Treatment of aggression with topiramate in male borderline patients, part II: 18-month follow-up

2008 ◽  
Vol 23 (2) ◽  
pp. 115-117 ◽  
Author(s):  
Marius K. Nickel ◽  
Thomas H. Loew

AbstractObjectiveWe previously tested topiramate, an anticonvulsant, in the treatment of aggression in men with borderline personality disorder (BPD) (Nickel M, Nickel C, Kaplan P, Lahmann C, Mühlbacher M, Tritt K, et al. Treatment of aggression with topiramate in male borderline patients: a double-blind, placebo-controlled study. Biol Psychiatry 2005;57:495–9), and found significant changes on most scales of the state–trait anger expression inventory (STAXI) and significant weight loss eight weeks later. The aim of this trial was to assess topiramate's efficacy in the long-term therapy for aggression in men with BPD.MethodsThis 18-month follow-up observation, in which the previous patients (topiramate group: n = 22; former placebo group: n = 22) were examined bianually, was carried out.ResultsAccording to the intent-to-treat principle, significant changes on all scales of the STAXI were observed in the subjects treated with topiramate. Additional significant weight loss was observed. All subjects tolerated topiramate relatively well.ConclusionsTopiramate appears to be an effective, relatively safe agent in the long-term treatment of patients with BPD. Mild, non-transient weight loss can be expected.

Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 2925-2925 ◽  
Author(s):  
Ricardo Spielberger ◽  
Christos Emmanouilides ◽  
William Bensinger ◽  
Alan Rong ◽  
Alessandra Cesano ◽  
...  

Abstract Oral mucositis (OM) is a severely debilitating side effect of chemoradiotherapy that often causes significant pain, diminished quality of life, and increased risk of infections. Palifermin decreases the incidence and duration of severe OM in HM pts receiving myelotoxic therapy and HSCT. Palifermin safety and efficacy have not been established in the non-HM setting. Since the rHuKGF receptor is not expressed by HM, palifermin is not expected to interfere with long-term disease outcomes in this pt population. Aim: We assessed palifermin’s effects on the long-term disease outcomes (survival, disease progression, secondary malignancies) in pts with HM. Methods: Long-term follow-up data were pooled from a 1998 phase 2, double-blind, placebo-controlled study (n=86) and a 2000 double-blind, placebo-controlled phase 3 study (n=212). The analysis included 152 pts treated with palifermin and 146 pts treated with placebo. Pts were assessed at 6-month intervals during the first year and annually thereafter until death or loss to follow-up. The Kaplan-Meier (K–M) method provided estimates of the safety endpoints. Data reported here are as of August 2004. Results: There were 298 pts (152 palifermin: 146 placebo) monitored for long-term follow-up. The median follow-up period was 23.3 months for palifermin and 23.5 months for placebo. The overall survival and progression-free survival curves (p=0.474 and p=0.253, respectively) are similar between the palifermin and placebo groups. Secondary malignancies occurred in only 6 of 152 (3%) palifermin and 5 of 146 (4%) placebo pts. All secondary malignancies were myelodysplastic syndromes: 9 patients with diagnoses of Non-Hodgkin’s lymphoma and 2 patients of Hodgkin’s Disease. The number of deaths was similar between the groups (30% palifermin; 27% placebo); most deaths occurred within 12 months of randomization and were attributable to the underlying HM disease. Conclusion: Use of palifermin for the prevention of severe OM has shown no negative impact on long-term disease outcomes, including survival, in the HSCT setting for patients with HM.


2021 ◽  
Vol 7 (1) ◽  
Author(s):  
Christoph U. Correll ◽  
Kenneth S. Koblan ◽  
Seth C. Hopkins ◽  
Yan Li ◽  
Heather Dworak ◽  
...  

AbstractUlotaront, a trace amine-associated receptor 1 (TAAR1) and serotonin 5-HT1A receptors agonist, has demonstrated efficacy in the treatment of patients with an acute exacerbation of schizophrenia in a 4-week, double-blind, placebo-controlled study. The aim of this 26-week open-label extension study was to evaluate the safety and effectiveness of ulotaront (25/50/75 mg/d) in patients who completed the initial 4-week study. Of the 193 4-week completers, 157 patients (81.3%) continued into the open-label extension study; 66.9% were completers. Among all extension phase patients, treatment with ulotaront was associated with minimal changes in body weight (mean [SD] change from double-blind baseline: −0.3 [3.7] kg), cholesterol (median change, −2.0 mg/dL), triglycerides (median, −5.0 mg/dL), and prolactin (female, median, −3.4 ng/mL; male, median, −2.7 ng/mL). Movement disorder scales showed no extrapyramidal effects. Twenty-six weeks of extension phase treatment was associated with a mean (95% CI) observed change from open-label baseline in the PANSS total score of −22.6 (−25.6, −19.6; effect size, 1.46), and a mean (95% CI) change in the CGI-Severity score of −1.0 (−1.2, −0.8; effect size, 1.07). Long-term treatment with the TAAR1 agonist ulotaront, in the daily dose range of 25–75 mg, was characterized by a relatively high completion rate, an adverse event profile notable for the absence of extrapyramidal-related adverse effects, a low liability for adverse weight and metabolic effects, and no effect on prolactin levels. Additional studies are needed to further confirm the long-term efficacy and safety of ulotaront.


Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 4811-4811
Author(s):  
Jean El-Cheikh ◽  
Anne-Marie Stoppa ◽  
Reda Bouabdallah ◽  
Diane Coso ◽  
Jean-Marc Schiano de Collela ◽  
...  

Abstract Bortezomib is a first-in-class proteasome inhibitor approved for the treatment of MM patients who have received at least one prior therapy. Classically, patients receive bortezomib 1.3 or 1.0 mg/m2 by IV bolus on days 1, 4, 8, and 11 of a 21-day cycle, associated or not to dexamethasone, for a total number of 8 cycles. Such administration schema is associated with a remarkable anti-tumor activity and response. However, a significant number of patients who are initially responders to bortezomib, will progress after drug discontinuation, raising the question of long term or maintenance treatment with bortezomib. The objective of this analysis was to evaluate the tolerance and safety profiles of long term treatment with bortezomib in a cohort of 16 patients with relapsed and/or refractory MM treated in a single institution. Eligible patients for this analysis are those who had relapsed MM, and who continued to receive bortezomib (1.3 or 1.0 mg/m2) as a long term therapy beyond the classical 8 cycles. All medical charts were uniformly reviewed in detail for assessment of toxicity, safety and response. The median age was 53 (range, 27–74) years. The majority of patients had already received at least one prior autologous or allogeneic stem cell transplantation (n=12; 75%). Also, 12 patients (75%) had received prior treatment with thalidomide at a median dose of 200 mg/day, for a median duration of 7 months. Before treatment with bortezomib, 7 patients (44%) already had some form of peripheral neuropathy (PN). With a median follow-up of 16 months from bortezomib initiation, patients from this series received a median of 10 (range, 9–16) cycles of bortezomib administered over a median period of 11 (range, 7–35) months. Overall, 6 patients had evidence of bortezomib-associated PN (38%; 4 grade 1, 1 grade 2 and 1 grade 3; sensory symptoms in all cases). Other bortezomib-related toxicities included thromobopenia (n=8; 50%; 1 grade 1, 5 grade 2, and 2 grade 3–4). General fatigue was also common and was encountered in 5 (31%) patients. Overall, bortezomib-associated toxicities led to dose reduction or increase of treatment cycle duration in 9 patients (56%), but none of the patients had to definitively discontinue treatment because of unacceptable toxicity. At last follow-up, 6 patients are still receiving bortezomib, 4 patients died from disease progression, no patient died from treatment-related causes, and the remaining 12 patients are still alive. Long term treatment with bortezomib was associated with an objective disease response rate in 87% (95%CI, 60–98%) of patients (n=14; 3 CR, 8 VGPR, 3 PR). The Kaplan-Meier estimate for overall survival is shown in the figure below. Though relatively small, results from this series suggest that long term treatment with bortezomib is feasible. Toxicity, tolerance and safety profiles of long term treatment are comparable to those observed with the standard schedule and manageable after dose reduction. Therefore, prospective studies aiming to optimize bortezomib administration schedule and duration (beyond the classical 8 cycles) are warranted, since such long term treatment can yield major objective disease response. Figure Figure


2020 ◽  
Vol 21 (1) ◽  
Author(s):  
Andrew Menzies-Gow ◽  
Sandhia Ponnarambil ◽  
John Downie ◽  
Karin Bowen ◽  
Åsa Hellqvist ◽  
...  

Abstract Background Tezepelumab is a human monoclonal antibody that blocks the activity of the epithelial cytokine thymic stromal lymphopoietin. The efficacy, safety and oral corticosteroid-sparing potential of tezepelumab are being investigated in two ongoing, phase 3, randomized, double-blind, placebo-controlled studies (NAVIGATOR [NCT03347279] and SOURCE [NCT03406078]). DESTINATION (NCT03706079) is a long-term extension (LTE) of these studies. Methods DESTINATION is a randomized, double-blind, placebo-controlled LTE study in adults (18–80 years old) and adolescents (12–17 years old) with severe, uncontrolled asthma who are receiving treatment with medium- or high-dose inhaled corticosteroids plus at least one additional controller medication with or without oral corticosteroids. The study population will comprise patients who complete the 52- and 48-week NAVIGATOR and SOURCE studies, respectively. Patients who were randomized to receive tezepelumab 210 mg every 4 weeks (Q4W) in either predecessor study will continue to receive this regimen for 1 year; those who were previously randomized to receive placebo will be re-randomized (1:1) to receive either tezepelumab 210 mg Q4W or placebo for 1 year. Patients will receive their prescribed controller medications throughout DESTINATION and study physicians will have the opportunity to down- or up-titrate dosage of these medications, if appropriate. The primary objective is to evaluate the long-term safety and tolerability of tezepelumab over 104 weeks (inclusive of the treatment period of either predecessor study). The secondary objective is to assess the long-term effect of tezepelumab on asthma exacerbations. Patients recruited from SOURCE will be followed up post-treatment for 12 weeks. Patients recruited from NAVIGATOR who complete 100 weeks of tezepelumab treatment will be eligible for either 12 weeks of follow-up or a 36-week extended follow-up during which the clinical benefit of tezepelumab after treatment cessation will be investigated. Discussion DESTINATION will evaluate the long-term safety, tolerability and efficacy of tezepelumab versus placebo with continued dosing for up to 2 years. DESTINATION will also evaluate the clinical effect of tezepelumab after treatment cessation. This LTE study aims to elucidate the long-term safety implications of receiving tezepelumab and to assess its potential long-term treatment benefits in patients with severe, uncontrolled asthma. Trial registration NCT03706079 (ClinicalTrials.gov). Registered 15 October 2018.


2017 ◽  
Vol 69 (3) ◽  
pp. 347-355 ◽  
Author(s):  
Arthur Kavanaugh ◽  
Philip J. Mease ◽  
Andreas M. Reimold ◽  
Hasan Tahir ◽  
Jürgen Rech ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document