Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
Nat Commun ; 14(1): 7700, 2023 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-38052779

RESUMEN

The opioid crisis in the United States is primarily driven by the highly potent synthetic opioid fentanyl leading to >70,000 overdose deaths annually; thus, new therapies for fentanyl overdose are urgently needed. Here, we present the first clinic-ready, fully human monoclonal antibody CSX-1004 with picomolar affinity for fentanyl and related analogs. In mice CSX-1004 reverses fentanyl antinociception and the intractable respiratory depression caused by the ultrapotent opioid carfentanil. Moreover, toxicokinetic evaluation in a repeat-dose rat study and human tissue cross-reactivity study reveals a favorable pharmacokinetic profile of CSX-1004 with no safety-related issues. Using a highly translational non-human primate (NHP) model of respiratory depression, we demonstrate CSX-1004-mediated protection from repeated fentanyl challenges for 3-4 weeks. Furthermore, treatment with CSX-1004 produces up to a 15-fold potency reduction of fentanyl in NHP respiration, antinociception and operant responding assays without affecting non-fentanyl opioids like oxycodone. Taken together, our data establish the feasibility of CSX-1004 as a promising candidate medication for preventing and reversing fentanyl-induced overdose.


Asunto(s)
Sobredosis de Droga , Insuficiencia Respiratoria , Humanos , Ratas , Ratones , Animales , Estados Unidos , Analgésicos Opioides/farmacocinética , Anticuerpos Monoclonales , Fentanilo , Sobredosis de Droga/tratamiento farmacológico , Insuficiencia Respiratoria/inducido químicamente , Insuficiencia Respiratoria/tratamiento farmacológico
2.
ACS Chem Biol ; 16(2): 277-282, 2021 02 19.
Artículo en Inglés | MEDLINE | ID: mdl-33533592

RESUMEN

The opioid epidemic remains a dire public health crisis with millions of people currently suffering from opioid use disorder (OUD) and tens of thousands dying each year. Synthetic opioids are most responsible for the crisis because of their extreme potency and ease of manufacture. Carfentanil for example has an estimated potency 10,000 times greater than morphine and thus is highly dangerous for human use. Herein, we report two synthetic opioid vaccines that elicited high-affinity antibodies against carfentanil and fentanyl with cross-reactivity to other synthetic opioids in mice and offered protection against opioid-induced respiratory depression, the primary cause of overdose deaths. These vaccines also successfully diminished drug biodistribution to the brain and shielded against opioid analgesic effects. Collectively, these findings provide new insights into the development of immunotherapeutic strategies aimed at opioid abuse and overdose.


Asunto(s)
Fentanilo/análogos & derivados , Trastornos Relacionados con Opioides/terapia , Insuficiencia Respiratoria/terapia , Vacunas Sintéticas/uso terapéutico , Animales , Fentanilo/inmunología , Fentanilo/farmacocinética , Fentanilo/uso terapéutico , Inmunoconjugados/inmunología , Inmunoconjugados/farmacocinética , Inmunoconjugados/uso terapéutico , Ratones , Insuficiencia Respiratoria/inducido químicamente , Toxoide Tetánico/inmunología , Vacunas Sintéticas/inmunología
3.
Drug Alcohol Depend ; 218: 108348, 2021 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-33268227

RESUMEN

AIM: Opioid-targeted vaccines are under consideration as candidate Opioid Use Disorder medications. We recently reported that a fentanyl-targeted vaccine produced a robust and long-lasting attenuation of fentanyl-vs-food choice in rats. In the current study, we evaluated an optimized fentanyl-targeted vaccine in rhesus monkeys to determine whether vaccine effectiveness to attenuate fentanyl choice translated to a species with greater phylogenetic similarity to humans. METHODS: Adult male (2) and female (3) rhesus monkeys were trained to respond under a concurrent schedule of food (1 g pellets) and intravenous fentanyl (0, 0.032-1 µg/kg/injection) reinforcement during daily 2 h sessions. Fentanyl choice dose-effect functions were determined daily and 7-day buprenorphine treatments (0.0032-0.032 mg/kg/h IV; n = 4-5) were determined for comparison to vaccine effects. Subsequently, a fentanyl-CRM197 conjugate vaccine was administered at week 0, 3, 8, 15 over a 29-week experimental period during which fentanyl choice dose-effect functions continued to be determined daily. RESULTS: Buprenorphine significantly decreased fentanyl choice and reciprocally increased food choice. Vaccination eliminated fentanyl choice and increased food choice in four-of-the-five monkeys. A transient and less robust vaccine effect was observed in the fifth monkey. Fentanyl-specific antibody concentrations peaked after the third vaccination to approximately 50 µg/mL while anti-fentanyl antibody affinity increased to a sustained low nanomolar level. CONCLUSION: These results translate fentanyl vaccine effectiveness from rats to rhesus monkeys to decrease fentanyl-vs-food choice, albeit with greater individual differences observed in monkeys. These results support the potential and further clinical evaluation of this fentanyl-targeted vaccine as a candidate Opioid Use Disorder medication.


Asunto(s)
Analgésicos Opioides/farmacología , Conducta Alimentaria/efectos de los fármacos , Fentanilo/farmacología , Vacunas , Animales , Conducta de Elección/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Femenino , Alimentos , Macaca mulatta , Masculino , Trastornos Relacionados con Opioides , Filogenia , Ratas , Refuerzo en Psicología , Autoadministración
4.
Mol Pharm ; 18(1): 228-235, 2021 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-33301675

RESUMEN

The nearly insurmountable adversity that accompanies opioid use disorder (OUD) creates life-altering complications for opioid users. To worsen matters, existing small-molecule drugs continue to inadequately address OUD due to their engagement of the opioid receptor, which can leave the user to deal with side effects and financial hardships from their repeated use. An alternative therapeutic approach utilizes endogenously generated antibodies through active vaccination to reduce the effect of opioids without modulating the opioid receptor. Here, we explore different adjuvants and storage conditions to improve opioid vaccine efficacy and shelf life. Our results revealed that inulin-based formulations (Advax) containing a CpG oligodeoxynucleotide (ODN) acted as effective adjuvants when combined with a heroin conjugate: immunized mice showed excellent recovery from heroin-induced antinociception accompanied by high titer, high opioid affinity serum antibodies similar to the immunopotentiating properties of traditional alum-based adjuvants. Moreover, nonhuman primates vaccinated with a heroin/fentanyl combination vaccine demonstrated potent antibody responses against opioids when formulated with both inulin and alum adjuvants. Finally, storing a freeze-dried opioid vaccine formulation maintained efficacy for up 1 year at room temperature. The results from our studies represent an advance toward a clinically feasible opioid vaccine.


Asunto(s)
Adyuvantes Inmunológicos/farmacología , Adyuvantes Farmacéuticos/farmacología , Fentanilo/inmunología , Heroína/inmunología , Vacunas Conjugadas/inmunología , Analgésicos Opioides/inmunología , Animales , Inmunización/métodos , Masculino , Ratones , Ratones Endogámicos BALB C , Oligodesoxirribonucleótidos/inmunología , Trastornos Relacionados con Opioides/inmunología , Vacunación/métodos
5.
Bioorg Med Chem Lett ; 30(17): 127388, 2020 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-32738981

RESUMEN

Heroin overdose and addiction remain significant health and economic burdens in the world today costing billions of dollars annually. Moreover, only limited pharmacotherapeutic options are available for treatment of heroin addiction. In our efforts to combat the public health threat posed by heroin addiction, we have developed vaccines against heroin. To expand upon our existing heroin-vaccine arsenal, we synthesized new aryl and alkyl sulfonate ester haptens; namely aryl-mono-sulfonate (HMsAc) and Aryl/alkyl-di-sulfonate (H(Ds)2) as carboxyl-isosteres of heroin then compared them to our model heroin-hapten (HAc) through vaccination studies. Heroin haptens were conjugated to the carrier protein CRM197 and the resulting CRM-immunoconjugates were used to vaccinate Swiss Webster mice following an established immunization protocol. Binding studies revealed that the highest affinity anti-heroin antibodies were generated by the HMsAc vaccine followed by the HAc and H(Ds)2 vaccines, respectively (HMsAc > HAc≫HDs2). However, neither the HMsAc nor H(Ds)2 vaccines were able to generate high affinity antibodies to the psychoactive metabolite 6-acetyl morphine (6-AM), in comparison to the HAc vaccine. Blood brain bio-distribution studies supported these binding results with vaccine efficiency following the trend HAc > HMsAc â‰« H(Ds)2 The work described herein provides insight into the use of hapten-isosteric replacement in vaccine drug design.


Asunto(s)
Alcanosulfonatos/química , Diseño de Fármacos , Haptenos/química , Heroína/química , Vacunas Sintéticas/inmunología , Animales , Anticuerpos/inmunología , Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Encéfalo/metabolismo , Haptenos/inmunología , Heroína/inmunología , Ratones , Vacunas Sintéticas/sangre , Vacunas Sintéticas/metabolismo
6.
J Am Chem Soc ; 142(31): 13294-13298, 2020 08 05.
Artículo en Inglés | MEDLINE | ID: mdl-32700530

RESUMEN

The United States is in the midst of an unprecedented epidemic of opioid substance use disorder, and while pharmacotherapies including opioid agonists and antagonists have shown success, they can be inadequate and frequently result in high recidivism. With these challenges facing opioid use disorder treatments immunopharmacotherapy is being explored as an alternative therapy option and is based upon antibody-opioid sequestering to block brain entry. Development of a heroin vaccine has become a major research focal point; however, producing an efficient vaccine against heroin has been particularly challenging because of the need to generate not only a potent immune response but one against heroin and its multiple psychoactive molecules. In this study, we explored the consequence of regioselective deuteration of a heroin hapten and its impact upon the immune response against heroin and its psychoactive metabolites. Deuterium (HdAc) and cognate protium heroin (HAc) haptens were compared head to head in an inclusive vaccine study. Strikingly the HdAc vaccine granted greater efficacy in blunting heroin analgesia in murine behavioral models compared to the HAc vaccine. Binding studies confirmed that the HdAc vaccine elicited both greater quantities and equivalent or higher affinity antibodies toward heroin and 6-AM. Blood-brain biodistribution experiments corroborated these affinity tests. These findings suggest that regioselective hapten deuteration could be useful for the resurrection of previous drug of abuse vaccines that have met limited success in the past.


Asunto(s)
Haptenos/química , Haptenos/inmunología , Heroína/inmunología , Vacunas/química , Vacunas/inmunología , Deuterio/química , Heroína/química , Conformación Molecular
7.
ACS Chem Neurosci ; 11(9): 1300-1310, 2020 05 06.
Artículo en Inglés | MEDLINE | ID: mdl-32271538

RESUMEN

Opioid-targeted vaccines represent an emerging treatment strategy for opioid use disorder. To determine whether concurrent vaccination against two commonly abused opioids (fentanyl and heroin) would confer broader spectrum opioid coverage, the current study evaluated dual fentanyl/heroin conjugate vaccine effectiveness using a warm water tail-withdrawal and a fentanyl/heroin-vs-food choice procedure in male and female rats across a 105-day observation period. Vaccine administration generated titers of high-affinity antibodies to both fentanyl and heroin sufficient to decrease the antinociceptive potency of fentanyl (25-fold), heroin (4.6-fold), and a 1:27 fentanyl/heroin mixture (7.5-fold). Vaccination did not alter the antinociceptive potency of the structurally dissimilar opioid agonist methadone. For comparison, continuous treatment with a naltrexone dose (0.032 mg/kg/h) shown previously to produce clinically relevant plasma-naltrexone levels decreased the antinociceptive potency of fentanyl, heroin, and the 1:27 fentanyl/heroin mixture by approximately 20-fold. Naltrexone treatment also shifted the potency of 1:27 fentanyl/heroin mixture in a drug-vs-food choice self-administration procedure 4.3-fold. In contrast, vaccination did not attenuate 1:27 fentanyl/heroin mixture self-administration in the drug-vs-food choice procedure. These data demonstrate that a vaccine can simultaneously attenuate the thermal antinociceptive effects of two structurally dissimilar opioids. However, the vaccine did not attenuate fentanyl/heroin mixture self-administration, suggesting a greater magnitude of vaccine responsiveness is required to decrease opioid reinforcement relative to antinociception.


Asunto(s)
Fentanilo , Vacunas , Analgésicos Opioides , Animales , Relación Dosis-Respuesta a Droga , Femenino , Heroína , Masculino , Naltrexona/farmacología , Ratas
8.
Drug Alcohol Depend ; 204: 107501, 2019 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-31479865

RESUMEN

BACKGROUND: One emerging strategy to address the opioid crisis includes opioid-targeted immunopharmacotherapies. This study compared effectiveness of a heroin-tetanus toxoid (TT) conjugate vaccine to antagonize heroin, 6-acetylmorphine (6-AM), morphine, and fentanyl antinociception in rats. METHODS: Adult male and female Sprague Dawley rats received three doses of active or control vaccine at weeks 0, 2, and 4. Vaccine pharmacological selectivity was assessed by comparing opioid dose-effect curves in 50 °C warm-water tail-withdrawal procedure before and after active or control heroin-TT vaccine. Route of heroin administration [subcutaneous (SC) vs. intravenous [IV)] was also examined as a determinant of vaccine effectiveness. Continuous naltrexone treatment (0.0032-0.032 mg/kg/h) effects on heroin, 6-AM, and morphine antinociceptive potency were also determined as a benchmark for minimal vaccine effectiveness. RESULTS: The heroin-TT vaccine decreased potency of SC heroin (5-fold), IV heroin (3-fold), and IV 6-AM (3-fold) for several weeks without affecting IV morphine or SC and IV fentanyl potency. The control vaccine did not alter potency of any opioid. Naltrexone dose-dependently decreased antinociceptive potency of SC heroin, and treatment with 0.01 mg/kg/h naltrexone produced similar, approximate 8-fold decreases in potencies of SC and IV heroin, IV 6-AM, and IV morphine. The combination of naltrexone and active vaccine was more effective than naltrexone alone to antagonize SC heroin but not IV heroin. CONCLUSIONS: The heroin-TT vaccine formulation examined is less effective, but more selective, than chronic naltrexone to attenuate heroin antinociception in rats. Furthermore, these results provide an empirical framework for future preclinical opioid vaccine research to benchmark effectiveness against naltrexone.


Asunto(s)
Fentanilo/antagonistas & inhibidores , Heroína/antagonistas & inhibidores , Heroína/farmacología , Derivados de la Morfina/antagonistas & inhibidores , Morfina/antagonistas & inhibidores , Naltrexona/farmacología , Vacunación , Analgésicos Opioides/antagonistas & inhibidores , Analgésicos Opioides/farmacología , Animales , Relación Dosis-Respuesta a Droga , Vías de Administración de Medicamentos , Femenino , Fentanilo/farmacología , Masculino , Morfina/farmacología , Derivados de la Morfina/farmacología , Antagonistas de Narcóticos/farmacología , Ratas , Toxoide Tetánico/farmacología
9.
Neuropharmacology ; 158: 107730, 2019 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-31369740

RESUMEN

One proposed factor contributing to the increased frequency of opioid overdose deaths is the emergence of novel synthetic opioids, including illicit fentanyl and fentanyl analogues. A treatment strategy currently under development to address the ongoing opioid crisis is immunopharmacotherapies or opioid-targeted vaccines. The present study determined the effectiveness and selectivity of a fentanyl-tetanus toxoid conjugate vaccine to alter the behavioral effects of fentanyl and a structurally dissimilar mu-opioid agonist oxycodone in male rhesus monkeys (n = 3-4). Fentanyl and oxycodone produced dose-dependent suppression of behavior in an assay of schedule-controlled responding and antinociception in an assay of thermal nociception (50 °C). Acute naltrexone (0.032 mg/kg) produced an approximate 10-fold potency shift for fentanyl to decrease operant responding. The fentanyl vaccine was administered at weeks 0, 2, 4, 9, 19, and 44 and fentanyl or oxycodone potencies in both behavioral assays were redetermined over the course of 49 weeks. The vaccine significantly and selectively shifted fentanyl potency at least 10-fold in both assays at several time points over the entire experimental period. Mid-point titer levels correlated with fentanyl antinociceptive potency shifts. Antibody affinity for fentanyl as measured by a competitive binding assay improved over time to approximately 3-4 nM. The fentanyl vaccine also increased fentanyl plasma levels approximately 6-fold consistent with the hypothesis that the vaccine sequesters fentanyl in the blood. Overall, these results support the continued development and evaluation of this fentanyl vaccine in humans to address the ongoing opioid crisis.


Asunto(s)
Analgésicos Opioides/inmunología , Fentanilo/inmunología , Oxicodona/farmacología , Toxoide Tetánico/inmunología , Vacunas Conjugadas/inmunología , Analgésicos Opioides/farmacología , Animales , Conducta Animal/efectos de los fármacos , Condicionamiento Operante/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Fentanilo/farmacología , Macaca mulatta , Masculino , Naltrexona/farmacología , Antagonistas de Narcóticos/farmacología , Nocicepción/efectos de los fármacos , Receptores Opioides mu , Toxoide Tetánico/farmacología , Vacunas/inmunología , Vacunas/farmacología , Vacunas Conjugadas/farmacología
10.
J Am Chem Soc ; 141(26): 10489-10503, 2019 07 03.
Artículo en Inglés | MEDLINE | ID: mdl-31187995

RESUMEN

Opioid abuse in the United States has been declared a national crisis and is exacerbated by an inexpensive, readily available, and illicit supply of synthetic opioids. Specifically, fentanyl and related analogues such as carfentanil pose a significant danger to opioid users due to their high potency and rapid acting depression of respiration. In recent years these synthetic opioids have become the number one cause of drug-related deaths. In our research efforts to combat the public health threat posed by synthetic opioids, we have developed monoclonal antibodies (mAbs) against the fentanyl class of drugs. The mAbs were generated in hybridomas derived from mice vaccinated with a fentanyl conjugate vaccine. Guided by a surface plasmon resonance (SPR) binding assay, we selected six hybridomas that produced mAbs with 10-11 M binding affinity for fentanyl, yet broad cross-reactivity with related fentanyl analogues. In mouse antinociception models, our lead mAb (6A4) could blunt the effects of both fentanyl and carfentanil in a dose-responsive manner. Additionally, mice pretreated with 6A4 displayed enhanced survival when subjected to fentanyl above LD50 doses. Pharmacokinetic analysis revealed that the antibody sequesters large amounts of these drugs in the blood, thus reducing drug biodistribution to the brain and other tissue. Lastly, the 6A4 mAb could effectively reverse fentanyl/carfentanil-induced antinociception comparable to the opioid antagonist naloxone, the standard of care drug for treating opioid overdose. While naloxone is known for its short half-life, we found the half-life of 6A4 to be approximately 6 days in mice, thus monoclonal antibodies could theoretically be useful in preventing renarcotization events in which opioid intoxication recurs following quick metabolism of naloxone. Our results as a whole demonstrate that monoclonal antibodies could be a desirable treatment modality for synthetic opioid overdose and possibly opioid use disorder.


Asunto(s)
Analgésicos Opioides/antagonistas & inhibidores , Anticuerpos Monoclonales/farmacología , Antagonistas de Narcóticos/farmacología , Dolor Nociceptivo/tratamiento farmacológico , Trastornos Relacionados con Opioides/tratamiento farmacológico , Analgésicos Opioides/administración & dosificación , Animales , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/química , Ratones , Antagonistas de Narcóticos/administración & dosificación , Antagonistas de Narcóticos/química , Resonancia por Plasmón de Superficie
11.
Neuropsychopharmacology ; 44(10): 1681-1689, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31043682

RESUMEN

The current opioid crisis remains a significant public health issue and there is a critical need for biomedical research to develop effective and easily deployable candidate treatments. One emerging treatment strategy for opioid use disorder includes immunopharmacotherapies or opioid-targeted vaccines. The present study determined the effectiveness of a fentanyl-tetanus toxoid conjugate vaccine to alter fentanyl self-administration using a fentanyl-vs.-food choice procedure in male and female rats under three experimental conditions. For comparison, continuous 7-day naltrexone (0.01-0.1 mg/kg/h) and 7-day clonidine (3.2-10 µg/kg/h) treatment effects were also determined on fentanyl-vs.-food choice. Male and female rats responded for concurrently available 18% diluted Ensure® (liquid food) and fentanyl (0-10 µg/kg/infusion) infusions during daily sessions. Under baseline and saline treatment conditions, fentanyl maintained a dose-dependent increase in fentanyl-vs.-food choice. First, fentanyl vaccine administration significantly blunted fentanyl reinforcement and increased food reinforcement for 15 weeks in non-opioid dependent rats. Second, surmountability experiments by increasing the unit fentanyl dose available during the self-administration session 10-fold empirically determined that the fentanyl vaccine produced an approximate 22-fold potency shift in fentanyl-vs.-food choice that was as effective as the clinically approved treatment naltrexone. Clonidine treatment significantly increased fentanyl-vs.-food choice. Lastly, fentanyl vaccine administration prevented the expression of withdrawal-associated increases in fentanyl-vs.-food choice following introduction of extended 12 h fentanyl access sessions. Overall, these results support the potential and further consideration of immunopharmacotherapies as candidate treatments to address the current opioid crisis.


Asunto(s)
Analgésicos Opioides/administración & dosificación , Analgésicos Opioides/inmunología , Conducta Animal/efectos de los fármacos , Conducta de Elección/efectos de los fármacos , Comportamiento de Búsqueda de Drogas/efectos de los fármacos , Fentanilo/administración & dosificación , Fentanilo/inmunología , Vacunas Conjugadas/farmacología , Agonistas de Receptores Adrenérgicos alfa 2/farmacología , Animales , Clonidina/farmacología , Conducta Alimentaria/efectos de los fármacos , Femenino , Alimentos , Masculino , Naltrexona/farmacología , Antagonistas de Narcóticos/farmacología , Trastornos Relacionados con Opioides , Ratas , Refuerzo en Psicología , Autoadministración , Síndrome de Abstinencia a Sustancias/fisiopatología , Toxoide Tetánico
12.
Mol Pharm ; 15(3): 1062-1072, 2018 03 05.
Artículo en Inglés | MEDLINE | ID: mdl-29420901

RESUMEN

In recent years, drug conjugate vaccines have shown promise as therapeutics for substance use disorder. As a means to improve the efficacy of a heroin conjugate vaccine, we systematically explored 20 vaccine formulations with varying combinations of carrier proteins and adjuvants. In regard to adjuvants, we explored a Toll-like receptor 9 (TLR9) agonist and a TLR3 agonist in the presence of alum. The TLR9 agonist was cytosine-guanine oligodeoxynucleotide 1826 (CpG ODN 1826), while the TLR3 agonist was virus-derived genomic doubled-stranded RNA (dsRNA). The vaccine formulations containing TLR3 or TLR9 agonist alone elicited strong antiheroin antibody titers and blockade of heroin-induced antinociception when formulated with alum; however, a combination of TLR3 and TLR9 adjuvants did not result in improved efficacy. Investigation of month-long stability of the two lead formulations revealed that the TLR9 but not the TLR3 formulation was stable when stored as a lyophilized solid or as a liquid over 30 days. Furthermore, mice immunized with the TLR9 + alum heroin vaccine gained significant protection from lethal heroin doses, suggesting that this vaccine formulation is suitable for mitigating the harmful effects of heroin, even following month-long storage at room temperature.


Asunto(s)
Analgésicos Opioides/farmacología , Sobredosis de Droga/prevención & control , Dependencia de Heroína/complicaciones , Heroína/farmacología , Vacunas Conjugadas/farmacología , Adyuvantes Inmunológicos/química , Adyuvantes Inmunológicos/farmacología , Analgésicos Opioides/química , Animales , Modelos Animales de Enfermedad , Sobredosis de Droga/etiología , Heroína/química , Humanos , Masculino , Ratones , Oligodesoxirribonucleótidos/química , Oligodesoxirribonucleótidos/farmacología , Receptor Toll-Like 3/agonistas , Receptor Toll-Like 9/agonistas , Vacunación/métodos , Vacunas Conjugadas/química
13.
J Am Chem Soc ; 139(25): 8601-8611, 2017 06 28.
Artículo en Inglés | MEDLINE | ID: mdl-28574716

RESUMEN

Heroin is a highly abused opioid and incurs a significant detriment to society worldwide. In an effort to expand the limited pharmacotherapy options for opioid use disorders, a heroin conjugate vaccine was developed through comprehensive evaluation of hapten structure, carrier protein, adjuvant and dosing. Immunization of mice with an optimized heroin-tetanus toxoid (TT) conjugate formulated with adjuvants alum and CpG oligodeoxynucleotide (ODN) generated heroin "immunoantagonism", reducing heroin potency by >15-fold. Moreover, the vaccine effects proved to be durable, persisting for over eight months. The lead vaccine was effective in rhesus monkeys, generating significant and sustained antidrug IgG titers in each subject. Characterization of both mouse and monkey antiheroin antibodies by surface plasmon resonance (SPR) revealed low nanomolar antiserum affinity for the key heroin metabolite, 6-acetylmorphine (6AM), with minimal cross reactivity to clinically used opioids. Following a series of heroin challenges over six months in vaccinated monkeys, drug-sequestering antibodies caused marked attenuation of heroin potency (>4-fold) in a schedule-controlled responding (SCR) behavioral assay. Overall, these preclinical results provide an empirical foundation supporting the further evaluation and potential clinical utility of an effective heroin vaccine in treating opioid use disorders.


Asunto(s)
Adyuvantes Inmunológicos/química , Diseño de Fármacos , Haptenos/química , Heroína , Vacunas Conjugadas , Inmunidad Humoral
14.
Pharmacol Rev ; 69(3): 298-315, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28634286

RESUMEN

Substance use disorder, especially in relation to opioids such as heroin and fentanyl, is a significant public health issue and has intensified in recent years. As a result, substantial interest exists in developing therapeutics to counteract the effects of abused drugs. A promising universal strategy for antagonizing the pharmacology of virtually any drug involves the development of a conjugate vaccine, wherein a hapten structurally similar to the target drug is conjugated to an immunogenic carrier protein. When formulated with adjuvants and immunized, the immunoconjugate should elicit serum IgG antibodies with the ability to sequester the target drug to prevent its entry to the brain, thereby acting as an immunoantagonist. Despite the failures of first-generation conjugate vaccines against cocaine and nicotine in clinical trials, second-generation vaccines have shown dramatically improved performance in preclinical models, thus renewing the potential clinical utility of conjugate vaccines in curbing substance use disorder. This review explores the critical design elements of drug conjugate vaccines such as hapten structure, adjuvant formulation, bioconjugate chemistry, and carrier protein selection. Methods for evaluating these vaccines are discussed, and recent progress in vaccine development for each drug is summarized.


Asunto(s)
Drogas Ilícitas/inmunología , Trastornos Relacionados con Sustancias/terapia , Vacunas Conjugadas/química , Vacunas Conjugadas/inmunología , Animales , Antagonismo de Drogas , Haptenos/química , Haptenos/inmunología , Humanos , Trastornos Relacionados con Sustancias/inmunología , Vacunas Conjugadas/administración & dosificación
15.
J Am Chem Soc ; 139(21): 7264-7272, 2017 05 31.
Artículo en Inglés | MEDLINE | ID: mdl-28475321

RESUMEN

Botulinum neurotoxin serotype A (BoNT/A) causes a debilitating and potentially fatal illness known as botulism. The toxin is also a bioterrorism threat, yet no pharmacological antagonist to counteract its effects has reached clinical approval. Existing strategies to negate BoNT/A intoxication have looked to antibodies, peptides, or organic small molecules as potential therapeutics. In this work, a departure from the traditional drug discovery mindset was pursued, in which the enzyme's susceptibility to metal ions was exploited. A screen of a series of metal salts showed marked inhibitory activity of group 11 and 12 metals against the BoNT/A light chain (LC) protease. Enzyme kinetics revealed that copper (I) and (II) cations displayed noncompetitive inhibition of the LC (Ki ≈ 1 µM), while mercury (II) cations were 10-fold more potent. Crystallographic and mutagenesis studies elucidated a key binding interaction between Cys165 on BoNT/A LC and the inhibitory metals. As potential copper prodrugs, ligand-copper complexes were examined in a cell-based model and were found to prevent BoNT/A cleavage of the endogenous protein substrate, SNAP-25, even at low µM concentrations of complexes. Further investigation of the complexes suggested a bioreductive mechanism causing intracellular release of copper, which directly inhibited the BoNT/A protease. In vivo experiments demonstrated that copper (II) dithiocarbamate and bis(thiosemicarbazone) complexes could delay BoNT/A-mediated lethality in a rodent model, indicating their potential for treating the harmful effects of BoNT/A intoxication. Our studies illustrate that metals can be therapeutically viable enzyme inhibitors; moreover, enzymes that share homology with BoNT LCs may be similarly targeted with metals.


Asunto(s)
Toxinas Botulínicas Tipo A/antagonistas & inhibidores , Complejos de Coordinación/farmacología , Cobre/farmacología , Inhibidores de Proteasas/farmacología , Tiocarbamatos/farmacología , Tiosemicarbazonas/farmacología , Animales , Toxinas Botulínicas Tipo A/metabolismo , Toxinas Botulínicas Tipo A/toxicidad , Complejos de Coordinación/química , Cobre/química , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Humanos , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Cinética , Ratones , Neuronas/citología , Neuronas/efectos de los fármacos , Inhibidores de Proteasas/química , Relación Estructura-Actividad , Tiocarbamatos/química , Tiosemicarbazonas/química
16.
Neuropharmacology ; 116: 1-8, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-27956054

RESUMEN

Recreational use of substituted cathinones continues to be an emerging public health problem in the United States; cathinone derivatives α-pyrrolidinopentiophenone (α-PVP) and 3,4-methylenedioxypyrovalerone (MDPV), which have been linked to human fatalities and show high potential for abuse liability in animal models, are of particular concern. The objective of this study was to develop an immunotherapeutic strategy for attenuating the effects of α-PVP and MDPV in rats, using drug-conjugate vaccines created to generate antibodies with neutralizing capacity. Immunoconjugates (α-PVP-KLH and MDPV-KLH) or the control carrier protein, keyhole limpet hemocyanin (KLH), were administered to groups (N = 12) of male Sprague-Dawley rats on Weeks 0, 2 and 4. Groups were administered α-PVP or MDPV (0.0, 0.25, 0.5, 1.0, 5.0 mg/kg, i.p.) in acute drug challenges and tested for changes in wheel activity. Increased wheel activity produced by α-PVP or MDPV in the controls was attenuated in the α-PVP-KLH and MDPV-KLH vaccinated groups, respectively. Rectal temperature decreases produced by MDPV in the controls were reduced in duration in the MDPV-KLH vaccine group. A separate group (N = 19) was trained to intravenously self-administer α-PVP (0.05, 0.1 mg/kg/inf) and vaccinated with KLH or α-PVP-KLH, post-acquisition. Self-administration in α-PVP-KLH rats was initially higher than in the KLH rats but then significantly decreased following a final vaccine booster, unlike the stable intake of KLH rats. The data demonstrate that active vaccination provides functional protection against the effects of α-PVP and MDPV, in vivo, and recommend additional development of vaccines as potential therapeutics for mitigating the effects of designer cathinone derivatives.


Asunto(s)
Benzodioxoles/farmacología , Estimulantes del Sistema Nervioso Central/farmacología , Pentanonas/farmacología , Psicotrópicos/farmacología , Pirrolidinas/farmacología , Trastornos Relacionados con Sustancias/prevención & control , Vacunas , Administración Intravesical , Animales , Benzodioxoles/sangre , Benzodioxoles/inmunología , Temperatura Corporal/efectos de los fármacos , Estimulantes del Sistema Nervioso Central/sangre , Estimulantes del Sistema Nervioso Central/inmunología , Drogas de Diseño/farmacocinética , Drogas de Diseño/farmacología , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Inmunoconjugados , Masculino , Metanfetamina/sangre , Metanfetamina/inmunología , Actividad Motora/efectos de los fármacos , Pentanonas/sangre , Pentanonas/inmunología , Psicotrópicos/sangre , Psicotrópicos/inmunología , Pirrolidinas/sangre , Pirrolidinas/inmunología , Ratas Sprague-Dawley , Autoadministración , Trastornos Relacionados con Sustancias/sangre , Trastornos Relacionados con Sustancias/inmunología , Vacunación , Cathinona Sintética
17.
J Med Chem ; 60(1): 338-348, 2017 01 12.
Artículo en Inglés | MEDLINE | ID: mdl-27966961

RESUMEN

Botulinum neurotoxin A (BoNT/A) is one of the most deadly toxins and is the etiological agent of the potentially fatal condition, botulism. Herein, we investigated 8-hydroxyquinoline (quinolin-8-ol) as a potential inhibitor scaffold for preventing the deadly neurochemical effects of the toxin. Quinolinols are known chelators that can disrupt the BoNT/A metalloprotease zinc-containing active site, thus impeding its proteolysis of the endogenous protein substrate, synaptosomal-associated protein 25 (SNAP-25). By use of this information, the structure-activity relationship (SAR) of the quinolinol-5-sulfonamide scaffold was explored through preparation of a crude sulfonamide library and evaluation of the library in a BoNT/A LC enzymatic assay. Potency optimization of the sulfonamide hit compounds was undertaken as informed by docking studies, granting a lead compound with a submicromolar Ki. These quinolinol analogues demonstrated inhibitory activity in a cell-based model for SNAP-25 cleavage and an ex vivo assay for BoNT/A-mediated muscle paralysis.


Asunto(s)
Toxinas Botulínicas Tipo A/antagonistas & inhibidores , Hidroxiquinolinas/farmacología , Animales , Toxinas Botulínicas Tipo A/metabolismo , Células Cultivadas , Hidroxiquinolinas/química , Masculino , Ratones , Simulación del Acoplamiento Molecular , Proteolisis , Relación Estructura-Actividad
18.
Chem Commun (Camb) ; 52(84): 12521-12524, 2016 Oct 13.
Artículo en Inglés | MEDLINE | ID: mdl-27722252

RESUMEN

In developing small-molecule inhibitors of botulinum neurotoxin serotype A light chain (BoNT/A LC), substituted picolinic acids were identified. Extensive investigation into the SAR of the picolinic acid scaffold revealed 5-(1-butyl-4-chloro-1H-indol-2-yl)picolinic acid (CBIP), which possessed low micromolar activity against BoNT/A. Kinetic and docking studies demonstrated binding of CBIP to the ß-exosite: a largely unexplored site on the LC that holds therapeutic relevance for botulism treatment.


Asunto(s)
Toxinas Botulínicas Tipo A/antagonistas & inhibidores , Quelantes del Hierro/farmacología , Ácidos Picolínicos/farmacología , Inhibidores de Proteasas/farmacología , Humanos
19.
J Med Chem ; 59(8): 3878-85, 2016 04 28.
Artículo en Inglés | MEDLINE | ID: mdl-27054372

RESUMEN

Methamphetamine (MA) addiction is a serious public health problem, and current methods to abate addiction and relapse are currently ineffective for mitigating this growing global epidemic. Development of a vaccine targeting MA would provide a complementary strategy to existing behavioral therapies, but this has proven challenging. Herein, we describe optimization of both hapten design and formulation, identifying a vaccine that elicited a robust anti-MA immune response in mice, decreasing methamphetamine-induced locomotor activity.


Asunto(s)
Haptenos/inmunología , Metanfetamina/inmunología , Vacunas/inmunología , Animales , Formación de Anticuerpos , Ensayo de Inmunoadsorción Enzimática , Locomoción/efectos de los fármacos , Metanfetamina/farmacología , Ratones , Vacunas/química
20.
Angew Chem Int Ed Engl ; 55(11): 3772-5, 2016 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-26879590

RESUMEN

Fentanyl is an addictive prescription opioid that is over 80 times more potent than morphine. The synthetic nature of fentanyl has enabled the creation of dangerous "designer drug" analogues that escape toxicology screening, yet display comparable potency to the parent drug. Alarmingly, a large number of fatalities have been linked to overdose of fentanyl derivatives. Herein, we report an effective immunotherapy for reducing the psychoactive effects of fentanyl class drugs. A single conjugate vaccine was created that elicited high levels of antibodies with cross-reactivity for a wide panel of fentanyl analogues. Moreover, vaccinated mice gained significant protection from lethal fentanyl doses. Lastly, a surface plasmon resonance (SPR)-based technique was established enabling drug-specificity profiling of antibodies derived directly from serum. Our newly developed fentanyl vaccine and analytical methods may assist in the battle against synthetic opioid abuse.


Asunto(s)
Analgésicos Opioides/toxicidad , Antídotos/uso terapéutico , Drogas de Diseño/síntesis química , Fentanilo/toxicidad , Vacunas/administración & dosificación , Animales , Fentanilo/farmacocinética , Ratones , Distribución Tisular
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA