Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Michael D. Hambuchen is active.

Publication


Featured researches published by Michael D. Hambuchen.


Cns & Neurological Disorders-drug Targets | 2011

Monoclonal antibodies as pharmacokinetic antagonists for the treatment of (+)-methamphetamine addiction.

S. Michael Owens; William T. Atchley; Michael D. Hambuchen; Eric C. Peterson; W. Brooks Gentry

Developing specific medications to treat (+)-methamphetamine (METH) addiction is a difficult challenge because METH has multiple sites of action that are intertwined with normal neurological function. As a result, no small molecule medication for the treatment of METH addiction has made it through the FDA clinical trials process. With the invention of a new generation of proteinbased therapies, it is now possible to consider treating drug addiction by an entirely different approach. This new approach is based on the discovery of very high affinity anti-METH monoclonal antibodies (mAbs), which are non-addictive and antagonize METH effects from the blood stream without entering the brain. Due to a very long biological half-life, anti-METH mAbs would only need to be administered once every 2-4 weeks, aiding in patient compliance. As a relapse prevention medication, anti-METH mAbs could reduce or prevent the rewarding effects of a relapse to METH use and thereby improve a patients probability of remaining in therapy and recovering from their addiction. In this review, we discuss the discovery process of anti-METH mAbs, with a focus on the preclinical development leading to high affinity anti-METH mAb antagonists.


Journal of Medicinal Chemistry | 2015

Combining Active Immunization with Monoclonal Antibody Therapy to Facilitate Early Initiation of a Long-acting Anti-methamphetamine Antibody Response

Michael D. Hambuchen; F. Ivy Carroll; Daniela Rüedi-Bettschen; Howard P. Hendrickson; Leah J. Hennings; Bruce E. Blough; Lawrence E. Brieaddy; Ramakrishna R. Pidaparthi; S. Michael Owens

We hypothesized that an anti-METH mAb could be used in combination with a METH-conjugate vaccine (MCV) to safely improve the overall quality and magnitude of the anti-METH immune response. The benefits would include immediate onset of action (from the mAb), timely increases in the immune responses (from the combined therapy) and duration of antibody response that could last for months (from the MCV). A novel METH-like hapten (METH-SSOO9) was synthesized and then conjugated to immunocyanin monomers of keyhole limpet hemocyanin (IC(KLH)) to create the MCV ICKLH-SOO9. The vaccine, in combination with previously discovered anti-METH mAb7F9, was then tested in rats for safety and potential efficacy. The combination antibody therapy allowed safe achievement of an early high anti-METH antibody response, which persisted throughout the study. Indeed, even after 4 months the METH vaccine antibodies still had the capacity to significantly reduce METH brain concentrations resulting from a 0.56 mg/kg METH dose.


Vaccine | 2014

Treatment of rats with an anti-(+)-methamphetamine monoclonal antibody shortens the duration of action of repeated (+)-methamphetamine challenges over a one month period

Michael D. Hambuchen; Daniela Rüedi-Bettschen; D. Keith Williams; Howard P. Hendrickson; S. Michael Owens

This study assessed clinical scenarios of continuing monoclonal antibody (mAb) treatment for (+)-methamphetamine (METH) addiction, and the implications of missing or discontinuing this therapy. We hypothesized that chronic anti-METH mAb7F9 (METH KD=9 nM) treatment of rats could significantly decrease METH-induced behaviors; even with repeated METH challenges, use of METH doses in excess of mAb binding sites, and after discontinuing mAb treatment which results in a 10-fold reduction in mAb7F9 serum concentrations. Male Sprague Dawley rats (n=6/group) were treated with i.v. saline or a loading dose of mAb7F9 to achieve instant steady-state conditions followed by two weekly (141 mg/kg) doses ending on day 14. METH (0.56 mg/kg) was administered 4h and three days after each saline or mAb7F9 treatment, and on day 21. This produced locomotion and rearing behavior that lasted about 120 min in control rats. In mAb7F9 treated rats, METH-induced distance traveled was significantly reduced from 60 to 120 min (P<0.05) on days 0-21 and rearing was significantly reduced from 60 to 120 min on days 0-17. METH serum concentrations determined 5h after METH dosing was significantly increased in mAb7F9-treated rats after all METH challenges. On days 24 and 28 (the final day), the rats were administered a 3-fold higher METH dose (1.68 mg/kg). MAb7F9 treated rats showed a substantially earlier termination of the METH-induced locomotion on both days, even though the METH dose exceeded mAb7F9s binding capacity. METH brain concentrations determined 5h after METH on day 28 were also significantly decreased in mAb7F9-treated rats. In conclusion, over one month, mAb7F9 significantly and continuously bound METH and reduced METH-induced locomotor effects even after discontinuation of mAb treatment and challenge with higher METH doses.


Human Vaccines & Immunotherapeutics | 2014

Pharmacological effects of two anti-methamphetamine monoclonal antibodies: Supporting data for lead candidate selection for clinical development

Elizabeth M. Laurenzana; Misty W Stevens; John C Frank; Michael D. Hambuchen; Howard P. Hendrickson; Sarah J. White; D. Keith Williams; S. Michael Owens; W. Brooks Gentry

This lead candidate selection study compared two anti-(+)-methamphetamine (METH) monoclonal antibodies (mAb) to determine their ability to reduce METH-induced locomotor effects and redistribute METH and (+)-amphetamine (AMP) in a preclinical overdose model. Both mAbs have high affinity for METH, but mAb4G9 has moderate and mAb7F9 has low affinity for AMP. In the placebo-controlled behavioral experiment, the effects of each mAb on the locomotor response to a single 1 mg/kg intravenous (IV) METH dose were determined in rats. The doses of mAb binding sites were administered such that they equaled 1, 0.56, 0.32, and 0.1 times the molar equivalent (mol-eq) of METH in the body 30 min after the METH dose. METH disposition was determined in separate animals that similarly received either a 1 or 0.32 mol-eq dose of mAb binding sites 30 min after a 1 mg/kg METH dose. Total METH-induced distance traveled was significantly reduced in rats that received the highest three doses of each mAb compared with saline. The duration of METH effects was also significantly reduced by mAb7F9 at the highest dose. The disposition of METH was altered dose-dependently by both mAbs as shown in reductions of volume of distribution and total clearance, and increases in elimination half-life. These data indicate that both mAbs are effective at reducing METH-induced behavior and favorably altering METH disposition. Both were therefore suitable for further preclinical testing as potential human medications for treating METH use; however, due to results reported here and in later studies, mAb7F9 was selected for clinical development.


Drug and Alcohol Dependence | 2015

The pharmacokinetics of methamphetamine self-administration in male and female rats

Alessandra Milesi-Hallé; Michael D. Hambuchen; Donald E. McMillan; S. Michael Owens

BACKGROUND Because methamphetamine (METH) pharmacokinetics after single iv doses show significant differences between male and female rats, we hypothesized that pharmacokinetic differences in METH disposition could be a contributing factor to the patterns of METH self-administration behaviors in rats. METHODS For the studies, we used a passive (non-contingent) METH dosing schedule consisting of 27 METH iv bolus injections (0.048mg/kg) over 2h derived from a previous active (contingent) METH self-administration behavioral study in male rats. After METH dosing of male and female Sprague-Dawley rats (n=5/group), METH and amphetamine serum concentrations were determined by LC-MS/MS. Pharmacokinetic analysis, including predictive mathematical simulations of the data, was then conducted. RESULTS Male and female rats achieved relatively stable METH serum concentrations within 20min, which remained constant from 20 to 120min. While not statistically different, METH clearance and volume of distribution values for females were 25% and 33% lower (respectively) than males. Linear regression analysis of predicted METH concentrations from pharmacokinetic simulations versus observed concentrations showed a substantially better correlation with male data than female data (r(2)=0.71 vs. 0.56; slope=0.95 vs. 0.45, respectively). At 120min, the time of predicted peak METH serum concentrations, female values were 42% higher than expected, while male values were within 3%. CONCLUSIONS Unlike METH male pharmacokinetic data, the female data was less predictable during multiple METH administrations and produced overall higher than expected METH concentrations. These findings demonstrate that METH pharmacokinetics could contribute to differences in METH self-administration behaviors in rats.


Drug and Alcohol Dependence | 2017

The pharmacokinetics of racemic MDPV and its (R) and (S) enantiomers in female and male rats

Michael D. Hambuchen; Howard P. Hendrickson; Melinda G. Gunnell; Samantha J. McClenahan; Laura E. Ewing; Dillon M. Gibson; Michael D. Berquist; S. Michael Owens

BACKGROUND These studies investigated the serum pharmacokinetic (PK) profile of racemic (3,4)-methylenedioxypyrovalerone [(R,S)-MDPV)] and its (R)- and (S)-enantiomers in female and male Sprague Dawley rats. METHODS Intravenous (R,S)-MDPV (3 and 5.6mg/kg) and single enantiomer of (R)- and (S)-MDPV (1.5mg/kg) were administered to both sexes for PK studies. Intraperitoneal (ip) bioavailability was determined at 3mg/kg (R,S)-MDPV. Locomotor activity studies were conducted after ip treatment with saline and 0.3-5.6mg/kg of (R,S)-MDPV. RESULTS PK values after iv (R,S)-MDPV showed a significant (p<0.05) sex-dependent differences in the volume of distribution at steady state (Vdss) for (R)- and (R,S)-MDPV at both (R,S)-MDPV doses. The female S/R enantiomeric ratios for area under the concentration time curve (AUCinf) and clearance were significantly lower and higher, respectively, than values determined in males. Importantly, there was no evidence of in vivo inversion of (R)-MDPV or (S)-MDPV to its antipode. There were, however, significant sex-dependent differences in volume of distribution after administration of the (R)-enantiomer. Bioavailability studies of ip (R,S)-MDPV showed greater variability and significantly greater bioavailability in male rats. Accordingly, there was a significantly greater maximal distance traveled measurement in male rats at a 3.0mg/kg dose. CONCLUSION PK sex differences in (R,S)-MDPV and enantiomers were most apparent in volume of distribution, which could be caused by differences in drug blood and tissue protein binding. The increased magnitude and variance in ip bioavailability in male compared to female rats could lead to sex-dependent differences in the pharmacological action caused by active enantiomer (S)-MDPV.


Analytical Methods | 2017

Chiral determination of 3,4-methylenedioxypyrovalerone enantiomers in rat serum

Michael D. Hambuchen; Howard P. Hendrickson; S. Michael Owens

The emerging stimulant drug of abuse (3,4)-methylenedioxypyrovalerone [(R,S)-MDPV] is self-administered as a racemic mixture by intranasal, iv, oral, and smoking routes. The individual enantiomers are known to have widely different pharmacological effects, with (S)-MDPV showing much greater potency than (R)-MDPV in pharmacological testing. The goal of these studies was to develop and validate an analytical method for quantitation of (R)-MDPV, (S)-MDPV and (R,S)-MDPV in small volumes of rat serum using a chiral separation column and liquid chromatography-mass spectrometry. The method was validated for selectivity, precision, accuracy, recovery, sensitivity, and reproducibility. The method was also used to determine the enantiomeric stability of the individual enantiomers during sample cleanup and analysis. The linear dynamic range of the calibration curve was 1 - 1000 ng/ml for each enantiomer. Concentration values for the lower limit of quantitation (1 ng/ml) were within 30% of their nominal value, but all other calibration standards were <20% of their nominal value. With proper storage and handling of samples, the two MDPV enantiomers were shown to remain stable in rat serum without any apparent racemization during the time needed for analysis. Finally, the ruggedness of the method was demonstrated with diluted and undiluted serum samples collected from Sprague Dawley rats in a preliminary pharmacokinetic study at 3 mg/kg of (R,S)-MDPV. In summary, the assay used a simple sample preparation method, reversed-phase chiral chromatography, and tandem mass spectrometry to achieve accurate and selective determinations of MDPV enantiomer concentrations in small volumes of serum.


Archive | 2016

Discovery and Development of an Anti-methamphetamine Monoclonal Antibody for Use in Treating Methamphetamine Abuse

Michael D. Hambuchen; Misty W Stevens; Melinda G. Gunnell; W. Brooks Gentry; S. Michael Owens

Anti-methamphetamine monoclonal antibodies (mAb) can reduce the pharmacological effects of methamphetamine (METH) in rodent models of METH abuse. Rather than a direct action in the brain, the mAb medication binds METH with high affinity in the bloodstream leading to a reduction and slowing of METH brain penetration. Through an extensive discovery and development process, prototype mouse anti-METH mAb medications were selected and tested in preclinical studies to select a final mAb with both high affinity for METH and long-term functionality in vivo. This antibody was then converted into a chimeric anti-METH mAb suitable for human use. In a Phase 1a clinical trial, the medication proved safe, with a half-life of 18 days. Because of this prolonged half-life, patients might only need mAb treatment once every 3 weeks to aid in the protection from relapse to METH abuse. Additional safety and efficacy will be tested in future clinical trials.


Human Vaccines & Immunotherapeutics | 2016

Chronic treatment of (+)-methamphetamine-induced locomotor effects in rats using one or a combination of two high affinity anti-methamphetamine monoclonal antibodies

Michael D. Hambuchen; Daniela Rüedi-Bettschen; Melinda G. Gunnell; Howard P. Hendrickson; S. Michael Owens

ABSTRACT We hypothesized that treatment of methamphetamine (METH) effects with a mixture of 2 high affinity anti-METH monoclonal antibodies (mAb) with differing molecular recognition for METH-like structures could increase efficacy compared to treatment with a single mAb. The antibodies studied were mAb7F9 (METH and amphetamine [AMP] KD = 7.7 and 270 nM) and mAb4G9 (16 nM and 110 nM, respectively) in a 50:50 mixture. Adult male Sprague Dawley Rats were treated with iv saline or a loading dose of mAb7F9-mAb4G9 (141 mg/kg of each mAb) followed by 2 weekly doses (70.5 mg/kg total) on days 7 and 14. METH challenge doses (0.56 mg/kg) were administered 4 hrs and 3 days after each mAb7F9-mAb4G9 treatment, and 7 days after the final treatment (day 21). Locomotor activity (0–4 hrs) and serum METH and AMP concentrations (at 5 hrs) were measured after each METH challenge. MAb7F9-mAb4G9 treatment significantly reduced the duration of locomotor activity after 6 of the 7 METH doses (P < 0.05) and significantly increased serum METH and AMP concentrations. Administering three-fold higher METH doses (1.68 mg/kg) on days 24 and 28 showed mAb7F9-mAb4G9 treatment had negligible effects on the duration of METH-induced locomotor activity. These data were then compared to previous monotherapy data. While mAb7F9-mAb4G9 therapy inhibited the effects of multiple METH challenge doses, the inhibition was not as profound or as long lasting as the effects of mAb7F9 treatment alone. These data demonstrate the importance of both mAb affinity and specificity in the production of effective, long-lasting anti-METH mAb therapies.


PLOS ONE | 2018

Development and testing of AAV-delivered single-chain variable fragments for the treatment of methamphetamine abuse

Charles E. Hay; Guillermo A. Gonzalez; Laura E. Ewing; E. Elizabeth Reichard; Michael D. Hambuchen; Nisha Nanaware-Kharade; Sinthia Alam; Chris T. Bolden; S. Michael Owens; Paris Margaritis; Eric C. Peterson

Methamphetamine (METH) substance abuse disorders have major impact on society, yet no medications have proven successful at preventing METH relapse or cravings. Anti-METH monoclonal antibodies can reduce METH brain concentrations; however, this therapy has limitations, including the need for repeated dosing throughout the course of addiction recovery. An adeno-associated viral (AAV)-delivered DNA sequence for a single-chain variable fragment could offer long-term, continuous expression of anti-METH antibody fragments. For these studies, we injected mice via tail vein with 1 x 1012 vector genomes of two AAV8 scFv constructs and measured long-term expression of the antibody fragments. Mice expressed each scFv for at least 212 days, achieving micromolar scFv concentrations in serum. In separate experiments 21 days and 50 days after injecting mice with AAV-scFvs mice were challenged with METH in vivo. The circulating scFvs were capable of decreasing brain METH concentrations by up to 60% and sequestering METH in serum for 2 to 3 hrs. These results suggest that AAV-delivered scFv could be a promising therapy to treat methamphetamine abuse.

Collaboration


Dive into the Michael D. Hambuchen's collaboration.

Top Co-Authors

Avatar

S. Michael Owens

University of Arkansas for Medical Sciences

View shared research outputs
Top Co-Authors

Avatar

Howard P. Hendrickson

University of Arkansas for Medical Sciences

View shared research outputs
Top Co-Authors

Avatar

Daniela Rüedi-Bettschen

University of Arkansas for Medical Sciences

View shared research outputs
Top Co-Authors

Avatar

Melinda G. Gunnell

University of Arkansas for Medical Sciences

View shared research outputs
Top Co-Authors

Avatar

W. Brooks Gentry

University of Arkansas for Medical Sciences

View shared research outputs
Top Co-Authors

Avatar

D. Keith Williams

University of Arkansas for Medical Sciences

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Laura E. Ewing

University of Arkansas for Medical Sciences

View shared research outputs
Top Co-Authors

Avatar

Alessandra Milesi-Hallé

University of Arkansas for Medical Sciences

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge