Abstract
(+)-Methamphetamine (METH) addiction is a chronic disease that interferes with fundamental brain-mediated behaviors and biological functions like eating. These studies present preclinical efficacy and safety profiles for a METH conjugate vaccine (ICKLH-SMO9) designed to treat METH abuse. ICKLH-SMO9 efficacy and safety were assessed over a 16-week period by monitoring general health and stability of responding in a food maintained behavioral paradigm. Male Sprague-Dawley rats were trained to lever press for food reinforcers until stable behavior was established. Rats (n=9/group) were then immunized with 100 µg of a control antigenic carrier protein (ICKLH-Cys) or ICKLH-SMO9 in Alhydrogel® adjuvant, with booster immunizations at 4, 8 and 12 weeks. Health, immunization site and behavior were assessed daily. No adverse effects were found. During weeks 14–16, when antibody titers and METH affinity (Kd = 13.9 ± 1.7 nM) were maximal, all rats received progressively higher METH doses (0.3–3.0 mg/kg) every 3–4 days, followed by behavioral testing. Even though the lower METH doses from 0.3-1.0 mg/kg produced no impairment in food maintained behavior, 3.0-mg/kg in control rats showed significantly (p<0.05) reduced response rates and number of reinforcers earned, as well as reduced food intake. In sharp contrast, the ICKLH-SMO9 group showed no changes in food maintained behavior at any METH dose, even though METH serum concentrations showed profound increases due to anti-METH antibody binding. These findings suggest the ICKLH-SMO9 vaccine is effective and safe at reducing adverse METH-induced effects, even at high METH doses.
Keywords: methamphetamine, vaccine, hapten, drug abuse, preclinical studies, rats
Introduction
(+)-Methamphetamine (METH) abuse is a worldwide problem, causing deleterious effects in users and enormous costs to communities [1,2]. While METH users report positive effects like increased energy, euphoria and appetite suppression [3,4]; chronic use results in addiction, organ system dysfunction, weight loss and neurotoxicity [4–6]. Rather than affecting a single site of action or organ system, METH use interferes with a range of medically important functions [7,8].
Because METH addiction is a root cause of most METH-associated health problems, a variety of addiction pharmacotherapies have been tested. Unfortunately no drug(s) have proven effective by United States FDA standards, but a few drugs are useful for acute supportive and symptomatic treatment [8]. The most successful addiction therapy is cognitive behavioral intervention, but even when patients successfully complete behavioral treatment, 36% use METH within 6 months and by 13 months 51% are back to METH use [9].
Combining behavioral treatments with medications to lessen the medical impact of METH use could potentially improve patient health and increase the chance of successful treatment [3,10]. An antibody-based medication could be either anti-METH monoclonal antibody (mAb) [2,11–13] or antibodies generated by active immunization with a METH conjugate vaccine [11,13–16].
Preclinical rodent studies show that active immunization with haptens (derived from drugs of abuse) conjugated to antigenic proteins can favorably alter behavioral and pharmacokinetic properties of addictive drugs like cocaine, heroin, METH, morphine, nicotine, and oxycodone [11,17–20]. Under optimal conditions active immunization can lead to high titer and high affinity immune responses [21–24], decrease drug-induced locomotor activity [14,15,25–28], prevent drug-induced reinstatement [11,28] and alter (i.e., increase or decrease) drug self-administration in animal studies [11,16,29–31].
Importantly, studies in rats show METH use during the immunization period does not affect the affinity or titer of the immune response to a METH-conjugate vaccine (MCV) [10]. This is clinically important since many patients are likely to use drugs of abuse during the development of an immune response. While vaccination is a promising treatment, two clinical trials of a nicotine vaccine and one trial of a cocaine vaccine failed to meet their Phase II clinical endpoints [21,23,24,32]. Two shared problems among all three vaccines were a large variation in response and less than optimal affinity for the drug of abuse.
The purpose of the present studies was to evaluate the efficacy and safety of a MCV [11,13] in rats, and to better understand the potential for clinically approved aluminum-based adjuvants like Alhydrogel® to augment the immune response. Behavioral studies showed that a high METH dose substantially impaired the control rats’ ability to continue stable food maintained behavior, but did not affect the ability of MCV immunized rats to respond for food, or eat the earned food reinforcers, which demonstrated an effective health benefit of the MCV.
Methods
Drugs and Chemicals
Reagents were purchased from Sigma Aldrich (St. Louis, MO) unless noted otherwise. (+)-Methamphetamine hydrochloride, (+)-amphetamine sulfate (AMP), and 3H-METH (23.5 Ci/mmol) were obtained from the NIDA Drug Supply Program. Doses were calculated as the free base.
Vaccines
The METH-like hapten HSMO9 (Figure 1) was covalently bound to immunocyanin to produce the MCV (ICKLH-SMO9) by the method of Carroll et al. [11]. Immunocyanin (ICKLH; Biosyn Corp., Carlsbad, CA) is derived from native keyhole limpet hemocyanin (KLH) and consisted of two stable subunit monomers (~360 and ~390 kDa). Both native KLH (8–32 million Da [33]) and ICKLH [25] are used in human vaccines. The control vaccine (ICKLH-Cys) was synthesized by conjugating the activated ICKLH to L-cysteine (Cys), instead of HSMO9.
Figure 1.
Chemical structures of METH, METH-like hapten (HSMO9; (S)-N-(2-(mercaptoethyl)-6-(3-(2-(methylamino)propyl)phenoxy)hexanamide), maleimide activated immunocyanin, ICKLH-Cys, and the MCV (ICKLH-SMO9). The large molecular weight immunocyanin (ICKLH) antigenic carrier protein is symbolically represented as a large circle. When the synthesis reactions for ICKLH-Cys and ICKLH-SMO9 were complete, a large excess of L-cysteine was added to the reaction mixture to block any potentially unreacted maleimide sites before use in immunizations.
Gel electrophoresis and Western blot analysis
The proteins were separated using the Fluorescent LP-Next Gel system (Amresco LLC, Solon, OH) and were then transferred to an Immobilon-NC Millipore membrane (Thermo Fisher Scientific, Waltham, MA) for Western blot analysis. Detection of the METH-like hapten was with anti-METH mAb4G9 [34] and an anti-mouse IgG-conjugated peroxidase secondary antibody.
Animal Subjects
All experiments were conducted in accordance with the NIH Guide for the Care and Use of Laboratory Animals and the Institutional Animal Care and Use Committee of the University of Arkansas for Medical Sciences. Adult male Sprague-Dawley rats (n=18; Charles River Laboratories, Raleigh, NC) were housed two per cage. Water was provided ad libitum and sufficient food was provided after behavioral sessions to maintain rat body weights at approximately 320 g.
Behavioral Apparatus
Experimental sessions were conducted in operant chambers (Med Associates, Georgia, VT) equipped with two levers, a white stimulus light above each lever with a house light on the opposite wall, a sound generator, and a food hopper connected to a pellet dispenser. Recording of responses and activation of lights and sound was computer controlled (MED-PC IV, Med Associates, Georgia, VT).
Food maintained behavior
Behavioral sessions were run Monday-Friday. Rats were trained to lever press for food reinforcers (45 mg grain-based Dustless Precision pellets; Bio-Serv, Frenchtown, NJ) under a second order fixed-interval (FI), fixed-ratio (FR) schedule of reinforcement. In the presence of the house and a white light above the reinforced lever, completion of every 5th response (FR5) during a 30s FI resulted in the concurrent flashing of the stimulus lights above both levers and sounding of a tone for 4s. Completion of the first FR after expiration of the FI resulted in the delivery of two food pellets (defined as one reinforcer), simultaneous with the flashing lights and sound activation. Daily sessions ended after 25 reinforcers earned, or two hrs, whichever occurred first. After responding stabilized, a 10-day baseline response measurement was collected. Responding was then extinguished by omitting the food reinforcer. Extinction sessions were run for a minimum of 10 days to achieve stable low responding. Following extinction, food reinforcers were made available and reacquisition of food maintained behavior was assessed for 10 sessions, at which point rats were back to baseline responding.
Immunization
Once trained, rats were assigned to immunization groups based on pre-established statistical criteria (as described in the statistics section) to prevent behavioral differences between groups. Rats were immunized subcutaneously with 100 µg of ICKLH-Cys or ICKLH-SMO9 in Alhydrogel® 85 adjuvant (Brenntag Biosector; Frederikssund, Denmark). Booster injections were at 4, 8 and 12 weeks.
Quantitation of anti-METH polyclonal antibody binding
Blood samples were collected two days before and 12 days after immunizations. Rapid equilibrium dialysis (RED; Thermo Scientific, Rockville, IL) and a [3H]-METH tracer were used to measure antibody functional titers. Each rat serum was diluted 1:50, 1:100 and 1:300 in 0.1 M sodium phosphate with 0.15 M NaCl, pH 7.35 buffer. Then aliquots of each dilution (containing 5 nM [3H]-METH) were added to one chamber of the RED device and buffer to the other chamber. Plates were incubated overnight at 4°C to reach equilibrium. The [3H]-METH in each chamber was quantitated by liquid scintillation spectrophotometry to determine the percentage of bound [3H]-METH in serum.
METH Kd (dissociation constant) determinations in ICKLH-SMO9 immunized rats
After the third boost, Kd values for METH binding in serum samples were determined using a bead-based RIA that is specific for IgG binding [13]. METH Kd values from each antiserum were determined after correction for the binding of [3H]-METH tracer in the presence of unlabeled METH [35]
METH challenge dosing
The goal was to test the ability of the ICKLH-Cys (control) and ICKLH-SMO9 vaccine to alter METH (and AMP metabolite) pharmacokinetics and METH-induced behavioral responses at week 14–16. Rats received subcutaneous saline followed by progressively increasing METH doses every 3–4 days (0.3–3.0 mg/kg METH). Immediately after dosing, a food maintained behavioral session was conducted. Testing of food maintained behavior continued Monday-Friday. Two hrs after saline or METH administration, a blood sample was collected to measure METH and AMP serum concentrations by LC-MS/MS [36].
Treatment safety observations
Rats were weighed and checked for signs of ill health Monday-Friday. Careful attention was given to sites of immunization for signs of inflammation, lesions or swelling. After METH challenge sessions, rats were observed in their home cage at regular intervals for unexpected changes in general appearance or behavior. The trained observer was blind to the treatments.
Statistical Analysis
Average number of responses, session time and response rate during acquisition, extinction and reacquisition of food maintained behavior was used to sort the rats into performance-matched immunization groups, verified by t-tests. Behavioral response data were analyzed using two-way repeated measures (RM) analysis of variance (ANOVA) (day × immunization group), followed by Bonferroni Multiple Comparison tests.
To assess food maintained behavior and body weight data during the immunization period, a two-way RM ANOVA (time point × immunization group) was conducted, followed by Bonferroni Multiple Comparison tests. To determine immunization effects on food maintained behavior after METH challenges, a two-way RM ANOVA was performed (METH dose × immunization group), followed by Bonferroni Multiple Comparisons.
Serum METH and AMP concentrations after METH challenges were analyzed using a two-way RM ANOVA (METH dose × immunization group), followed by Bonferroni Multiple Comparison tests. Statistical analyses were conducted using SigmaStat (Aspire Software International, Ashburn, VA). Comparisons were considered statistically different at p<0.05.
Results
Gel and Western blot analysis
While the initial electrophoresis step after maleimide activation of ICKLH did not separate the ~360 and ~390 kDa proteins (Figure 2), the final ICKLH-Cys and ICKLH-SMO9 antigens showed two bands, with a significant and equal amount of METH hapten conjugated to both immunocyanin monomers.
Figure 2.
Gel electrophoresis and Western blot analysis of ICKLH, ICKLH-Cys and ICKLH-SMO9. Protein concentration for all lanes ranged from 0.73–0.75 mg/ml. The upper panel shows the migration of ICKLH carrier protein (Lane 1). ICKLH-Cys is shown in Lanes 2 and 5, and the final MCV ICKLH-SMO9 is shown in Lanes 3 and 4. The proteins shown in Lanes 2–3 were treated with an additional large excess of L-cysteine as a final step in the synthesis to “cap” or block potentially unreacted maleimide groups in the ICKLH-Cys (control) and ICKLH-SMO9 antigens. The purpose of this step was to reduce the possibility of unwanted covalent binding to protein and cellular sites in vivo. The proteins in Lanes 4 and 5 were not treated with an excess of Cys. The lower panel shows the same proteins analyzed by Western blot. The antibody for the Western blot analysis was anti-METH mAb4G9 [13,34]. The immune reactivity of the ICKLH-SMO9 protein with mAb4G9 appeared unaffected by the addition of excess L-cysteine, and did not show immunoreactivity with the proteins in other lanes, which did not contain the METH hapten.
Food maintained behavior performance testing and METH binding over time
Table 1 summarizes the food maintained behavior results before the assignment to immunization groups. Daily testing during the immunization period showed no differences between immunization groups for any measures (p>0.23). Figure 3 shows the serum METH binding after each immunization, as measured with RED and [3H]-METH binding.
Table 1.
Summary of food maintained behavioral testing results before the assignment to immunization groups. All rats were trained to stable performance by 13.8 ± 0.3 days.
| Test stage | Responses (number) |
Response Rate (responses/s) |
Reinforcers (number) |
Session time (min) |
|---|---|---|---|---|
| Baseline | 414.3 ± 28.0 | 0.51 ± 0.04 | 25.0 | 15.4 ± 0.2 |
| Extinction | 35.2 ± 9.7 | 0.005 ± 0.001 | 5.0 ± 1.4 | 120 ± 6.9 |
| Reacquisition | Within 4 days, baseline levels of performance were reestablished. | |||
All values are the mean ± S.E.M. (n=18 rats)
Figure 3.
Development of anti-METH polyclonal antibody binding titers in rats using ICKLH-SMO9 (open circles indicate individual rat results) or ICKLH-Cys (without METH haptens, closed circle at week 14 along the x-axis) antigens with Alhydrogel® adjuvant during a 14-week period. The dashed line crosses each data set at the mean value for the group (n = 9 per group). Serum samples were obtained two days before and 12 days after each immunization. Arrows indicate the time of booster immunizations. The Kd value for METH binding at week 14 was 13.7 ± 1.7 nM (n=7). We were unable to accurately measure the Kd values in serum samples from the two rats with the lowest titers.
Both immunization groups showed similar behavioral responses by all measures following METH doses from 0.3 to 1.0 mg/kg (Figure 4). The major changes occurred after the 3.0 mg/kg METH challenge dose, when the ICKLH-Cys immunized rats showed decreased responding that was not statistically different (p<0.07), decreased response rates (p<0.001), decreased reinforcers earned (p<0.001), and increased session time (p<0.001) relative to ICKLH-SMO9 treated rats.
Figure 4.
Effects of ICKLH-Cys (control) and ICKLH-SMO9 immunization on food maintained behavior immediately after METH challenge doses from 0.3 to 3.0 mg/kg. The graphs show the number of lever presses on the reinforced lever (upper left), response rates (responses per second, upper right), session times (lower left) and number of food pellet reinforcers earned (lower right). Data are expressed as mean ± S.E.M. (n = 9 per group). The ** indicates a significant difference (p < 0.001) relative to the ICKLH-Cys control rats.
METH and AMP concentrations and Kd values in serum after immunization
Rats from both treatment groups showed METH-dose dependent increases in METH serum concentrations (Figure 5). However, ICKLH-SMO9 immunized rats showed significantly and substantively higher METH serum levels than control rats at all METH doses, except the lowest dose (p<0.06, Fig 5). AMP concentrations also increased in a dose-dependent manner, but only at 3.0 mg/kg did the METH ICKLH-SMO9 immunized rats show significantly higher AMP levels. The Kd value for METH binding at week 14 was 13.7 ± 1.7 nM (n=7).
Figure 5.
METH (upper graph) and AMP (lower graph) serum concentrations measured in rat serum samples collected 2 hrs after a METH challenge dose from 0.3 to 3.0 mg/kg in ICKLH-Cys control (open circle) and ICKLH-SMO9 immunized (closed circle) rats. The progressively increasing METH challenge doses were administered every 3–4 days from weeks 14–16. Each point represents an individual rat response and the horizontal line indicates the mean of the group. The * indicates a significant difference between ICKLH-Cys (control) and ICKLH-SMO9 immunized rats.
General safety assessment
Rat body weights showed a small increase from an initial weight of 327 ± 5.4 g to 349 ± 4.7 g (mean ± S.E.M.) after 16 weeks, but there were no differences between the ICKLH-Cys and ICKLH-SMO9 immunization groups (p>0.41). Daily health and behavioral monitoring did not reveal any signs of ill health or adverse effects attributable to the immunization, METH dosing, or the combination of immunization and dosing.
Discussion
These studies provide a preclinical efficacy and safety profile for a MCV designed to treat human METH addiction. We chose to use the ability of the animals to maintain stable food maintained behavior as an indication of efficacy, as well as a general measure of animal health. While this is not a standard test of vaccine protection from addiction behaviors, we hypothesized it could be an important measure since METH has significant negative health effects on eating behavior and body weight in humans [4]. Indeed, METH is approved in the US to treat obesity where other diets or medications have failed to produce weight loss.
At the highest METH challenge dose, control rats showed significant impairment (Figure 4) in their ability to continue normal behaviors. In contrast, the ICKLH-SMO9 treated rats appeared unaffected. An important observation is that even though some of the control rats did not seem overtly impaired in their performance of food maintained behavior after 3.0 mg/kg METH, five out of nine rats did not eat the food earned during the session. By comparison eight out of nine of the ICKLH-SMO9 immunized rats responded normally for the food, and all nine ate all earned food pellets. The significant adverse effect on control rats at this dose could be explained by METH-induced disruptions in a chain of behaviors, suggesting rats were able to respond on the lever, but unable to complete the process of eating the food. It is also possible the animals were too profoundly affected to eat the food reward because of METH-induced behaviors like sniffing, rearing, repetitive head and limb movement [37].
One of the most impressive behavioral findings was that the ICKLH-SMO9 immunized rats had sufficient antibody binding capacity to protect against the pharmacological effects of the highest dose of METH. Especially since it was administered after a 0.3, 0.56, and 1.0 mg/kg dose of METH. These progressively increasing doses, followed by the final 3.0 mg/kg METH dose should have substantially exceeded the stoichiometric binding capacity of the antibodies generated by active immunization. Thus the mechanism of action cannot be solely dependent on antibody binding capacity, as previously demonstrated [38,39].
In contrast to control rats, eight of the nine the ICKLH-SMO9 immunized rats were able to complete the behavioral task at the 3.0 mg/kg METH dose. The one adversely affected rat showed increased session time and failed to obtain all reinforcers within the 2 hr session. Although a Grubb’s outlier test showed this rat’s data were a statistical outlier in the ICKLH-SMO9 treatment group, we did not remove the data set. This rat’s immune response was the third lowest anti-METH titer on week 14 (Figure 3), and the only rat in this group to not respond normally for the food.
The finding that a low-titer rat was not fully protected from the METH-induced behavioral effects appears to partially mimic recent findings in clinical trials for cocaine and nicotine vaccines, where only a portion of the human populations achieved even a modest immune response [21,23,32]. In our study all ICKLH-SMO9 immunized rats had significantly higher METH serum concentrations compared to METH serum concentrations in controls (Figure 5), and even the rat with the lowest measured titer had elevated METH concentrations compared to controls. Thus, we think improvements in the choice of adjuvant, immunization schedule, and METH hapten epitope density could improve the uniformity and magnitude of the immune response. These studies are ongoing.
Based on our experience with METH-induced stimulant and stereotypic behaviors (e.g., [40], we expected the highest doses of METH (1.0 and 3.0 mg/kg) to produce significant and long lasting (e.g., up to 4–6 hrs) stereotypic and locomotor behaviors that would interfere with accurate and timely behavioral responses (Figure 4). A possible reason for not observing disruptions in their behavioral task may lie in the timing of the testing. We started the 2 hr testing period immediately after the METH challenge, and on average the rats completed the task within 15 min, up to the 1 mg/kg METH dose. However, the peak for potential disruptive locomotor activity and stereotype behaviors occurs at about 20 min after a 1 mg/kg METH dose [40,41] and even later after a 3.0 mg/kg dose.
The polyclonal antibodies generated by ICKLH-SMO9 immunization showed a high affinity for METH (Kd = 13.7 nM, see Figure 3 legend), which was comparable to three of the highest affinity anti-METH mAb produced in our laboratory (i.e., 6.8, 9, and 16 nM for METH) [13]. Studies of another anti-METH KLH-based vaccine report polyclonal anti-METH antibody Kd values of 130 nM in mice [42] and 30.4 nM in male Sprague-Dawley rats [14]. While the rats were the same outbred strain as in the current studies, most other study aspects differed including use of intact native KLH instead of pharmaceutical grade ICKLH monomers, a different hapten design (named MH6), a different immunization schedule, and the use of Sigma Adjuvant System instead of Alhydrogel®.
Antibody titers in ICKLH-SMO9 immunized rats rose steadily across the immunization period, resulting in an approximate 2-fold increase in titers from the first to third boost. However, the individual week 14 rat titers (and earlier) were variable (Figure 3), ranging from 40–88% (n=9) with a percent coefficient of variation of 23%. In our experience with rodents (unpublished observation), Alhydrogel® usually elicits lower antibody titers than other more potent preclinical adjuvants like Freund’s Complete adjuvant [43] or Sigma Adjuvant System. However, Alhydrogel® is one of only a few adjuvants approved for human use by the United States FDA. We think that the poor and variable immune responses in previous clinical trials of anti-cocaine and anti-nicotine vaccines was in part due to the poor ability of aluminum-based adjuvants like Alhydrogel® to stimulate robust immune responses [21], [23], [32]. This will need to be addressed in future anti-addiction vaccine trails.
Another essential aspect of vaccine development is safety of the vaccine protocol, and potential for adverse interactions with the antibody target (i.e., METH). Our MCV produced no reactions at the injection site or observable adverse health effects. Food maintained behavior (prior to METH) and home cage behavior were not different between control and ICKLH-SMO9 immunized rats, indicating no apparent negative effects. When METH was administered there were no observable additive or synergistic effects of the MCV on METH-induced effects.
Conclusions
In conclusion, repeated immunization with a high dose of ICKLH-SMO9 produced no adverse effects on health, body weight or performance during food maintained behavioral testing. Even when rats were challenged with increasing METH doses over a two-week period, there were no apparent additive or synergistic interactions between the immunization and METH. Administration of 3.0 mg/kg METH dramatically impaired food maintained behavior in control, but not ICKLH-SMO9 immunized rats. Even though low METH challenge doses did not seem to affect behavior differently in the treatment groups, ICKLH-SMO9 immunized rats still had higher METH serum concentrations for all doses, indicating that the antibodies generated by the ICKLH-SMO9 vaccine were effective at sequestering METH in the bloodstream. In a broader perspective these data suggest anti-METH vaccines could provide improvements in general health even when patients use METH at exceedingly high doses.
Highlights.
A vaccine (ICKLH-SMO9) for treatment of methamphetamine abuse was tested in rats
Repeated immunization with ICKLH-SMO9 caused no apparent adverse effect
ICKlh-SMO9 protects from methamphetamine-induced reductions in responding for food
METH serum concentrations substantially increased due to anti-METH antibody binding
These studies suggest the vaccine is effective and safe
Acknowledgements
This work was supported by grants from the National Institute on Drug Abuse (U01 DA23900 and DA05477) and the National Center for Advancing Translational Sciences (UL1TR000039).
Abbreviations
- AMP
(+)-amphetamine
- Cys
L-cysteine
- FI
fixed-interval schedule of behavioral reinforcement
- FR
fixed-ratio schedule of behavioral reinforcement
- [3H]-METH
(+)-[2’,6’ -3H(n)]-methamphetamine
- HSMO9
the METH-like hapten (S)-N-(2-(mercaptoethyl)-6-(3-(2-(methylamino)propyl)phenoxy)hexanamide
- ICKLH
immunocyanin monomers derived from native KLH
- ICKLH-Cys
maleimide activated immunocyanin end capped with cysteine
- ICKLH-SMO9
the METH-conjugate vaccine
- Kd
equilibrium dissociation constant
- KLH
keyhole limpet hemocyanin
- LC-MS/MS
liquid chromatography coupled to tandem mass spectrometry
- mAb
monoclonal antibody
- METH
(+)-methamphetamine
- MCV
METH-conjugate vaccine
- RED
rapid equilibrium dialysis
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
References
- 1.Nicosia N, Pacula RL, Kilmer B, Lundberg R, Chiesa J. The Economic Cost of Methamphetamine Use in the United States. Santa Monica, CA: Rand Corporation; 2009. [Google Scholar]
- 2.Owens S, Atchley W, Hambuchen M, Peterson E, Gentry W. Monoclonal antibodies as pharmacokinetic antagonists for the treatment of (+)-methamphetamine addiction. CNS Neurol Disord-DR. 2011;10:892–898. doi: 10.2174/187152711799219370. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Gentry WB, Rüedi-Bettschen D, Owens SM. Anti-(+)-methamphetamine monoclonal antibody antagonists designed to prevent the progression of human diseases of addiction. Clin Pharmacol Ther. 2010;88:390–393. doi: 10.1038/clpt.2010.155. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Cho AK. Ice: a new dosage form of an old drug. Science. 1990;249:631–634. doi: 10.1126/science.249.4969.631. [DOI] [PubMed] [Google Scholar]
- 5.Volkow ND, Chang L, Wang GJ, Fowler JS, Franceschi D, Sedler M, et al. Loss of dopamine transporters in methamphetamine abusers recovers with protracted abstinence. J Neurosci. 2001;21:9414–9418. doi: 10.1523/JNEUROSCI.21-23-09414.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Sekine Y, Ouchi Y, Takei N, Yoshikawa E, Nakamura K, Futatsubashi M, et al. Brain serotonin transporter density and aggression in abstinent methamphetamine abusers. Arch Gen Psychiatry. 2006;63:90–100. doi: 10.1001/archpsyc.63.1.90. [DOI] [PubMed] [Google Scholar]
- 7.Richards JR, Bretz SW, Johnson EB, Turnipseed SD, Brofeldt BT, Derlet RW. Methamphetamine abuse and emergency department utilization. West J Med. 1999;170:198–202. [PMC free article] [PubMed] [Google Scholar]
- 8.Albertson TE, Derlet RW, Van Hoozen BE. Methamphetamine and the expanding complications of amphetamines. West J Med. 1999;170:214–219. [PMC free article] [PubMed] [Google Scholar]
- 9.Brecht ML, Mayrhauser von C, Anglin MD. Predictors of relapse after treatment for methamphetamine use. J Psychoactive Drugs. 2000;32:211–220. doi: 10.1080/02791072.2000.10400231. [DOI] [PubMed] [Google Scholar]
- 10.Byrnes-Blake KA, Carroll FI, Abraham P, Owens SM. Generation of anti-(+)methamphetamine antibodies is not impeded by (+)methamphetamine administration during active immunization of rats. Int Immunopharmacol. 2001;1:329–338. doi: 10.1016/s1567-5769(00)00019-9. [DOI] [PubMed] [Google Scholar]
- 11.Carroll FI, Blough BE, Pidaparthi RR, Abraham P, Gong PK, Deng L, et al. Synthesis of Mercapto-(+)-methamphetamine Haptens and Their Use for Obtaining Improved Epitope Density on (+)-Methamphetamine Conjugate Vaccines. J Med Chem. 2011;54:5221–5228. doi: 10.1021/jm2004943. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Peterson EC, Laurenzana EM, Atchley WT, Hendrickson HP, Owens SM. Development and preclinical testing of a high-affinity single-chain antibody against (+)-methamphetamine. J Pharmacol Exp Ther. 2008;325:124–133. doi: 10.1124/jpet.107.134395. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Carroll FI, Abraham P, Gong PK, Pidaparthi RR, Blough BE, Che Y, et al. The synthesis of haptens and their use for the development of monoclonal antibodies for treating methamphetamine abuse. J Med Chem. 2009;52:7301–7309. doi: 10.1021/jm901134w. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Miller ML, Moreno AY, Aarde SM, Creehan KM, Vandewater SA, Vaillancourt BD, et al. A Methamphetamine Vaccine Attenuates Methamphetamine-Induced Disruptions in Thermoregulation and Activity in Rats. Biol Psychiatry. 2012:1–8. doi: 10.1016/j.biopsych.2012.09.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Shen XY, Kosten TA, Lopez AY, Kinsey BM, Kosten TR, Orson FM. A vaccine against methamphetamine attenuates its behavioral effects in mice. Drug Alcohol Depend. 2013;129:41–48. doi: 10.1016/j.drugalcdep.2012.09.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Duryee MJ, Bevins RA, Reichel CM, Murray JE, Dong Y, Thiele GM, et al. Immune responses to methamphetamine by active immunization with peptide-based, molecular adjuvant-containing vaccines. Vaccine. 2009;27:2981–2988. doi: 10.1016/j.vaccine.2009.02.105. [DOI] [PubMed] [Google Scholar]
- 17.Pravetoni M, Le Naour M, Harmon TM, Tucker AM, Portoghese PS, Pentel PR. An oxycodone conjugate vaccine elicits drug-specific antibodies that reduce oxycodone distribution to brain and hot-plate analgesia. J Pharmacol Exp Ther. 2012;341:225–232. doi: 10.1124/jpet.111.189506. Epub2012Jan18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Pravetoni M, Raleigh MD, Le Naour M, Tucker AM, Harmon TM, Jones JM, et al. Co- administration of morphine and oxycodone vaccines reduces the distribution of 6-monoacetylmorphine and oxycodone to brain in rats. Vaccine. 2012;30:4617–4624. doi: 10.1016/j.vaccine.2012.04.101. Epub2012May11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Satoskar SD, Keyler DE, LeSage MG, Raphael DE, Ross CA, Pentel PR. Tissue-dependent effects of immunization with a nicotine conjugate vaccine on the distribution of nicotine in rats. Int Immunopharmacol. 2003;3:957–970. doi: 10.1016/S1567-5769(03)00094-8. [DOI] [PubMed] [Google Scholar]
- 20.Kinsey BM, Jackson DC, Orson FM. Anti-drug vaccines to treat substance abuse. Immunol Cell Biol. 2009;87:309–314. doi: 10.1038/icb.2009.17. [DOI] [PubMed] [Google Scholar]
- 21.Kosten TR, Rosen M, Bond J, Settles M, Roberts JSC, Shields J, et al. Human therapeutic cocaine vaccine: safety and immunogenicity. Vaccine. 2002;20:1196–1204. doi: 10.1016/s0264-410x(01)00425-x. [DOI] [PubMed] [Google Scholar]
- 22.Martell BA, Mitchell E, Poling J, Gonsai K, Kosten TR. Vaccine pharmacotherapy for the treatment of cocaine dependence. Biol Psychiatry. 2005;58:158–164. doi: 10.1016/j.biopsych.2005.04.032. [DOI] [PubMed] [Google Scholar]
- 23.Cornuz J, Zwahlen S, Jungi WF, Osterwalder J, Klingler K, van Melle G, et al. A vaccine against nicotine for smoking cessation: a randomized controlled trial. PloS One. 2008;3:e2547. doi: 10.1371/journal.pone.0002547. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Maurer P, Jennings GT, Willers J, Rohner F, Lindman Y, Roubicek K, et al. A therapeutic vaccine for nicotine dependence: preclinical efficacy, and Phase I safety and immunogenicity. Eur J Immunol. 2005;35:2031–2040. doi: 10.1002/eji.200526285. [DOI] [PubMed] [Google Scholar]
- 25.Miller JS, Curtsinger J, Berthold M, Malvey K, Bliss RL, Le CT, et al. Diminished neo-antigen response to keyhole limpet hemocyanin (KLH) vaccines in patients after treatment with chemotherapy or hematopoietic cell transplantation. Clin Immunol. 2005;117:144–151. doi: 10.1016/j.clim.2005.07.005. [DOI] [PubMed] [Google Scholar]
- 26.Carrera MR, Ashley JA, Wirsching P, Koob GF, Janda KD. A second-generation vaccine protects against the psychoactive effects of cocaine. Proc Natl Acad Sci USA. 2001;98:1988–1992. doi: 10.1073/pnas.041610998. Epub2001Feb6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Carrera MR, Meijler MM, Janda KD. Cocaine pharmacology and current pharmacotherapies for its abuse. Bioorg Med Chem. 2004;12:5019–5030. doi: 10.1016/j.bmc.2004.06.018. [DOI] [PubMed] [Google Scholar]
- 28.Li Q-Q, Luo Y-X, Sun C-Y, Xue Y-X, Zhu W-L, Shi H-S, et al. A morphine/heroin vaccine with new hapten design attenuates behavioral effects in rats. J Neurochem. 2011;119:1271–1281. doi: 10.1111/j.1471-4159.2011.07502.x. [DOI] [PubMed] [Google Scholar]
- 29.Kantak KM, Collins SL, Lipman EG, Bond J, Giovanoni K, Fox BS. Evaluation of anti-cocaine antibodies and a cocaine vaccine in a rat self-administration model. Psychopharmacology. 2000;148:251–262. doi: 10.1007/s002130050049. [DOI] [PubMed] [Google Scholar]
- 30.Kantak KM, Collins SL, Bond J, Fox BS. Time course of changes in cocaine self-administration behavior in rats during immunization with the cocaine vaccine IPC-1010. Psychopharmacology. 2001;153:334–340. doi: 10.1007/s002130000555. [DOI] [PubMed] [Google Scholar]
- 31.Moreno AY, Janda KD. Immunopharmacotherapy: vaccination strategies as a treatment for drug abuse and dependence. Pharmacol Biochem Behav. 2009;92:199–205. doi: 10.1016/j.pbb.2009.01.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Hatsukami DK, Jorenby DE, Gonzales D, Rigotti NA, Glover ED, Oncken CA, et al. Immunogenicity and Smoking-Cessation Outcomes for a Novel Nicotine Immunotherapeutic. Clin Pharmacol Ther. 2011;89:392–399. doi: 10.1038/clpt.2010.317. Epub2011Jan26. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Slovin SF, Keding SJ, Ragupathi G. Carbohydrate vaccines as immunotherapy for cancer. Immunol Cell Biol. 2005;83:418–428. doi: 10.1111/j.1440-1711.2005.01350.x. [DOI] [PubMed] [Google Scholar]
- 34.Peterson EC, Gunnell M, Che Y, Goforth RL, Carroll FI, Henry R, et al. Using hapten design to discover therapeutic monoclonal antibodies for treating methamphetamine abuse. J Pharmacol Exp Ther. 2007;322:30–39. doi: 10.1124/jpet.106.117150. [DOI] [PubMed] [Google Scholar]
- 35.Akera T, Cheng VK. A simple method for the determination of affinity and binding site concentration in receptor binding studies. Biochim Biophys Acta. 1977;470:412–423. doi: 10.1016/0005-2736(77)90132-8. [DOI] [PubMed] [Google Scholar]
- 36.Hendrickson H, Laurenzana E, Owens SM. Quantitative determination of total methamphetamine and active metabolites in rat tissue by liquid chromatography with tandem mass spectrometric detection. Aaps J. 2006;8:E709–E717. doi: 10.1208/aapsj080480. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Segal DS, Kuczenski R. Individual differences in responsiveness to single and repeated amphetamine administration: behavioral characteristics and neurochemical correlates. J Pharmacol Exp Ther. 1987;242:917–926. [PubMed] [Google Scholar]
- 38.Proksch JW, Gentry WB, Owens SM. Anti-phencyclidine monoclonal antibodies provide long-term reductions in brain phencyclidine concentrations during chronic phencyclidine administration in rats. J Pharmacol Exp Ther. 2000;292:831–837. [PubMed] [Google Scholar]
- 39.Pitas G, Laurenzana EM, Williams DK, Owens SM, Gentry WB. Anti-phencyclidine monoclonal antibody binding capacity is not the only determinant of effectiveness, disproving the concept that antibody capacity is easily surmounted. Drug Metab Dispos. 2006;34:906–912. doi: 10.1124/dmd.105.005934. [DOI] [PubMed] [Google Scholar]
- 40.Milesi-Hallé A, McMillan DE, Laurenzana EM, Byrnes-Blake KA, Owens SM. Sex differences in (+)-amphetamine- and (+)-methamphetamine-induced behavioral response in male and female Sprague-Dawley rats. Pharmacol Biochem Behav. 2007;86:140–149. doi: 10.1016/j.pbb.2006.12.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Gentry WB, Ghafoor AU, Wessinger WD, Laurenzana EM, Hendrickson HP, Owens SM. (+)-Methamphetamine-induced spontaneous behavior in rats depends on route of (+)METH administration. Pharmacol Biochem Behav. 2004;79:751–760. doi: 10.1016/j.pbb.2004.10.006. [DOI] [PubMed] [Google Scholar]
- 42.Moreno AY, Azar MR, Warren NA, Dickerson TJ, Koob GF, Janda KD. A critical evaluation of a nicotine vaccine within a self-administration behavioral model. Mol Pharm. 2010;7:431–441. doi: 10.1021/mp900213u. [DOI] [PubMed] [Google Scholar]
- 43.Pinto VV, Salanti A, Joergensen LM, Dahlback M, Resende M, Ditlev SB, et al. The effect of adjuvants on the immune response induced by a DBL4varepsilon-ID4 VAR2CSA based Plasmodium falciparum vaccine against placental malaria. Vaccine. 2012;30:572–579. doi: 10.1016/j.vaccine.2011.11.068. Epub2011Nov26. [DOI] [PubMed] [Google Scholar]





