBM is ongoing in 2 global, randomized trials (NCT02573324, NCT02343406).


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Brain Neoplasms/drug therapy , Gene Amplification , Glioblastoma/drug therapy , Neoplasm Recurrence, Local/drug therapy , Adult , Aged , Antibodies, Monoclonal, Humanized/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Cohort Studies , ErbB Receptors/genetics , Female , Follow-Up Studies , Glioblastoma/genetics , Glioblastoma/pathology , Humans , International Agencies , Male , Maximum Tolerated Dose , Middle Aged , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/pathology , Prognosis , Temozolomide/administration & dosage , Tissue Distribution , Young Adult
8.
J Clin Psychiatry ; 79(3)2018.
Article in English | MEDLINE | ID: mdl-28922590

ABSTRACT

OBJECTIVE: To evaluate the efficacy and safety of the α7-nicotinic receptor agonist ABT-126 for treatment of cognitive impairment in stable subjects with schizophrenia who smoke. METHODS: A 12-week double-blind, placebo-controlled, parallel-group study was conducted from August 2012 to March 2014. Subjects with a diagnosis of schizophrenia based on DSM-IV-TR criteria (confirmed by the Mini-International Neuropsychiatric Interview version 6.0.0) were randomized 1:1:1 to ABT-126 25 mg, ABT-126 75 mg, or placebo once daily while maintained on their background antipsychotic medication. The primary endpoint was the change from baseline on the Measurement and Treatment Research to Improve Cognition in Schizophrenia (MATRICS) Consensus Cognitive Battery (MCCB) neurocognitive composite score; the primary analysis compared ABT-126 with placebo at week 12 using a mixed-effects model for repeated measures. Secondary endpoints included the change from baseline on the University of California San Diego Performance-based Skills Assessment-2 Extended-Range, the 16-item Negative Symptom Assessment scale (NSA-16), and safety assessments. RESULTS: Of the 157 randomized subjects, 82% completed the study. The mean baseline MCCB neurocognitive composite score for the entire study sample was 28.8; scores were similar across groups. No statistical difference in the change from baseline score between any of the ABT-126 dose groups and placebo was observed on the MCCB neurocognitive composite score (ABT-126 25 mg, +0.28; ABT-126 75 mg, +0.41; placebo, +1.42). Differences in the NSA-16 total score were seen with ABT-126 75 mg versus placebo at week 6 (-2.79; P = .011) and week 12 (-1.94; P = .053). Adverse events with ABT-126 were similar to placebo, except for constipation (5.8% for ABT-126 vs 0% for placebo). CONCLUSIONS: ABT-126 did not demonstrate a procognitive effect in subjects with stable schizophrenia who smoke. A trend for improvement in negative symptoms was observed with the high dose. The safety profile of ABT-126 was similar to placebo. TRIAL REGISTRATION: ClinicalTrials.gov identifier: NCT01678755​.


Subject(s)
Antipsychotic Agents/therapeutic use , Cognitive Dysfunction/drug therapy , Cognitive Dysfunction/physiopathology , Outcome Assessment, Health Care , Quinuclidines/pharmacology , Schizophrenia/drug therapy , Smoking , Thiadiazoles/pharmacology , alpha7 Nicotinic Acetylcholine Receptor/agonists , Adult , Aged , Cognitive Dysfunction/complications , Cognitive Dysfunction/epidemiology , Comorbidity , Double-Blind Method , Female , Humans , Male , Middle Aged , Quinuclidines/administration & dosage , Quinuclidines/adverse effects , Schizophrenia/complications , Schizophrenia/epidemiology , Severity of Illness Index , Smoking/epidemiology , Thiadiazoles/administration & dosage , Thiadiazoles/adverse effects , Young Adult
9.
Cancer Chemother Pharmacol ; 80(6): 1209-1217, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29075855

ABSTRACT

PURPOSE: Patients with recurrent glioblastoma (rGBM) have a poor prognosis. Epidermal growth factor receptor (EGFR) gene amplification is present in ~ 50% of glioblastomas (GBMs). Depatuxizumab mafodotin (depatux-m), formerly ABT-414, is an antibody-drug conjugate that preferentially binds cells with EGFR amplification, is internalized and releases a potent antimicrotubule agent, monomethyl auristatin F (MMAF). Here we report the safety, pharmacokinetics, and efficacy of depatux-m monotherapy at the recommended Phase 2 dose (RPTD) in patients with EGFR-amplified, rGBM. METHODS: M12-356 (NCT01800695) is an open-label study with three escalation and expansion cohorts. Sixty-six patients with EGFR-amplified, rGBM were treated with depatux-m monotherapy at 1.25 mg/kg intravenously every 2 weeks. Adults with measurable rGBM, who were bevacizumab-naïve, with EGFR amplification were eligible. RESULTS: Among 66 patients, median age was 58 years (range 35-80). All patients were previously treated with radiotherapy/temozolomide. The most common adverse events (AEs) were eye related (91%), including blurred vision (65%), dry eye (29%), keratitis, and photophobia (27% each). Grade 3/4 AEs occurred in 42% of all patients, and ocular Grade 3/4 AEs occurred in 33% of patients overall. One patient (2%) had a Grade 4 ocular AE. Ocular AEs were manageable and usually resolved once treatment with depatux-m ceased. The objective response rate was 6.8%, the 6-month progression-free survival rate was 28.8%, and the 6-month overall survival rate was 72.5%. CONCLUSION: Depatux-m monotherapy displayed frequent but mostly Grade 1/2 ocular toxicities. A PFS6 of 28.8% was observed in this rGBM population, warranting further study.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Glioblastoma/drug therapy , Immunoconjugates/therapeutic use , Adult , Aged , Aged, 80 and over , ErbB Receptors/genetics , Female , Glioblastoma/pathology , Humans , Male , Middle Aged
10.
JMIR Mhealth Uhealth ; 5(2): e18, 2017 Feb 21.
Article in English | MEDLINE | ID: mdl-28223265

ABSTRACT

BACKGROUND: Accurately monitoring and collecting drug adherence data can allow for better understanding and interpretation of the outcomes of clinical trials. Most clinical trials use a combination of pill counts and self-reported data to measure drug adherence, despite the drawbacks of relying on these types of indirect measures. It is assumed that doses are taken, but the exact timing of these events is often incomplete and imprecise. OBJECTIVE: The objective of this pilot study was to evaluate the use of a novel artificial intelligence (AI) platform (AiCure) on mobile devices for measuring medication adherence, compared with modified directly observed therapy (mDOT) in a substudy of a Phase 2 trial of the α7 nicotinic receptor agonist (ABT-126) in subjects with schizophrenia. METHODS: AI platform generated adherence measures were compared with adherence inferred from drug concentration measurements. RESULTS: The mean cumulative pharmacokinetic adherence over 24 weeks was 89.7% (standard deviation [SD] 24.92) for subjects receiving ABT-126 who were monitored using the AI platform, compared with 71.9% (SD 39.81) for subjects receiving ABT-126 who were monitored by mDOT. The difference was 17.9% (95% CI -2 to 37.7; P=.08). CONCLUSIONS: Using drug levels, this substudy demonstrates the potential of AI platforms to increase adherence, rapidly detect nonadherence, and predict future nonadherence. Subjects monitored using the AI platform demonstrated a percentage change in adherence of 25% over the mDOT group. Subjects were able to use the technology successfully for up to 6 months in an ambulatory setting with early termination rates that are comparable to subjects outside of the substudy. TRIAL REGISTRATION: ClinicalTrials.gov NCT01655680 https://clinicaltrials.gov/ct2/show/NCT01655680?term=NCT01655680.

11.
Am J Psychiatry ; 173(8): 827-35, 2016 Aug 01.
Article in English | MEDLINE | ID: mdl-26940805

ABSTRACT

OBJECTIVE: The authors sought to evaluate the efficacy and safety of ABT-126, a selective α7 nicotinic receptor partial agonist, in stable patients with schizophrenia. METHOD: A 12-week, double-blind, placebo-controlled, parallel-group phase 2 study was conducted in 22 centers in the United States. Clinically stable patients with schizophrenia were randomly assigned to receive once-daily dosing with 10 mg of ABT-126, 25 mg of ABT-126, or placebo. The primary efficacy measure was change from baseline to week 12 on the MATRICS Consensus Cognitive Battery (MCCB) composite score compared with placebo, tested by a one-sided t test. Secondary measures included MCCB domain scores and UCSD Performance-Based Skills Assessment total score, each tested by two-sided t tests. RESULTS: A total of 207 subjects were randomized, of whom 165 (81%) completed the study. ABT-126 showed an improvement that fell short of significance on the MCCB composite score at week 12 (least squares mean difference from placebo, 1.3 and 1.5 for the 10 mg and 25 mg groups, respectively). A significant treatment-by-smoking status interaction was observed on the mean change from baseline to final MCCB composite score: nonsmokers (N=69) demonstrated a difference from placebo of 2.9 (SE=1.4) in the 10 mg group and 5.2 (SE=1.6) in the 25 mg group, whereas no differences were observed in smokers (N=113). Among the nonsmokers in the ABT-126 25 mg group (N=19), significant improvements compared with placebo occurred at final assessment for verbal learning (least squares mean difference=5.5, SE=1.9), working memory (least squares mean difference=5.4, SE=2.0), and attention/vigilance (least squares mean difference=8.7, SE=2.5). The most frequently reported adverse events for ABT-126 were dizziness, diarrhea, and fatigue (all <8% incidence). CONCLUSIONS: ABT-126 demonstrated a procognitive effect in nonsmoking subjects, particularly in verbal learning, working memory, and attention.


Subject(s)
Cognitive Dysfunction/drug therapy , Cognitive Dysfunction/psychology , Nootropic Agents/therapeutic use , Quinuclidines/therapeutic use , Schizophrenia/drug therapy , Schizophrenic Psychology , Thiadiazoles/therapeutic use , alpha7 Nicotinic Acetylcholine Receptor/agonists , Adult , Attention/drug effects , Cognitive Dysfunction/diagnosis , Dose-Response Relationship, Drug , Double-Blind Method , Female , Humans , Male , Memory, Short-Term/drug effects , Middle Aged , Neuropsychological Tests/statistics & numerical data , Nootropic Agents/adverse effects , Psychometrics , Quinuclidines/adverse effects , Schizophrenia/diagnosis , Smoking/adverse effects , Thiadiazoles/adverse effects , Treatment Outcome , Verbal Learning/drug effects
12.
J Clin Pharmacol ; 56(9): 1151-64, 2016 09.
Article in English | MEDLINE | ID: mdl-26634893

ABSTRACT

Accounting for subject nonadherence and eliminating inappropriate subjects in clinical trials are critical elements of a successful study. Nonadherence can increase variance, lower study power, and reduce the magnitude of treatment effects. Inappropriate subjects (including those who do not have the illness under study, fail to report exclusionary conditions, falsely report medication adherence, or participate in concurrent trials) confound safety and efficacy signals. This paper, a product of the International Society for CNS Clinical Trial Methodology (ISCTM) Working Group on Nonadherence in Clinical Trials, explores and models nonadherence in clinical trials and puts forth specific recommendations to identify and mitigate its negative effects. These include statistical analyses of nonadherence data, novel protocol design, and the use of biomarkers, subject registries, and/or medication adherence technologies.


Subject(s)
Clinical Trials as Topic/standards , Internationality , Patient Compliance/psychology , Clinical Trials as Topic/methods , Female , Humans , Male , Medication Adherence/psychology , Patient Participation/methods , Patient Participation/psychology
13.
Schizophr Bull ; 40(6): 1433-42, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24516190

ABSTRACT

INTRODUCTION: ABT-288 is a highly potent histamine-3 receptor antagonist that has demonstrated pro-cognitive effects in preclinical models relevant to schizophrenia. This study evaluated the efficacy and safety of two doses of ABT-288 in the treatment of cognitive impairment associated with schizophrenia. METHODS: A randomized, double-blind, placebo-controlled, parallel-group 12-week study was conducted at 23 centers in the United States. Clinically stable subjects with schizophrenia were randomized in an equal ratio to ABT-288 10 mg, ABT-288 25 mg, or placebo once daily while continuing their antipsychotic regimen. The primary efficacy measure was the change from baseline to day 84 evaluation on the Measurement and Treatment Research to Improve Cognition in Schizophrenia Consensus Cognitive Battery (MCCB) composite score vs placebo. Secondary measures included cognitive functioning and psychiatric scales. Safety assessments and sparse pharmacokinetic sampling were also conducted. RESULTS: A total of 214 subjects were randomized. The mean baseline MCCB composite score was 28.4. Approximately 80% of subjects completed the study. The MCCB composite score mean change from baseline to day 84 was numerically worse for both the 10 mg (1.90, P = .618) and 25 mg (0.64, P = .946) doses of ABT-288 vs placebo (2.19). Results from the secondary measures were consistent with the primary analysis. Subjects' schizophrenia symptoms remained stable throughout the study as evidenced by stable Positive and Negative Syndrome Scale scores. Overall, study medication was tolerated; however, an increased incidence of psychosis-related and sleep-related adverse events was associated with ABT-288. DISCUSSION: Neither dose of ABT-288 resulted in cognitive improvement in clinically stable adults with schizophrenia.


Subject(s)
Cognition Disorders/drug therapy , Histamine H3 Antagonists/pharmacology , Pyridazines/pharmacology , Pyrroles/pharmacology , Schizophrenia/drug therapy , Adult , Cognition Disorders/etiology , Double-Blind Method , Female , Histamine H3 Antagonists/administration & dosage , Histamine H3 Antagonists/adverse effects , Humans , Male , Middle Aged , Pyridazines/administration & dosage , Pyridazines/adverse effects , Pyrroles/administration & dosage , Pyrroles/adverse effects , Schizophrenia/complications , Treatment Failure
14.
J Psychopharmacol ; 27(10): 894-902, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23926239

ABSTRACT

Abnormal serotonin type 1A (5-HT1A) receptor function and binding have been implicated in the pathophysiology of mood disorders. Preclinical studies have consistently shown that stress decreases the gene expression of 5-HT1A receptors in experimental animals, and that the associated increase in hormone secretion plays a crucial role in mediating this effect. Chronic administration of the mood stabilizers lithium and divalproex (valproate semisodium) reduces glucocorticoid signaling and function in the hippocampus. Lithium has further been shown to enhance 5-HT1A receptor function. To assess whether these effects translate to human subject with bipolar disorder (BD), positron emission tomography (PET) and [18F]trans-4-fluoro-N-(2-[4-(2-methoxyphenyl) piperazino]-ethyl)-N-(2-pyridyl) cyclohexanecarboxamide ([(18)F]FCWAY) were used to acquire PET images of 5-HT1A receptor binding in 10 subjects with BD, before and after treatment with lithium or divalproex. Mean 5-HT1A binding potential (BPP) significantly increased following mood stabilizer treatment, most prominently in the mesiotemporal cortex (hippocampus plus amygdala). When mood state was also controlled for, treatment was associated with increases in BPP in widespread cortical areas. These preliminary findings are consistent with the hypothesis that these mood stabilizers enhance 5-HT1A receptor expression in BD, which may underscore an important component of these agents' mechanism of action.


Subject(s)
Bipolar Disorder/drug therapy , Bipolar Disorder/metabolism , Lithium Compounds/pharmacology , Lithium Compounds/therapeutic use , Receptor, Serotonin, 5-HT1A/metabolism , Valproic Acid/pharmacology , Valproic Acid/therapeutic use , Adult , Amygdala/diagnostic imaging , Amygdala/drug effects , Amygdala/metabolism , Bipolar Disorder/diagnostic imaging , Female , Functional Neuroimaging , Hippocampus/diagnostic imaging , Hippocampus/drug effects , Hippocampus/metabolism , Humans , Male , Middle Aged , Radioligand Assay , Radionuclide Imaging , Young Adult
15.
Eur Neuropsychopharmacol ; 23(8): 822-9, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23434290

ABSTRACT

Multiple lines of evidence suggest that serotonin type 1A (5-HT(1A)) receptor dysfunction is involved in the pathophysiology of mood disorders, and that alterations in 5-HT(1A) receptor function play a role in the mechanisms of antidepressant and mood stabilizer treatment. The literature is in disagreement, however, as to whether 5-HT(1A) receptor binding abnormalities exist in bipolar disorder (BD). We acquired PET images of 5-HT(1A) receptor binding in 26 unmedicated BD subjects and 37 healthy controls using [¹8F]FCWAY, a highly selective 5-HT(1A) receptor radio-ligand. The mean 5-HT(1A) receptor binding potential (BP(P)) was significantly lower in BD subjects compared to controls in cortical regions where 5-HT(1A) receptors are expressed post-synaptically, most prominently in the mesiotemporal cortex. Post-hoc assessments involving other receptor specific binding parameters suggested that this difference particularly affected the females with BD. The mean BPP did not differ between groups in the raphe nucleus, however, where 5-HT(1A) receptors are predominantly expressed pre-synaptically. Across subjects the BPP in the mesiotemporal cortex was inversely correlated with trough plasma cortisol levels, consistent with preclinical literature indicating that hippocampal 5-HT(1A) receptor expression is inhibited by glucocorticoid receptor stimulation. These findings suggest that 5-HT(1A) receptor binding is abnormally reduced in BD, and this abnormality may particularly involve the postsynaptic 5-HT(1A) receptor system of individuals with a tendency toward cortisol hypersecretion.


Subject(s)
Bipolar Disorder/metabolism , Cerebral Cortex/metabolism , Down-Regulation , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Post-Synaptic Density/metabolism , Receptor, Serotonin, 5-HT1A/metabolism , Adrenal Cortex/metabolism , Adrenal Cortex/physiopathology , Adrenocortical Hyperfunction/physiopathology , Adult , Bipolar Disorder/blood , Bipolar Disorder/physiopathology , Diagnostic and Statistical Manual of Mental Disorders , Female , Humans , Hydrocortisone/blood , Hydrocortisone/metabolism , Male , Middle Aged , Positron-Emission Tomography , Radioligand Assay , Sex Characteristics , Young Adult
16.
Neuropsychopharmacology ; 38(3): 405-13, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23032073

ABSTRACT

Dysregulation of the neuronal nicotinic acetylcholine receptor (NNR) system has been implicated in attention-deficit/hyperactivity disorder (ADHD), and nicotinic agonists improve attention across preclinical species and humans. Hence, a randomized, double-blind, placebo-controlled, crossover study was designed to determine the safety and efficacy of a novel α4ß2 NNR agonist (ABT-894 (3-(5,6-dichloro-pyridin-3-yl)-1(S),5 (S)-3,6-diazabicyclo[3.2.0]heptane)) in adults with ADHD. Participants (N=243) were randomized to one of four dose regimens of ABT-894 (1, 2, and 4 mg once daily (QD)) or 4 mg twice daily (BID) or the active comparator atomoxetine (40 mg BID) vs placebo for 28 days. Following a 2-week washout period, participants crossed over to the alternative treatment condition (active or placebo) for an additional 28 days. Primary efficacy was based on an investigator-rated Conners' Adult ADHD Rating Scale (CAARS:Inv) Total score at the end of each 4-week treatment period. Additional secondary outcome measures were assessed. A total of 238 patients were assessed for safety end points, 236 patients were included in the intent-to-treat data set, and 196 were included in the completers data set, which was the prespecified, primary data set for efficacy. Both the 4 mg BID ABT-894 and atomoxetine groups demonstrated significant improvement on the primary outcome compared with placebo. Several secondary outcome measures were also significantly improved with 4 mg BID ABT-894. Overall, ABT-894 was well tolerated at all dose levels. These results provide initial proof of concept for the use of α4ß2 agonists in the treatment of adults with ADHD. Further investigation of ABT-894, including higher doses, is therefore warranted.


Subject(s)
Attention Deficit Disorder with Hyperactivity/drug therapy , Attention Deficit Disorder with Hyperactivity/epidemiology , Nicotinic Agonists/therapeutic use , Receptors, Nicotinic/physiology , Adult , Cross-Over Studies , Double-Blind Method , Female , Humans , Male , Middle Aged , Young Adult
17.
J Clin Psychiatry ; 73(6): 783-9, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22795204

ABSTRACT

OBJECTIVE: ABT-089, an α4ß2 neuronal nicotinic receptor partial agonist (generic name pozanicline), has demonstrated efficacy in adults with attention-deficit/hyperactivity disorder (ADHD) at doses of 40 mg once daily and 40 mg twice daily. The purpose of this exploratory pilot study was to obtain initial safety, tolerability, and efficacy data for an ABT-089 80-mg once-daily regimen to inform a decision of whether to include an 80-mg once-daily dose regimen in subsequent, definitive (phase 3) efficacy studies. METHOD: This phase 2, randomized, double-blind, parallel-group, placebo-controlled pilot study was conducted at 12 sites from March to August 2008. A screening/washout period of up to 4 weeks was followed by an 8-week double-blind treatment period. Eligible subjects met DSM-IV-TR criteria for ADHD and were randomized in a 1:1:1 ratio to ABT-089 40 mg once daily, ABT-089 80 mg once daily, or placebo. The primary efficacy variable was reduction from baseline to the final evaluation in the investigator-rated Conners' Adult ADHD Rating Scale for each active treatment group versus placebo. Safety assessments and pharmacokinetic sampling were also conducted. RESULTS: A total of 160 subjects were randomized, with 137 (86%) completing the trial. No statistically significant treatment effects were observed with either ABT-089 dose for any efficacy measures. The most commonly reported adverse events in the active treatment groups were nasopharyngitis (6.6%), upper respiratory tract infection (6.6%), and somnolence (5.7%). The incidence of adverse events did not differ significantly between active groups and placebo. There were no clinically significant laboratory, electrocardiogram, or physical examination findings. CONCLUSIONS: ABT-089 was generally well tolerated at doses up to 80 mg. Because ABT-089 is a weak partial neuronal nicotinic receptor agonist, the results may not predict the potential efficacy for other, more potent neuronal nicotinic receptor agonists. TRIAL REGISTRATION: ClinicalTrials.gov identifier: NCT00640185.


Subject(s)
Attention Deficit Disorder with Hyperactivity/drug therapy , Nicotinic Agonists/adverse effects , Nicotinic Agonists/therapeutic use , Pyridines/adverse effects , Pyridines/therapeutic use , Pyrrolidines/adverse effects , Pyrrolidines/therapeutic use , Adolescent , Adult , Dose-Response Relationship, Drug , Double-Blind Method , Drug Partial Agonism , Female , Humans , Male , Middle Aged , Pilot Projects , Psychiatric Status Rating Scales/statistics & numerical data , Pyridines/agonists , Pyridines/pharmacokinetics , Pyrrolidines/agonists , Pyrrolidines/pharmacokinetics , Receptors, Nicotinic/drug effects
18.
Int J Psychophysiol ; 80(3): 182-91, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21414364

ABSTRACT

Electrophysiology, behavioral, and neuroimaging studies have revealed sex-related differences in autonomic cardiac control, as reflected in measurements of heart rate variability (HRV). Imaging studies indicate that the neurobiological correlates of autonomic nervous system (ANS) function can be investigated by measuring indices of HRV during the performance of mildly strenuous motor tasks or mildly stressful cognitive tasks. In this preliminary study, fifteen male and seven female healthy subjects underwent H(2)(15)O-positron emission tomography (PET) and electrocardiograph (ECG) recording while performing a handgrip motor task and an n-back task. Indices of HRV were calculated and correlated with regional cerebral blood flow (rCBF). We hypothesized that sex differences would be evident in brain regions known to participate in autonomic regulation: the anterior insula, the anterior cingulate cortex, the orbitofrontal cortex, and the amygdala. Our study found that associations between rCBF and parasympathetic indices differed significantly between female and male subjects in the amygdala. Females showed a positive correlation between rCBF and parasympathetic indices while males exhibited negative correlations. This differential correlation of amygdala rCBF and parasympathetic activity between males and females may reflect differences in parasympathetic/sympathetic balance between sexes, consistent with known sexual dimorphism in the amygdala and closely related structures such as the hypothalamus. These preliminary imaging results are consistent with earlier reports of significant correlation between brain activity and HRV, and extend these findings by demonstrating prominent sex differences in the neural control of the ANS. While the generalizability of our results was limited by the small size of the study samples, the relatively robust effect size of the differences found between groups encourages further work in larger samples to characterize sex differences in the neural correlates of autonomic arousal.


Subject(s)
Arousal/physiology , Autonomic Nervous System/diagnostic imaging , Autonomic Nervous System/physiology , Positron-Emission Tomography/methods , Sex Characteristics , Adult , Amygdala/blood supply , Amygdala/diagnostic imaging , Amygdala/physiology , Cerebrovascular Circulation/physiology , Female , Frontal Lobe/blood supply , Frontal Lobe/diagnostic imaging , Frontal Lobe/physiology , Gyrus Cinguli/blood supply , Gyrus Cinguli/diagnostic imaging , Gyrus Cinguli/physiology , Hand Strength/physiology , Humans , Male , Oxygen Radioisotopes , Pilot Projects , Young Adult
19.
Psychiatry Res ; 191(1): 1-8, 2011 Jan 30.
Article in English | MEDLINE | ID: mdl-21129936

ABSTRACT

Research indicates that major depressive disorder (MDD) is associated with alterations in autonomic control, particularly cardiac control as measured by heart rate variability (HRV). In this preliminary study, we investigated the neural correlates of autonomic control by measuring both HRV and associated brain activity during the performance of mildly stressful tasks. Medically healthy female subjects with MDD (N=10) and healthy controls (N=7) underwent H(2)(15)O-positron emission tomography (PET) and electrocardiographic ECG recording while performing a handgrip motor task and an n-back task. Indices of HRV were calculated and correlated with regional cerebral blood flow (rCBF). Differences in the rCBF and HRV correlations between depressed and healthy subjects were evident in both the medial and lateral orbital cortices. In addition, these areas appeared to be involved in different facets of autonomic control with regard to sympathetic or parasympathetic dominance of cardiac control. These results are consistent with the known roles of networks within the orbital cortex in both autonomic control and the pathophysiology of MDD.


Subject(s)
Cerebrovascular Circulation/physiology , Cognition/physiology , Depressive Disorder/physiopathology , Hand Strength/physiology , Heart Rate/physiology , Adult , Analysis of Variance , Brain/diagnostic imaging , Brain Mapping , Depressive Disorder/diagnostic imaging , Depressive Disorder/pathology , Depressive Disorder/psychology , Electrocardiography/methods , Female , Humans , Neuropsychological Tests , Positron-Emission Tomography/methods , Statistics as Topic , Young Adult
20.
Biol Psychiatry ; 69(4): 336-43, 2011 Feb 15.
Article in English | MEDLINE | ID: mdl-21094939

ABSTRACT

BACKGROUND: Increased activity of the habenula has been implicated in the etiology of major depressive disorder (MDD), in which reductions in habenula volume are present after death. We conducted the first magnetic resonance imaging analysis of habenula volume in MDD and bipolar disorder (BD). METHODS: High-resolution images (resolution approximately .4 mm(3)) were acquired with a 3T scanner, and a pulse sequence was optimized for tissue contrast resolution. The habenula was manually segmented by one rater blind to diagnosis. Seventy-four healthy control subjects (HC) were compared with both medicated (lithium/divalproex, n = 15) and unmedicated, depressed BD (n = 22) patients; unmedicated, depressed MDD patients (n = 28); and unmedicated MDD patients in remission (n = 32). RESULTS: The unmedicated BD patients displayed significantly smaller absolute (p < .01) and normalized (p < .05) habenula volumes than the HC subjects. In post hoc assessments analyzing men and women separately, the currently-depressed women with MDD had smaller absolute (p < .05) habenula volumes than the HC women. None of the other psychiatric groups differed significantly from the HC group. CONCLUSIONS: We provide further evidence for the involvement of the habenula in affective illness but suggest that a reduction in volume might be more pronounced in unmedicated, depressed BD subjects and female currently depressed MDD subjects. The habenula plays major roles in the long-term modification of monoamine transmission and behavioral responses to stress and in the suppression of dopamine cell activity after the absence of an expected reward. A reduction in habenula volume might thus have functional consequences that contribute to the risk for developing affective disease.


Subject(s)
Bipolar Disorder/pathology , Depressive Disorder, Major/pathology , Habenula/pathology , Adult , Analysis of Variance , Antidepressive Agents/therapeutic use , Antimanic Agents/therapeutic use , Bipolar Disorder/drug therapy , Brain Mapping , Depressive Disorder, Major/drug therapy , Female , Fluoxetine/therapeutic use , Humans , Lithium Carbonate/therapeutic use , Magnetic Resonance Imaging , Male , Middle Aged , Organ Size , Valproic Acid/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...