Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 973
Filter
1.
Turk Psikiyatri Derg ; 35(3): 245-247, 2024.
Article in English, Turkish | MEDLINE | ID: mdl-39224997

ABSTRACT

Naltrexone is an µ opioid receptor antagonist that is used in alcohol and opiate use disorder. Naltrexone does not constitute tolerance and dependence, and cessation of the drug does not cause withdrawal symptoms. Sustained release form of naltrexone has been developed due to patient compliance issues. There is currently only one sustainedrelease form available in Turkey, which is inserted subcutaneously. In this case report, we present, a probable serious side effect of sustained release naltrexone implant. A 36 years old male with alcohol use disorder, developed a sudden clouding of consciousness one hour after the naltrexone implant application followed by anterograde amnesia in the next 8-10 hours. We were not able to detect any medical or neurological reasons for the altered mental status but after the removal of the naltrexone implant, the symptoms improved. To the best of our knowledge, this is the first case to report clouding of consciousness and anterograde amnesia after naltrexone implantation. Keywords: Naltrexone Implant, Side Effect, Alcohol Use Disorder, Lethargy, Consciousness.


Subject(s)
Naltrexone , Narcotic Antagonists , Humans , Naltrexone/adverse effects , Naltrexone/administration & dosage , Naltrexone/therapeutic use , Male , Narcotic Antagonists/adverse effects , Narcotic Antagonists/administration & dosage , Narcotic Antagonists/therapeutic use , Adult , Alcoholism/complications , Alcoholism/drug therapy , Diagnosis, Differential , Unconsciousness/chemically induced , Drug Implants/adverse effects
2.
J Opioid Manag ; 20(4): B14, 2024.
Article in English | MEDLINE | ID: mdl-39321068

ABSTRACT

BACKGROUND: Bone marrow transplant (BMT) offers potential cure for cancer and a spectrum of otherwise incur- able diseases. The BMT process can cause multi-systemic pain in patients with sickle cell disease (SCD) refractory to high-dose opioid analgesics during BMT because of their pre-existing opioid-tolerance. Because of frequent pain resulting in hyperalgesia and chronic opioid use, SCD patients undergoing BMT often experience excruciating pain uncontrolled by exceedingly high-dose opioids with severe and intolerable adverse effects. PURPOSE/HYPOTHESIS: There is a small but growing body of literature about the successful buprenorphine effect for SCD's chronic pain management that had failed sufficient pain relief by the traditional full-agonist opioids in the outpatient setting. However, the buprenorphine use for acute inpatient pain management has not been previously researched. Pilot prospective clinical trial with buprenorphine-based pain management for acute BMT-related pain was initiated for SCD patients' pain uncontrolled by full-agonist opioids. Procedures/data/observations: Buprenorphine was started as scheduled and as-needed analgesics, supplemented by full-agonist opioids upon consultation for uncontrolled BMT-related pain of SCD patients. Patients' 24-hour opioid requirement by morphine equivalent daily doses (MEDD) were assessed at 3 time points: 1)immediately before pain escalation; 2)consultation; 3)discharge. MEDDs were compared to those of patients treated with full-agonist opioids only. Cases treated by full-agonist opioids (morphine/hydromorphone/fentanyl/methadone/oxycodone) had MEDD escalation by 1230-16300% by discharge compared to immediately before BMT-related pain escalation. Buprenorphine-supported cases had significantly smaller MEDD increase by 220-317%. CONCLUSIONS/APPLICATIONS: Our case series suggests superior pain control by adding Buprenorphine prior to opioid dose escalation during BMT for SCD. Buprenorphine may provide the advantageous effect for other patients with complex pain background and experiencing difficult pain management during BMT due to pre-existing hyperalgesia and high opioid-tolerance. The evidence for buprenorphine's analgesic effect is moderate but growing, and more randomized controlled trials comparing the buprenorphine and other standard opioids are needed.


Subject(s)
Analgesics, Opioid , Anemia, Sickle Cell , Buprenorphine , Hematopoietic Stem Cell Transplantation , Pain, Intractable , Humans , Anemia, Sickle Cell/complications , Analgesics, Opioid/therapeutic use , Analgesics, Opioid/administration & dosage , Analgesics, Opioid/adverse effects , Buprenorphine/therapeutic use , Buprenorphine/administration & dosage , Buprenorphine/adverse effects , Hematopoietic Stem Cell Transplantation/adverse effects , Male , Adult , Pain, Intractable/drug therapy , Pain, Intractable/etiology , Pain, Intractable/diagnosis , Female , Pilot Projects , Prospective Studies , Narcotic Antagonists/adverse effects , Narcotic Antagonists/therapeutic use , Narcotic Antagonists/administration & dosage , Treatment Outcome , Pain Measurement , Middle Aged , Young Adult
3.
JAMA Netw Open ; 7(9): e2435895, 2024 Sep 03.
Article in English | MEDLINE | ID: mdl-39331392

ABSTRACT

Importance: Buprenorphine treatment of opioid use disorder (OUD) is safe and effective, but opioid withdrawal during treatment initiation is associated with poor retention in care. As fentanyl has replaced heroin in the drug supply, case reports and surveys have indicated increased concern for buprenorphine-precipitated withdrawal (PW); however, some observational studies have found a low incidence of PW. Objective: To estimate buprenorphine PW incidence and assess factors associated with PW among emergency department (ED) or hospitalized patients. Design, Setting, and Participants: This retrospective cohort study at 3 academic hospitals in Philadelphia, Pennsylvania, included adults with OUD who underwent traditional or high-dose buprenorphine initiation between January 1, 2020, and December 31, 2021. Exclusion criteria included low-dose buprenorphine initiation and missing documentation of opioid withdrawal severity within 4 hours of receiving buprenorphine. Exposure: Buprenorphine initiation with an initial dose of at least 2 mg of sublingual buprenorphine after a Clinical Opiate Withdrawal Scale (COWS) score of 8 or higher. Additional exposures included 4 predefined factors potentially associated with PW: severity of opioid withdrawal before buprenorphine (COWS score of 8-12 vs ≥13), initial buprenorphine dose (2 vs 4 or ≥8 mg), body mass index (BMI) (<25 vs 25 to <30 or ≥30; calculated as weight in kilograms divided by height in meters squared), and urine fentanyl concentration (0 to <20 vs 20 to <200 or ≥200 ng/mL). Main Outcome and Measures: The main outcome was PW incidence, defined as a 5-point or greater increase in COWS score from immediately before to within 4 hours after buprenorphine initiation. Logistic regression was used to estimate the odds of PW associated with the 4 aforementioned predefined factors. Results: The cohort included 226 patients (150 [66.4%] male; mean [SD] age, 38.6 [10.8] years). Overall, 26 patients (11.5%) met criteria for PW. Among patients with PW, median change in COWS score was 9 points (IQR, 6-13 points). Of 123 patients with confirmed fentanyl use, 20 (16.3%) had PW. In unadjusted and adjusted models, BMI of 30 or greater compared with less than 25 (adjusted odds ratio [AOR], 5.12; 95% CI, 1.31-19.92) and urine fentanyl concentration of 200 ng/mL or greater compared with less than 20 ng/mL (AOR, 8.37; 95% CI, 1.60-43.89) were associated with PW. Conclusions and Relevance: In this retrospective cohort study, 11.5% of patients developed PW after buprenorphine initiation in ED or hospital settings. Future studies should confirm the rate of PW and assess whether bioaccumulated fentanyl is a risk factor for PW.


Subject(s)
Buprenorphine , Fentanyl , Opioid-Related Disorders , Substance Withdrawal Syndrome , Humans , Buprenorphine/adverse effects , Buprenorphine/therapeutic use , Fentanyl/adverse effects , Fentanyl/therapeutic use , Male , Female , Retrospective Studies , Adult , Middle Aged , Opioid-Related Disorders/drug therapy , Analgesics, Opioid/adverse effects , Analgesics, Opioid/therapeutic use , Opiate Substitution Treatment/methods , Opiate Substitution Treatment/adverse effects , Hospitalization/statistics & numerical data , Narcotic Antagonists/therapeutic use , Narcotic Antagonists/adverse effects , Narcotic Antagonists/administration & dosage , Philadelphia/epidemiology , Incidence
4.
Clin Pharmacol Drug Dev ; 13(9): 974-984, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39110083

ABSTRACT

The pharmacokinetics (PK) of naloxegol were characterized in pediatric subjects, aged 6 months or older to less than 18 years who either have or are at risk of developing opioid-induced constipation following single dose administration. Subjects grouped as aged 12 years or older to less than 18 years, 6 months or older to less than 12 years, and 6 months or older to less than 6 years, received a single oral dose of naloxegol at doses that were estimated to achieve plasma exposures comparable to adult 12.5- or 25-mg doses. Intensive and sparse plasma naloxegol samples were collected to assess naloxegol concentrations. Data were combined with previously collected adult PK data and used to estimate PK parameters using population PK analyses. Naloxegol PK was described using a 2-compartment model with Weibull-type absorption. Neither age nor body weight was identified as a significant covariate indicating similar PK properties in adult and pediatric subjects. PK estimates in the youngest age group were approximately 80% less than those in adults (12.5-mg equivalent dose). Exposures in the other pediatric groups were similar to those in adult equivalent doses. The PK of naloxegol were characterized as linear over the dose range, with no clinically significant covariates and comparable PK characteristics in adults and pediatric subjects aged 6 months or older.


Subject(s)
Analgesics, Opioid , Morphinans , Polyethylene Glycols , Humans , Morphinans/pharmacokinetics , Morphinans/administration & dosage , Morphinans/adverse effects , Child , Child, Preschool , Male , Analgesics, Opioid/pharmacokinetics , Analgesics, Opioid/administration & dosage , Analgesics, Opioid/adverse effects , Female , Adolescent , Infant , Polyethylene Glycols/pharmacokinetics , Polyethylene Glycols/administration & dosage , Polyethylene Glycols/adverse effects , Models, Biological , Narcotic Antagonists/pharmacokinetics , Narcotic Antagonists/administration & dosage , Narcotic Antagonists/adverse effects , Age Factors , Opioid-Induced Constipation , Adult , Administration, Oral
5.
JAMA ; 332(10): 805-816, 2024 09 10.
Article in English | MEDLINE | ID: mdl-39133511

ABSTRACT

Importance: Buprenorphine combined with naloxone is commonly used to treat opioid use disorders outside of pregnancy. In pregnancy, buprenorphine alone is generally recommended because of limited perinatal safety data on the combination product. Objective: To compare perinatal outcomes following prenatal exposure to buprenorphine with naloxone vs buprenorphine alone. Design, Settings, and Participants: Population-based cohort study using health care utilization data from Medicaid-insured beneficiaries in the US from 2000 to 2018. The cohort was restricted to pregnant individuals linked to their liveborn infants, with maternal Medicaid enrollment from 3 months before pregnancy to 1 month after delivery and infant enrollment for the first 3 months after birth, unless they died sooner. Exposure: Use of buprenorphine with naloxone vs buprenorphine alone during the first trimester based on outpatient dispensings. Main Outcomes and Measures: Outcomes included major congenital malformations, low birth weight, neonatal abstinence syndrome, neonatal intensive care unit admission, preterm birth, respiratory symptoms, small for gestational age, cesarean delivery, and maternal morbidity. Confounder-adjusted risk ratios were calculated using propensity score overlap weights. Results: This study identified 3369 pregnant individuals exposed to buprenorphine with naloxone during the first trimester (mean [SD] age, 28.8 [4.6] years) and 5326 exposed to buprenorphine alone or who switched from the combination to buprenorphine alone by the end of the first trimester (mean [SD] age, 28.3 [4.5] years). When comparing buprenorphine combined with naloxone with buprenorphine alone, a lower risk for neonatal abstinence syndrome (absolute risk, 37.4% vs 55.8%; weighted relative risk, 0.77 [95% CI, 0.70-0.84]) and a modestly lower risk for neonatal intensive care unit admission (absolute risk, 30.6% vs 34.9%; weighted relative risk, 0.91 [95% CI, 0.85-0.98]) and small for gestational age (absolute risk, 10.0% vs 12.4%; weighted relative risk, 0.86 [95% CI, 0.75-0.98]) was observed. For maternal morbidity, the comparative rates were 2.6% vs 2.9%, respectively, and the weighted relative risk was 0.90 (95% CI, 0.68-1.19). No differences were observed with respect to major congenital malformations overall, low birth weight, preterm birth, respiratory symptoms, or cesarean delivery. Results were consistent across sensitivity analyses. Conclusions and Relevance: There were similar and, in some instances, more favorable neonatal and maternal outcomes for pregnancies exposed to buprenorphine combined with naloxone compared with buprenorphine alone. For the outcomes assessed, compared with buprenorphine alone, buprenorphine with naloxone during pregnancy appears to be a safe treatment option. This supports the view that both formulations are reasonable options for the treatment of opioid use disorder in pregnancy, affirming flexibility in collaborative treatment decision-making.


Subject(s)
Buprenorphine, Naloxone Drug Combination , Buprenorphine , Narcotic Antagonists , Opioid-Related Disorders , Prenatal Exposure Delayed Effects , Adult , Female , Humans , Infant, Newborn , Pregnancy , Young Adult , Abnormalities, Drug-Induced/epidemiology , Buprenorphine/administration & dosage , Buprenorphine/adverse effects , Buprenorphine, Naloxone Drug Combination/administration & dosage , Buprenorphine, Naloxone Drug Combination/adverse effects , Cesarean Section/statistics & numerical data , Cohort Studies , Infant, Low Birth Weight , Infant, Small for Gestational Age , Narcotic Antagonists/administration & dosage , Narcotic Antagonists/adverse effects , Neonatal Abstinence Syndrome/drug therapy , Opiate Substitution Treatment/adverse effects , Opiate Substitution Treatment/methods , Opioid-Related Disorders/drug therapy , Pregnancy Complications/drug therapy , Pregnancy Outcome , Pregnancy Trimester, First , Premature Birth/chemically induced , Premature Birth/epidemiology , Prenatal Exposure Delayed Effects/chemically induced , Prenatal Exposure Delayed Effects/epidemiology , United States
6.
Support Care Cancer ; 32(8): 504, 2024 Jul 10.
Article in English | MEDLINE | ID: mdl-38985364

ABSTRACT

PURPOSE: In this study, we aimed to evaluate the safety and effectiveness of naldemedine for treating opioid-induced constipation (OIC) in patients with advanced cancer, who are receiving palliative care, and particularly explored its early effects. METHODS: Palliative care teams and inpatient palliative care units across 14 institutions in Japan were included in this multicenter, prospective, observational study. Patients who were newly prescribed a daily oral dose of 0.2 mg naldemedine were enrolled. The spontaneous bowel movement (SBM) within 24 h after the first dose of naldemedine was considered the primary outcome, whereas, the secondary outcomes included weekly changes in SBM frequency and adverse events. RESULTS: A total of 204 patients were enrolled and 184 completed the 7-day study. The average age of the participants (103 males, 101 females) was 63 ± 14 years. The primary cancer was detected in the lungs (23.5%), gastrointestinal tract (13.7%), and urological organs (9.3%). A considerable proportion of patients (34.8%) had ECOG performance status of 3-4. Most patients were undergoing active cancer treatment, however, 40.7% of the patients were receiving the best supportive care. Within 24 h of the first naldemedine dose, 146 patients (71.6%, 95% CI: 65.4-77.8%) experienced SBMs. The weekly SBM counts increased in 62.7% of the participants. The major adverse events included diarrhea and abdominal pain, detected in 17.6% and 5.4% of the patients, respectively. However, no serious adverse events were observed. CONCLUSION: Conclusively, naldemedine is effective and safe for OIC treatments in real-world palliative care settings. TRIAL REGISTRATION NUMBER: UMIN000031381, registered 20/02/2018.


Subject(s)
Analgesics, Opioid , Naltrexone , Narcotic Antagonists , Neoplasms , Opioid-Induced Constipation , Palliative Care , Humans , Male , Female , Middle Aged , Prospective Studies , Palliative Care/methods , Aged , Neoplasms/drug therapy , Neoplasms/complications , Opioid-Induced Constipation/drug therapy , Naltrexone/analogs & derivatives , Naltrexone/therapeutic use , Naltrexone/administration & dosage , Naltrexone/adverse effects , Analgesics, Opioid/adverse effects , Analgesics, Opioid/administration & dosage , Analgesics, Opioid/therapeutic use , Narcotic Antagonists/administration & dosage , Narcotic Antagonists/therapeutic use , Narcotic Antagonists/adverse effects , Japan , Adult , Constipation/chemically induced , Constipation/drug therapy , Aged, 80 and over , Cancer Pain/drug therapy , Treatment Outcome
7.
BMJ Case Rep ; 17(5)2024 May 02.
Article in English | MEDLINE | ID: mdl-38697684

ABSTRACT

Acute eosinophilic pneumonia (AEP) is a rare cause of acute respiratory failure. Clinical presentations can range from dyspnoea, fever and cough, to rapidly progressive and potentially fulminant respiratory failure. While its exact cause is often unknown, associations with inhalational injuries and exposures to new medications have been described.We report a case of a middle-aged, non-smoking man with a history of alcohol use disorder. He presented with 4 days of shortness of breath that started hours after taking injectable naltrexone (Vivitrol). The patient had rapidly worsening hypoxaemia, necessitating emergent bronchoscopy with transbronchial biopsies and bronchoalveolar lavage which showed 66% eosinophils. The patient was intubated for the procedure and unable to get extubated due to worsening hypoxaemic respiratory failure with high fractional inspired oxygen requirements. Chest radiograph showed worsening lung infiltrates and with a high index of suspicion for AEP, he was started empirically on methylprednisolone. He had rapid improvement in his respiratory status and was extubated on day 5 of admission then discharged on day 8. Histopathological examination confirmed acute/subacute eosinophilic pneumonia. A 3-week post-discharge follow-up chest radiograph confirmed the full resolution of pulmonary infiltrates.Naltrexone-induced AEP is rare, with only six other cases reported in the literature. Careful history taking and prompt evaluation for AEP are important given the potential for rapid progression to acute hypoxic respiratory failure and the excellent response to steroid treatment.


Subject(s)
Naltrexone , Pulmonary Eosinophilia , Humans , Male , Pulmonary Eosinophilia/chemically induced , Pulmonary Eosinophilia/diagnosis , Naltrexone/therapeutic use , Naltrexone/adverse effects , Middle Aged , Narcotic Antagonists/therapeutic use , Narcotic Antagonists/adverse effects , Narcotic Antagonists/administration & dosage , Methylprednisolone/therapeutic use , Respiratory Insufficiency/chemically induced , Bronchoscopy , Acute Disease , Dyspnea
8.
Early Hum Dev ; 194: 106051, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38815498

ABSTRACT

BACKGROUND: Naltrexone is a medication used to treat both opioid and alcohol use disorder with limited experience in pregnant individuals, particularly in comparison to more commonly utilized treatments such as buprenorphine-naloxone. The long-term outcomes of infants exposed to naltrexone has not been previously examined. AIMS: To compare the neurobehavioral outcomes of naltrexone versus buprenorphine-naloxone exposed infants. STUDY DESIGN: Multi-centered prospective cohort study. SUBJECTS: Pregnant people on prescribed buprenorphine-naloxone or naltrexone were enrolled during pregnancy and the dyad followed until 12 months after delivery. OUTCOME MEASURES: Infants were evaluated at 4-6 weeks corrected gestational age (CGA) using the NICU Neonatal Neurobehavioral Scale (NNNS) and at the 12-month CGA visit using the Ages and Stages Questionnaire, Third Edition (ASQ-3). RESULTS: There were 7 dyads in the naltrexone group and 34 in the buprenorphine-naloxone group. On the NNNS, infants exposed to naltrexone had higher median scores for arousal and excitability, and lower median scores for attention and regulation at 4-6 weeks CGA compared to the buprenorphine-naloxone group. None of the infants in the naltrexone group were monitored for NOWS and had shorter length of hospital stay compared with the buprenorphine-naloxone group. Although no statistically significant differences were observed, more infants in the buprenorphine-naloxone group were identified as at risk for development delays in the communication, problem solving, and personal social domains of the ASQ-3 at 12 months CGA. Results should be interpreted with caution given this study's small sample size and lack of a prospective comparison cohort. CONCLUSIONS: In this small cohort, there are differences noted in infant neurobehavior by NNNS at 4-6 weeks of age when comparing the buprenorphine-naloxone and naltrexone groups. At 12 months, ASQ-3 scores were similar but with percentage differences in potential development delay risk observed between the two groups. Larger cohort studies are needed to determine the long-term child outcomes after naltrexone exposure in pregnancy.


Subject(s)
Naltrexone , Narcotic Antagonists , Humans , Female , Pregnancy , Infant, Newborn , Adult , Naltrexone/adverse effects , Naltrexone/administration & dosage , Naltrexone/therapeutic use , Narcotic Antagonists/adverse effects , Narcotic Antagonists/administration & dosage , Narcotic Antagonists/therapeutic use , Buprenorphine/adverse effects , Buprenorphine/administration & dosage , Prenatal Exposure Delayed Effects/chemically induced , Male , Buprenorphine, Naloxone Drug Combination/adverse effects , Buprenorphine, Naloxone Drug Combination/therapeutic use , Buprenorphine, Naloxone Drug Combination/administration & dosage , Child Development/drug effects , Infant , Infant Behavior/drug effects , Prospective Studies , Opioid-Related Disorders/drug therapy , Naloxone/administration & dosage , Naloxone/adverse effects , Naloxone/therapeutic use , Pregnancy Complications/drug therapy
9.
Am J Emerg Med ; 81: 127-128, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38723364

ABSTRACT

Extended-release formulations of buprenorphine offer less frequent dosing, provide consistent medication delivery, and improve adherence for treatment of opioid use disorder (OUD). Although buprenorphine is a partial agonist with seemingly less precipitated withdrawal and easier initiation than full opioid agonists used for OUD, its use is not benign and understanding of the different extended-release formulations is necessary. We report a case of a patient that received a long-acting buprenorphine formulation (Sublocade®) administered subcutaneously that presented to the emergency department with tachycardia, hyperglycemia, elevated anion gap, and sustained nausea and vomiting refractory to pharmacotherapy requiring surgical removal of the buprenorphine depot for resolution of nausea and vomiting symptoms.


Subject(s)
Buprenorphine , Delayed-Action Preparations , Opioid-Related Disorders , Humans , Buprenorphine/administration & dosage , Buprenorphine/adverse effects , Opioid-Related Disorders/drug therapy , Male , Analgesics, Opioid/adverse effects , Analgesics, Opioid/administration & dosage , Narcotic Antagonists/administration & dosage , Narcotic Antagonists/therapeutic use , Narcotic Antagonists/adverse effects , Adult , Female , Vomiting/chemically induced , Vomiting/drug therapy , Nausea/chemically induced , Nausea/drug therapy
10.
Trials ; 25(1): 286, 2024 Apr 27.
Article in English | MEDLINE | ID: mdl-38678289

ABSTRACT

BACKGROUND: The fragility index is a statistical measure of the robustness or "stability" of a statistically significant result. It has been adapted to assess the robustness of statistically significant outcomes from randomized controlled trials. By hypothetically switching some non-responders to responders, for instance, this metric measures how many individuals would need to have responded for a statistically significant finding to become non-statistically significant. The purpose of this study is to assess the fragility index of randomized controlled trials evaluating opioid substitution and antagonist therapies for opioid use disorder. This will provide an indication as to the robustness of trials in the field and the confidence that should be placed in the trials' outcomes, potentially identifying ways to improve clinical research in the field. This is especially important as opioid use disorder has become a global epidemic, and the incidence of opioid related fatalities have climbed 500% in the past two decades. METHODS: Six databases were searched from inception to September 25, 2021, for randomized controlled trials evaluating opioid substitution and antagonist therapies for opioid use disorder, and meeting the necessary requirements for fragility index calculation. Specifically, we included all parallel arm or two-by-two factorial design RCTs that assessed the effectiveness of any opioid substitution and antagonist therapies using a binary primary outcome and reported a statistically significant result. The fragility index of each study was calculated using methods described by Walsh and colleagues. The risk of bias of included studies was assessed using the Revised Cochrane Risk of Bias tool for randomized trials. RESULTS: Ten studies with a median sample size of 82.5 (interquartile range (IQR) 58, 179, range 52-226) were eligible for inclusion. Overall risk of bias was deemed to be low in seven studies, have some concerns in two studies, and be high in one study. The median fragility index was 7.5 (IQR 4, 12, range 1-26). CONCLUSIONS: Our results suggest that approximately eight participants are needed to overturn the conclusions of the majority of trials in opioid use disorder. Future work should focus on maximizing transparency in reporting of study results, by reporting confidence intervals, fragility indexes, and emphasizing the clinical relevance of findings. TRIAL REGISTRATION: PROSPERO CRD42013006507. Registered on November 25, 2013.


Subject(s)
Narcotic Antagonists , Opiate Substitution Treatment , Opioid-Related Disorders , Randomized Controlled Trials as Topic , Humans , Analgesics, Opioid/therapeutic use , Analgesics, Opioid/adverse effects , Data Interpretation, Statistical , Narcotic Antagonists/therapeutic use , Narcotic Antagonists/adverse effects , Opiate Substitution Treatment/methods , Opioid-Related Disorders/drug therapy , Research Design , Treatment Outcome
11.
J Addict Med ; 18(2): 212-214, 2024.
Article in English | MEDLINE | ID: mdl-38345228

ABSTRACT

ABSTRACT: Monthly long-acting injectable buprenorphine (LAI-BUP) is a treatment option for moderate to severe opioid use disorder. Safe administration of LAI-BUP requires preexisting opioid tolerance to prevent sedation and respiratory depression. In the event of adverse medication effects including oversedation, LAI-BUP can be surgically excised up to 14 days after administration ( https://www.sublocadehcp.com/dosing-administration ). However, the manufacturer does not provide guidance on the proper procedure for excision, and no case reports have been published documenting this procedure. We report a case of a man with methamphetamine use disorder and multiple unintentional fentanyl overdoses who inadvertently received LAI-BUP for overdose protection. This resulted in significant sedation for days, ultimately necessitating excision 5 days after administration. His sedation improved moderately at 24 hours after excision and significantly by 36 hours after excision. Providers seeking to use LAI-BUP to prevent overdose among those with unintentional opioid exposure must ensure sublingual buprenorphine tolerance before injection to avoid iatrogenic harm. Although manufacturer instructions mention that LAI-BUP can be excised under local anesthesia within 14 days of insertion, ideal excision is best performed in a setting with surgical instruments and cautery-such as the operating room-as the depot can adhere strongly to the surrounding subcutaneous tissue.


Subject(s)
Buprenorphine , Opioid-Related Disorders , Humans , Male , Administration, Sublingual , Analgesics, Opioid/adverse effects , Buprenorphine/adverse effects , Drug Tolerance , Narcotic Antagonists/adverse effects , Opioid-Related Disorders/drug therapy
13.
Clin Pharmacol Ther ; 115(3): 506-514, 2024 03.
Article in English | MEDLINE | ID: mdl-38009933

ABSTRACT

Methadone and buprenorphine/naloxone are opioid agonist therapies for opioid use disorder treatment. Genetic factors contribute to individual differences in opioid response; however, little is known regarding genetic associations with clinical outcomes in people receiving opioid agonist therapies. Participants diagnosed with opioid use disorder, principally consisting of prescription opioids (licit or illicit), were randomized to methadone or buprenorphine/naloxone for 24 weeks of daily treatment (NCT03033732). Urine was collected at 12 biweekly study visits and analyzed for non-treatment opioids. Variants in genes involved in methadone metabolism (CYP2B6, CYP2C19, and CYP3A4), buprenorphine metabolism (CYP3A4 and UGT2B7), and µ-opioid receptor function (OPRM1) were genotyped and analyzed for their association with the number of non-treatment opioid-free urine screens. Primary analyses focused on the last 12 weeks (6 study visits, post-titration) of treatment among those reporting White ethnicity. Additional sensitivity and exploratory analyses were performed. Among methadone-treated participants (n = 52), the OPRM1 rs1799971 AA genotype (vs. G-genotypes, i.e., having one or two G alleles) was associated with greater opioid-free urine screens (incidence rate ratio = 5.24, 95% confidence interval (CI) = 2.43-11.26, P = 0.000023); longitudinal analyses showed a significant genotype-by-time interaction over the full 24 weeks (12 study visits, ß = -0.28, 95% CI = -0.45 to -0.11, P = 0.0015). Exploratory analyses suggest an OPRM1 rs1799971 genotype effect on retention. No evidence of association was found between other genetic variants, including in metabolic variants, and non-treatment opioid-free urine screens in the methadone or buprenorphine/naloxone arms. Those with the OPRM1 rs1799971 G-genotypes may have a poorer response to methadone maintenance treatment, an effect that persisted through 24 weeks of treatment.


Subject(s)
Buprenorphine , Opioid-Related Disorders , Humans , Methadone/therapeutic use , Analgesics, Opioid/adverse effects , Pharmacogenetics , Cytochrome P-450 CYP3A , Narcotic Antagonists/adverse effects , Buprenorphine, Naloxone Drug Combination/therapeutic use , Buprenorphine/therapeutic use , Opioid-Related Disorders/drug therapy , Opioid-Related Disorders/genetics , Opiate Substitution Treatment
14.
J Oncol Pharm Pract ; 30(1): 4-8, 2024 Jan.
Article in English | MEDLINE | ID: mdl-36946143

ABSTRACT

INTRODUCTION: Peripherally acting µ-opioid receptor antagonists (PAMORAs) are used in the treatment of opioid induced constipation without impacting the actions of opioid analgesics. Subcutaneous methylnaltrexone was one of the first PAMORAs approved in April 2008 for the treatment of opioid induced constipation in adult patients. The safety and effectiveness of methylnaltrexone has not been established in pediatric patients. In this study, the use of subcutaneous methylnaltrexone in pediatric patients is analyzed and reviewed. The primary outcome is occurrence of a bowel movement within 24 h after methylnaltrexone (MNTX) administration and the number of bowel movements following treatment with methylnaltrexone. Secondary outcomes include safety in this patient cohort. METHODS: This is a retrospective study of 79 pediatric patients with opioid induced constipation. Patients were administered methylnaltrexone during their inpatient stay. Data on bowel activity after methylnaltrexone was obtained from the hospital information system. RESULTS: Out of the 79 patients who received methylnaltrexone, there were seven patients from whom data could not be analyzed. Of the 72 patients whose data was available, 38% (N = 27) were documented as having a bowel movement, 62% (N = 45) did not have a bowel movement. Reported adverse events were minimal with nausea (N = 3), vomiting (N = 1), and flatulence (N = 6). CONCLUSION: Methylnaltrexone appears safe in the pediatric population and produces bowel movements in more than a third of pediatric patients. It is a feasible and safe option for opioid induced constipation in pediatric patients.


Subject(s)
Naltrexone/analogs & derivatives , Neoplasms , Opioid-Induced Constipation , Adult , Humans , Child , Analgesics, Opioid/adverse effects , Opioid-Induced Constipation/drug therapy , Retrospective Studies , Constipation/chemically induced , Constipation/drug therapy , Narcotic Antagonists/adverse effects , Neoplasms/drug therapy , Quaternary Ammonium Compounds
15.
Neurogastroenterol Motil ; 35(12): e14694, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37869768

ABSTRACT

BACKGROUND: Constipation is frequent in critically ill patients, and potentially related to adverse outcomes. Peripherally-active mu-opioid receptor antagonists (PAMORAs) are approved for opioid-induced constipation, but information on their efficacy and safety in critically ill patients is limited. We present a single-center, retrospective, case-series of the use of naldemedine for opioid-associated constipation, and we systematically reviewed the use of PAMORAs in critically ill patients. METHODS: Case-series included consecutive mechanically-ventilated patients; constipation was defined as absence of bowel movements for >3 days. Naldemedine was administered after failure of the local laxation protocol. Systematic review: PubMed was searched for studies of PAMORAs to treat opioid-induced constipation in adult critically ill patients. PRIMARY OUTCOMES: time to laxation, and number of patients laxating at the shortest follow-up. SECONDARY OUTCOMES: gastric residual volumes and adverse events. KEY RESULTS: A total of 13 patients were included in the case-series; the most common diagnosis was COVID-19 ARDS. Patients had their first bowel movement 1 [0;2] day after naldemedine. Daily gastric residual volume was 725 [405;1805] before vs. 250 [45;1090] mL after naldemedine, p = 0.0078. Systematic review identified nine studies (two RCTs, one prospective case-series, three retrospective case-series and three case-reports). Outcomes were similar between groups, with a trend toward a lower gastric residual volume in PAMORAs group. CONCLUSIONS & INFERENCES: In a highly-selected case-series of patients with refractory, opioid-associated constipation, naldemedine was safe and associated to reduced gastric residuals and promoting laxation. In the systematic review and meta-analysis, the use of PAMORAs (mainly methylnaltrexone) was safe and associated with a reduced intolerance to enteral feeding but no difference in the time to laxation.


Subject(s)
Narcotic Antagonists , Opioid-Induced Constipation , Adult , Humans , Narcotic Antagonists/adverse effects , Analgesics, Opioid/adverse effects , Constipation/chemically induced , Constipation/drug therapy , Retrospective Studies , Critical Illness , Naltrexone/therapeutic use , Naltrexone/pharmacology , Laxatives/therapeutic use
16.
Expert Opin Drug Metab Toxicol ; 19(10): 671-679, 2023.
Article in English | MEDLINE | ID: mdl-37795596

ABSTRACT

INTRODUCTION: Drug-induced respiratory depression is potentially fatal and can be caused by various drugs such as synthetic opioids and tranquilizers. The only class of respiratory depressants that has a specific reversal agent are opioids, such as naloxone. These reversal agents have limited utility in situations of polysubstance ingestion with agents from multiple respiratory depressant classes. Hence, there is an unmet need for drugs that stimulate breathing irrespective of the underlying cause of respiratory depression, i.e. mechanism agnostic respiratory stimulants. AREAS COVERED: In this review, we discuss agnostic respiratory stimulants, tested in humans with promising results, i.e. ampakines, drugs that act at the carotid bodies, N-methyl-D-aspartate receptor antagonist ketamine, and orexin receptor-2-agonist danavorexton, and others that demonstrated positive effects in animals but not yet in humans. EXPERT OPINION: Rapid, effective rescuing of individuals who overdosed on respiratory depressants saves lives. While naloxone is the preferred drug for reversing opioid-induced respiratory depression, its effectiveness is limited in cases involving non-opioids. While several agnostic respiratory stimulants showed promise in humans, further research is needed to optimize dosing, evaluate safety and efficacy in deeper respiratory depression (apnea). Additionally, future studies should combine agnostic stimulants with naloxone, to improve rapid, effective rescue from drug overdoses.


Subject(s)
Drug Overdose , Ketamine , Respiratory Insufficiency , Respiratory System Agents , Animals , Humans , Respiratory System Agents/adverse effects , Analgesics, Opioid/adverse effects , Naloxone/adverse effects , Respiratory Insufficiency/chemically induced , Respiratory Insufficiency/drug therapy , Ketamine/adverse effects , Drug Overdose/drug therapy , Narcotic Antagonists/adverse effects
18.
J Subst Use Addict Treat ; 154: 209155, 2023 11.
Article in English | MEDLINE | ID: mdl-37657559

ABSTRACT

BACKGROUND: Few studies investigate the natural history of patients on long-term treatment for opioid use disorder (OUD). We evaluated the long-term efficacy, safety, and tolerability experience of monthly extended-release buprenorphine (BUP-XR) in participants seeking treatment for OUD, via integrated analysis of phase 3 studies. METHODS: Study 1 was a 24-week randomized, double-blind, placebo-controlled trial of participants receiving monthly injections of BUP-XR (300 mg × 2, 100 mg × 4 [n = 203] or 300 mg × 6 [n = 201]) or placebo (n = 100). Study 2 was a 48-week, open-label trial enrolling 257 participants who completed study 1 and 412 de novo participants, to receive 6 and 12 BUP-XR injections, respectively. Study 3 was a 24-week, open-label extension enrolling 208 participants who completed study 2 for 6 additional injections. We assessed opioid abstinence as the proportion of urine opioid negative participants by visit and the percentage of each participant's negative opioid assessments during the first 6 months. RESULTS: In total, 916 participants were treated with BUP-XR or placebo. By the end of 18 months, 92.7 % of the de novo cohort and 81.8 % of the study 1 cohort were urine negative for opioids. Among early nonresponders (percentage of abstinence ≤20 %), 73.1 % were urine negative after 18 months. The longer treatment period was well tolerated, with no new safety concerns, and a low incidence of opioid withdrawal signs and symptoms, and hepatic disorder. CONCLUSIONS: Extending BUP-XR treatment beyond 6 months sustained improvement in opioid abstinence and was well tolerated, supporting clinical benefit up to 18 months. TRIAL REGISTRATION: NCT02357901 (study 1); NCT02510014 (study 2); NCT02896296 (study 3).


Subject(s)
Buprenorphine , Opioid-Related Disorders , Humans , Buprenorphine/adverse effects , Analgesics, Opioid/adverse effects , Narcotic Antagonists/adverse effects , Naltrexone , Opioid-Related Disorders/drug therapy , Injections, Subcutaneous
19.
Am J Addict ; 32(5): 469-478, 2023 09.
Article in English | MEDLINE | ID: mdl-37308805

ABSTRACT

BACKGROUND AND OBJECTIVES: Buprenorphine/naloxone (BUP-NX) and methadone are used to treat opioid use disorder (OUD), yet there is insufficient evidence on the impact of doses on interventions' effectiveness and safety when treating OUD attributable to other opioids than heroin. METHODS: We explored associations between methadone and BUP-NX doses and treatment outcomes using data from OPTIMA, a 24-week, pragmatic, open-label, multicenter, pan-Canadian, randomized controlled, two-arm parallel trial with participants (N = 272) with OUD who primarily use opioids other than heroin. Participants were randomized to receive flexible take-home BUP-NX (n = 138) or standard supervised methadone treatment (n = 134). We examined associations between highest BUP-NX and methadone doses, and (1) percentage of opioid-positive urine drug screens (UDS); (2) retention in the assigned treatment; and (3) adverse events (AEs). RESULTS: The mean (SD) highest BUP-NX and methadone dose were 17.31 mg/day (8.59) and 67.70 mg/day (34.70). BUP-NX and methadone doses were not associated with opioid-positive UDS percentages or AEs. Methadone dose was associated with higher retention in treatment (odds ratio [OR]: 1.025; 95% confidence interval [CI]: 1.010; 1.041), while BUP-NX dose was not (OR: 1.055; 95% CI: 0.990; 1.124). Higher methadone doses (70-110 mg/day) offered higher odds of treatment retention. DISCUSSION AND CONCLUSION: Methadone dose was associated with higher retention, which may be related to its full µ-opioid receptor agonism. Future research should notably ascertain the effect of pace of titration on a wide range of outcomes. SCIENTIFIC SIGNIFICANCE: Our results extend previous findings of high doses of methadone increasing retention to be applied in our population using opioids other than heroin, including highly potent opioids.


Subject(s)
Buprenorphine , Opioid-Related Disorders , Humans , Buprenorphine, Naloxone Drug Combination/therapeutic use , Methadone/adverse effects , Analgesics, Opioid/adverse effects , Heroin , Opiate Substitution Treatment/adverse effects , Opiate Substitution Treatment/methods , Canada , Opioid-Related Disorders/drug therapy , Buprenorphine/adverse effects , Prescriptions , Narcotic Antagonists/adverse effects
20.
Drug Dev Res ; 84(6): 1279-1284, 2023 09.
Article in English | MEDLINE | ID: mdl-37317059

ABSTRACT

This study was conducted to determine the safety of topical naltrexone treatment in Göttingen swine. Efficacy of topical naltrexone was performed previously in Sprague-Dawley rats. In this study, 25 male and female mini-pigs received topical naltrexone once daily for 30 days. The gel at doses of 1%, 2%, and 10% naltrexone was applied at a dose volume of 0.01 ml/cm2 to an area of unbroken skin encompassing 10% of the animal's surface. Body and food consumption, skin and organ morphology, and clinical signs, including blood analyses were taken periodically. Naltrexone levels in serum were measured at the time of death. No adverse observations were made in the cutaneous skin, autopsied organs, or biochemical parameters. The no-observed adverse effect level (NOAEL) was considered to be 2% topical application daily. The conclusions from the veterinarians and researchers are that topical naltrexone at 1% or 2% can be used safely in clinical efficacy studies.


Subject(s)
Diabetes Mellitus, Experimental , Naltrexone , Rats , Male , Animals , Female , Swine , Narcotic Antagonists/adverse effects , Swine, Miniature , Rats, Sprague-Dawley , Diabetes Mellitus, Experimental/drug therapy
SELECTION OF CITATIONS
SEARCH DETAIL