Sandbox Reserved 1474: Difference between revisions
No edit summary |
No edit summary |
||
(15 intermediate revisions by the same user not shown) | |||
Line 94: | Line 94: | ||
===Human-mouse chimeric monoclonal antibody(mAb) Ch-mAb7F9=== | ===Human-mouse chimeric monoclonal antibody(mAb) Ch-mAb7F9=== | ||
IgG mAbs are typically chimeric, humanized, or fully human proteins. The longest t1/2lamdaz values are usually achieved when the antibody does not bind to tissue sites and is not prematurely cleared due to antigenicity. | IgG mAbs are typically chimeric, humanized, or fully human proteins. The longest t1/2lamdaz values are usually achieved when the antibody does not bind to tissue sites and is not prematurely cleared due to antigenicity<ref name="Preclinical characterization of an anti-methamphetamine monoclonal antibody for human use"/> | ||
Ch-mAb7F9, a chimeric mAb is produced as a treatment medication for METH abuse. In vitro, it is shown only binds to (+)METH (KD=6.9nM), (+)AMP( | . | ||
Ch-mAb7F9, a chimeric mAb is produced as a treatment medication for METH abuse based on the murine anti-METH mAb7F9<ref name="Pharmacological effects of two anti-methamphetamine monoclonal antibodies"/>. It is created by preserving mAb7f9 variable region with human IgG2 constant domains<ref name="Preclinical characterization of an anti-methamphetamine monoclonal antibody for human use"/> to minimize the risk of effector function. In vitro, it is shown only binds to (+)METH (KD=6.9nM)<ref name="Preclinical characterization of an anti-methamphetamine monoclonal antibody for human use"/> | |||
, (+)AMP(KI = 350 nM)<ref name="Preclinical characterization of an anti-methamphetamine monoclonal antibody for human use"/> | |||
, (+)MDMA(kI=6.7nM)<ref name="Preclinical characterization of an anti-methamphetamine monoclonal antibody for human use"/> | |||
. | |||
===Preclinical characterization of Ch-mAb7F9 for human use=== | ===Preclinical characterization of Ch-mAb7F9 for human use=== | ||
====Cross reactions in vitro ligand binding studies==== | |||
It did not bind endogenous neurotransmitters or other medications and was not bound by protein C1q, result of the test that was conducted to determine the potential for complement activation, which is an undesired effector function, thus it is unlikely to stimulate in vivo complement-dependent cytotoxicity. <ref name="Preclinical characterization of an anti-methamphetamine monoclonal antibody for human use" /> | |||
Just as mAb7F9, ch-mAb counterpart does not bind any of these compounds except (+)AMP, (+)METH and (+)MDMA well enough to raise clinical effect and in vitro with only one interesting exception: (-)MDMA, which was capable of inhibiting [3H]METH binding at greater than 50% and it was the only ligand with a KI (ocncentration of inhibitor that prevents 50% of the [H3] from binding) less than 1 microMolar among other tested compounds. However, due to Ecstasy is a racemic mixture contains both (+) and (-) MDMA and the ability that mAb7F9 to bind both forms of MDMA may actually improve its utility as a potential treatment for MDMA abuse. | |||
<table><tr><td colspan='2'>Table 2 <ref name="Preclinical characterization of an anti-methamphetamine monoclonal antibody for human use" />Ligands tested in Ch-mAb7F9 cross-reactivity study.<br></td></tr> | |||
<tr id='Related stimulates'><td class="sblockLbl"><b>Related stimulates</b></td><td class="sblockDat"> | |||
(+)-Methamphetamine, | |||
(+)-Amphetamine, | |||
(+)-MDMA, | |||
(-)-MDMA, | |||
(+)-MDA | |||
</td></tr> | |||
<tr id='Neurotransmitters'> | |||
==== | <td class="sblockLbl"><b>neurotransmitters </b></td><td class="sblockDat"> | ||
Dopamine, | |||
== | (-)-Norepinephrine, | ||
(-)-Epinephrine, | |||
Serotonin, | |||
γ-aminobutyric acid, | |||
L-Glutamate | |||
</td></tr> | |||
<tr id='Medications'><td class="sblockLbl"><b>Medications </b></td><td class="sblockDat"> | |||
(+)-Pseudoephedrine, | |||
(+)-Norpseudoephedrine, | |||
(-)-Phenylephrine, | |||
(±)-Ephedrine, | |||
2-Phenylethylamine, | |||
Tyramine, | |||
</td></tr> | |||
<tr id='Drugs of abuse'><td class="sblockLbl"><b>Drugs of abuse </b></td><td class="sblockDat"> | |||
Cocaine, | |||
Morphine, | |||
Phencyclidine | |||
</td></tr> | |||
</table> | </table> | ||
====Isothermal titration calorimetry potency studies==== | |||
Isothermal titration calorimetry analysis of ch-mAb7F9 binding to METH provided thermodynamic and stoichiometry measurements for its potency.<ref name="Preclinical characterization of an anti-methamphetamine monoclonal antibody for human use" /> | |||
<table><tr><td colspan='2'> Antibody thermodynamic values and stoichiometry for target binding<ref name="Preclinical characterization of an anti-methamphetamine monoclonal antibody for human use" />.<br></td></tr> | |||
<tr id='Antibody'><td class="sblockLbl"><b>Antibody </b></td><td class="sblockDat"> Ch-mAb7F9</td></tr> | |||
<tr id='Delta G'><td class="sblockLbl"><b>ΔG (kJ/mol) </b></td><td class="sblockDat"> -43 </td></tr> | |||
<tr id='Delta H'><td class="sblockLbl"><b>ΔH (kJ/mol) </b></td><td class="sblockDat"> -45 </td></tr> | |||
<tr id='-TdeltaS'><td class="sblockLbl"><b>-TΔS (kJ/mol) </b></td><td class="sblockDat">1 </td></tr> | |||
<tr id='Stoichiometry(N)'><td class="sblockLbl"><b>Stoichiometry </b></td><td class="sblockDat">1.89 </td></tr> | |||
</table> | |||
The gibbs free energy change is very similar to other antibodies reported in study. Like others, binding is mostly driven by a favorable enthalpy change that compensates for an almost nonexistent entropy penalty. The stoichiometry is expected to be 2 binding sites per antibody molecule and it is almost completely active. | |||
====Pharmacokinetics studies in rats==== | ====Pharmacokinetics studies in rats==== | ||
METH had little effect on ch-mAb7F9 disposition, ch-mAb7F9 substantially altered METH disposition. <ref name="Preclinical characterization of an anti-methamphetamine monoclonal antibody for human use" /> | METH had little effect on ch-mAb7F9 disposition, ch-mAb7F9 substantially altered METH disposition. Both in vitro and in vivo demonstrated ch-mAb7F9 is pharmacologically similar to its murine counter part<ref name="Preclinical characterization of an anti-methamphetamine monoclonal antibody for human use" />. Ch-mAb decreased the METH Vd by 5 and 25 fold at the 15 and 150mg/kg doses. Although METH t1/2lamdaz is increased 2 to 5 folds due to the decreased ClT to a greater degree, METH elimination was still rapid compared to ch-mAb elimination. (2-7 hrs v.s 10-13 d). | ||
In the current studies, a t1/2lamdaZ of 10-13 days for ch-mAb7F9 in rats was observed. The half life is predicted to be 3 weeks in human roughly 3 folds of that of rat due to the volume of distribution at steady state (Vdss) of IgG in both species is similar yet the clearance time (Clt) in humans is one third of that in rats. | |||
== Human Study == | == Human Study == | ||
===Phase 1 Study: First Human study of a chimeric anti-methamphetamine monoclonal antibody in healthy volunteers=== | ===Phase 1 Study: First Human study of a chimeric anti-methamphetamine monoclonal antibody in healthy volunteers=== | ||
The first study is conducted in healthy 42 volunteers, 10s of which received saline placebo as control group.<ref name="First human study of a chimeric anti-methamphetamine monoclonal antibody in healthy volunteers"/>. Single, escalating doses of ch-mAb7F9 over the range of 0.2 to 20mg/kg (5 dose groups) were administered and followed for 147 d for pharmacokinetic and immugenicity studies<ref name="First human study of a chimeric anti-methamphetamine monoclonal antibody in healthy volunteers"/>. No serious adverse reactions or discontinuations form the study due to adverse events<ref name="First human study of a chimeric anti-methamphetamine monoclonal antibody in healthy volunteers"/>. No trends emerged of adverse events<ref name="First human study of a chimeric anti-methamphetamine monoclonal antibody in healthy volunteers"/>. Half life of 17-19 d in the 3 highest does groups an volume of distribution of 5-6L suggesting antibody is confined primarily to the vascular compartment<ref name="First human study of a chimeric anti-methamphetamine monoclonal antibody in healthy volunteers"/>. | |||
Serum ch-mAb7F9 concentration is plotted and reported for 147 d<ref name="First human study of a chimeric anti-methamphetamine monoclonal antibody in healthy volunteers"/>. | |||
Most common AE include: increased blood creatine phosphokinase, upper respiratory tract infection, decreased hemoglobin, headache, increased aspartate aminotransferase and alanine aminotransferase, proteinuria, decreased white blood cell count, and nasal congestion<ref name="First human study of a chimeric anti-methamphetamine monoclonal antibody in healthy volunteers"/>. AEs considered by the investigator to be related to study medication were limited to single events in the 2mg/kg group <ref name="First human study of a chimeric anti-methamphetamine monoclonal antibody in healthy volunteers"/>and included infusion reaction, bronchospasm, and proteinuria<ref name="First human study of a chimeric anti-methamphetamine monoclonal antibody in healthy volunteers"/>. The 3 Grade 4 AEs(life-threatening) were all elevations in blood creatine phosphokinases levels and were considered unrelated to the ch-mAb and resolved without treatment. 6 events as Grade 3 and 47 grade 2 and 160 events as grade 1 (mild)<ref name="First human study of a chimeric anti-methamphetamine monoclonal antibody in healthy volunteers"/>. | |||
Four(12.5%) of the 32 subjects receiving ch-mAb7F9 were confirmed to have developed a human anti-chimeric antibody response by the end of the study (147 d)<ref name="First human study of a chimeric anti-methamphetamine monoclonal antibody in healthy volunteers"/>;however, this response did not appear to be dose related.<ref name="First human study of a chimeric anti-methamphetamine monoclonal antibody in healthy volunteers " /> | |||
Line 157: | Line 197: | ||
After METH infusion 3.2mg/kg/day<ref name="Development and preclinical testing of a high-affinity single-chain antibody against (+)-methamphetamine"/> by osmotic minipumps which resulted average steady state serum concentration of 25ng/ml after 24h<ref name="Development and preclinical testing of a high-affinity single-chain antibody against (+)-methamphetamine"/>, mice were administered scFv6H4 which has led to drastic increase of serum concentration of METH<ref name="Development and preclinical testing of a high-affinity single-chain antibody against (+)-methamphetamine"/>, determined by liquid chromatography-tandem mass spectrometry as described previously<ref name ="Development of a liquid chromatography-tandem mass spectrometric method for the determination of methamphetamine and amphetamine using small volumes of rat serum"/>. The compare of the first 480 min METH concentration with control group was reported<ref name="Development and preclinical testing of a high-affinity single-chain antibody against (+)-methamphetamine"/>. scFv6H4 was reported stable in serum in vitro yet unstable in Urine in vitro<ref name="Development and preclinical testing of a high-affinity single-chain antibody against (+)-methamphetamine"/>. The concentration of scFv in serum in vivo was determined by SEC<ref name="Development and preclinical testing of a high-affinity single-chain antibody against (+)-methamphetamine"/>, the method has been previously described <ref name ="Pharmacokinetic mechanisms for obtaining high renal coelimination of phencyclidine and a monoclonal antiphencyclidine antigen-binding fragment of immunoglobulin G in the rat"/>. Monomer of scFv was reportd to have been completely eliminated in the serum within the first 30 minutes<ref name="Development and preclinical testing of a high-affinity single-chain antibody against (+)-methamphetamine"/>, yet the multivalent larger proteins persisted for >240 mins corresponding to the reported t1/2lamdaz to be 228 +/- 38 min<ref name="Development and preclinical testing of a high-affinity single-chain antibody against (+)-methamphetamine"/>. It is interesting that it was reported the divalent form did not decrease for the first 10 minutes, as if while it is been eliminated, it is also been formed from the mono scFvs. Pharmacokinetic parameters were reported for mono and multi scFvs respectively<ref name="Development and preclinical testing of a high-affinity single-chain antibody against (+)-methamphetamine"/>. | After METH infusion 3.2mg/kg/day<ref name="Development and preclinical testing of a high-affinity single-chain antibody against (+)-methamphetamine"/> by osmotic minipumps which resulted average steady state serum concentration of 25ng/ml after 24h<ref name="Development and preclinical testing of a high-affinity single-chain antibody against (+)-methamphetamine"/>, mice were administered scFv6H4 which has led to drastic increase of serum concentration of METH<ref name="Development and preclinical testing of a high-affinity single-chain antibody against (+)-methamphetamine"/>, determined by liquid chromatography-tandem mass spectrometry as described previously<ref name ="Development of a liquid chromatography-tandem mass spectrometric method for the determination of methamphetamine and amphetamine using small volumes of rat serum"/>. The compare of the first 480 min METH concentration with control group was reported<ref name="Development and preclinical testing of a high-affinity single-chain antibody against (+)-methamphetamine"/>. scFv6H4 was reported stable in serum in vitro yet unstable in Urine in vitro<ref name="Development and preclinical testing of a high-affinity single-chain antibody against (+)-methamphetamine"/>. The concentration of scFv in serum in vivo was determined by SEC<ref name="Development and preclinical testing of a high-affinity single-chain antibody against (+)-methamphetamine"/>, the method has been previously described <ref name ="Pharmacokinetic mechanisms for obtaining high renal coelimination of phencyclidine and a monoclonal antiphencyclidine antigen-binding fragment of immunoglobulin G in the rat"/>. Monomer of scFv was reportd to have been completely eliminated in the serum within the first 30 minutes<ref name="Development and preclinical testing of a high-affinity single-chain antibody against (+)-methamphetamine"/>, yet the multivalent larger proteins persisted for >240 mins corresponding to the reported t1/2lamdaz to be 228 +/- 38 min<ref name="Development and preclinical testing of a high-affinity single-chain antibody against (+)-methamphetamine"/>. It is interesting that it was reported the divalent form did not decrease for the first 10 minutes, as if while it is been eliminated, it is also been formed from the mono scFvs. Pharmacokinetic parameters were reported for mono and multi scFvs respectively<ref name="Development and preclinical testing of a high-affinity single-chain antibody against (+)-methamphetamine"/>. | ||
Line 177: | Line 214: | ||
|RESOURCES=<span class='plainlinks'>[http://oca.weizmann.ac.il/oca-docs/fgij/fg.htm?mol=4lar FirstGlance], [http://oca.weizmann.ac.il/oca-bin/ocaids?id=4lar OCA], [http://pdbe.org/4lar PDBe], [http://www.rcsb.org/pdb/explore.do?structureId=4lar RCSB], [http://www.ebi.ac.uk/pdbsum/4lar PDBsum], [http://prosat.h-its.org/prosat/prosatexe?pdbcode=4lar ProSAT]</span> | |RESOURCES=<span class='plainlinks'>[http://oca.weizmann.ac.il/oca-docs/fgij/fg.htm?mol=4lar FirstGlance], [http://oca.weizmann.ac.il/oca-bin/ocaids?id=4lar OCA], [http://pdbe.org/4lar PDBe], [http://www.rcsb.org/pdb/explore.do?structureId=4lar RCSB], [http://www.ebi.ac.uk/pdbsum/4lar PDBsum], [http://prosat.h-its.org/prosat/prosatexe?pdbcode=4lar ProSAT]</span> | ||
}} | }} | ||
{{ STRUCTURE | {{ STRUCTURE | ||
|PDB=3gkz | |PDB=3gkz | ||
Line 191: | Line 225: | ||
|RESOURCES=<span class='plainlinks'>[http://oca.weizmann.ac.il/oca-docs/fgij/fg.htm?mol=4lar FirstGlance], [http://oca.weizmann.ac.il/oca-bin/ocaids?id=4lar OCA], [http://pdbe.org/4lar PDBe], [http://www.rcsb.org/pdb/explore.do?structureId=4lar RCSB], [http://www.ebi.ac.uk/pdbsum/4lar PDBsum], [http://prosat.h-its.org/prosat/prosatexe?pdbcode=4lar ProSAT]</span> | |RESOURCES=<span class='plainlinks'>[http://oca.weizmann.ac.il/oca-docs/fgij/fg.htm?mol=4lar FirstGlance], [http://oca.weizmann.ac.il/oca-bin/ocaids?id=4lar OCA], [http://pdbe.org/4lar PDBe], [http://www.rcsb.org/pdb/explore.do?structureId=4lar RCSB], [http://www.ebi.ac.uk/pdbsum/4lar PDBsum], [http://prosat.h-its.org/prosat/prosatexe?pdbcode=4lar ProSAT]</span> | ||
}} | }} | ||
===Aromatic-Aromatic Interaction: A Mechanism of Protein Structure Stabilization=== | ===Aromatic-Aromatic Interaction: A Mechanism of Protein Structure Stabilization=== | ||
The entrance of the binding pocket is lined with seven amino residues, one residue from each of them H1, H2, and H3 loops, 3 from the L3 loop, one from the beta-strand-3c of the heavy chain. These aromatic residues form a hydrophobic barrel around the aromatic portion of METH. | The entrance of the binding pocket is lined with seven amino residues, one residue from each of them H1, H2, and H3 loops, 3 from the L3 loop, one from the beta-strand-3c of the heavy chain. These aromatic residues form a hydrophobic barrel around the aromatic portion of METH<ref name="Crystal structures of a therapeutic single chain antibody in complex with two drugs of abuse-methamphetamine and 3,4-methylenedioxymethamphetamine"/>. | ||
===Hydrophilic interactions of METH=== | ===Hydrophilic interactions of METH=== | ||
The protonated secondary amine of METH anchors the ligand deep in the pocket. There is a salt bridge between the cationic nitrogen of METH and the carboxyl oxygen of Glutamate. In addition, the cationic nitrogen forms a hydrogen bond to Hisdine of the light chain<ref name="Crystal structures of a therapeutic single chain antibody in complex with two drugs of abuse-methamphetamine and 3,4-methylenedioxymethamphetamine"/>. | |||
===Water molecules in the binding cavity=== | ===Water molecules in the binding cavity=== | ||
two water molecules are in the pocket stablized by hydrogen bonding bewteen and with the side chain residues<ref name="Crystal structures of a therapeutic single chain antibody in complex with two drugs of abuse-methamphetamine and 3,4-methylenedioxymethamphetamine"/>. | |||
{{ STRUCTURE | {{ STRUCTURE | ||
Line 220: | Line 247: | ||
}} | }} | ||
Latest revision as of 20:44, 18 December 2018
Immunotherapy for treating Methamphetamine AbuseImmunotherapy for treating Methamphetamine Abuse
IntroductionIntroduction
Traditional treatment for addiction and its challengeTraditional treatment for addiction and its challenge
(+)-Methamphetamine (METH) abuse are mediated via multiple neurotransmitter sites. Medications that target single site of action (e.g., the dopamine transporter) have proven ineffective and in some cases addictive. No medication has been approved by the Food and Drug Administration for the treatment of methamphetamine abuse. Traditional treatment regimes begin with medical detox, during which the substance is slowly weaned out of the user’s body. Because there are no specific pharmacological therapies for excessive methamphetamine use, supportive care is aimed to manage symptoms instead of removing causative agent. Patients are left susceptible to potential neurotoxic effects of methamphetamine[1]. Post detox long-term behavioral modification programs aims at reconstructing positive behaviors and learning new and drug-free ways to cope with life stressors that trigger the desire to use meth, however, to protect patients against relapse remains a profound challenge and there is great need for outcome-based treatment research. Anti-METH mAb reside in the blood and extracellular fluid compartments. It could act as a pharmacokinetic antagonist and alter distribution of METH by high affinity binding and reduce its amount and delivery to its site of action in the brain and other vulnerable organs (heart), therefore favorably alter its metabolism and eliminate the drug.[1]. A monoclonal antibody (mAb) with high affinity binding to METH and a long acting duration (2-3 weeks) could provide a novel therapeutic strategy for patients who have serious problems with compliance [2] therefore will increase the probability of a success treatment.
Two major approaches to developing drug-specific immunotherapies: active and passive immunizationTwo major approaches to developing drug-specific immunotherapies: active and passive immunization
Active immunizationActive immunization
Active immunization involves conjugating a drug-like hapten to a carrier protein and using traditional immunization approaches to generate a specific immune response over time[3].Active vaccines against METH have shown potential for reducing CNS effects such as horizontal locomotion and self administration in animal models[4]. Vaccine has shown to attenuate methamphetamine-Induced disruptions in thermoregulation and activity in Rats[5]. It has also shown to protect rats from METH-induced impairment of behavioral responding for food[6].
Furthermore, it is important to determine whether or not drug use would interfere with antibody generation during the immunization period because it is unlikely that most drug abusers will abstain from drug abuse during the recovery process. In animals immunized against (+)METH, anti-(+)METH antibody titers and affinity for (+)METH were the same, regardless of whether or not they were repeatedly challenged with (+)METH [7].This indicates chronic drug use would less likely to interfere with antibody generation during active immunization.
Passive immunotherapyPassive immunotherapy
Passive immunotherapy involves treatment with carefully selected, preformed monoclonal antibodies or antibody fragments against a drug of abuse[3] .These mAbs are generated by first vaccinating a host animal and then creating mAb-secreting hybridoma cell lines, or, alternatively, by recombinant DNA methods that utilize phage, yeast, or ribosome display[3]. If needed, these mAbs are converted to a human-compatible form(e.g., chimeric, humanized, or fully human immunoglobulin)[3].
There are currently two forms of passive mAbs in preclinical testing: a long-acting intact immunoglobulin G (IgG) (150kDa) form for treating addiction and overdose, and an extremely short-acting single chain variable fragment (scFv; 27kDa) for treatment of overdose.[3] The long half-life of IgG is due partly to the ability of the constant region of the antibody to be salvaged from catabolism by the neonatal Fc receptor (FcRn)[8].
Combining active immunization with monoclonal antibody therapyCombining active immunization with monoclonal antibody therapy
An anti-METH mAb could be used in combination with a METH-conjugate vaccine (MCV). 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).[9]
Materials and methodsMaterials and methods
ChemicalsChemicals
[(+)-3H]METH and [(+/-)-3H]AMP were used for synthesizing haptens.
Synthesis of METH-like haptensSynthesis of METH-like haptens
Because drugs of abuse are too small to generate an immune response on their own, a critical step toward making an effective drug-specific vaccine is to synthesize a hapten that maintains the chemical and structural properties of the original drug and has an added, carefully placed chemical linker with a distal moiety of a functional group that can be easily conjugated to a larger carrier antigenic protein.[3]. The complete synthesis (+)-METH P6 was reported 2001[7] . The chemical structures of other stereospecific (+) haptens were reported 2007[10].
It was reported that 1)linkers located distal to the chiral center of the small drug molecule favors generation of stereospecific antibodies 2)longer flexible linker broadens recognition of meth like molecules 3) spacers equal to or greater than six atoms produce higher affinity mAb<>.
Antigen synthesisAntigen synthesis
Carrier proteins, such as keyhole limpet hemocyanin or modified bacterial toxoids that are safe for use in humans. Ovalbumin, bovine serum albumin, or many others are used in animal models[3]. Bovine serum albumin was reported to covalently bound to (+)-METH P6 and ovalbumin to (+)METH MO10 to produce mAb6H4 and mAb4G9 respectively[10] by two-step carbodiimide procedure[11] which forms a peptide bond between the carboxyl group of the hapten linker arm and free amino groups of lysine side chains in the respective proteins[10].
At the end, all antigens were purified on a gel filtration column in a phosphate-buffered saline after dialysis[1].
Immunization, Screening, and Hybridoma GenerationImmunization, Screening, and Hybridoma Generation
Initial subcutaneous immunization of 100 micrograms of the (+)-METH P6 antigen emulsified with TiterMax adjuvant was followed with monthly boosts of 50 microgram dosage. For all other antigen immunizations, Freund's adjuvants were used[10] and dosage and boost intervals have also been reported[10]. Serum samples were taken via tail bleed periodically to measure IgG titers by enzyme-linked immunosorbent essay (ELISA)[10]. Wells were coated with the original hapten that conjugated to a different protein to avoid selecting carrier protein-reactive antibodies[10]. The mouse with the highest anti-METH serum titer was chosen for monoclonal antibody production[1].(cell line [10]). After fusion of mouse B-cells with a myeloma cell line, hybridomas which has both the antibody-producing ability of the mouse B-cell and exaggerated longevity and reproductivity of the fusion partner myeloma were identified by ELISA and sub cloned to monoclonality[12]. To generate large amounts of monoclonal antibody, mice were injected with hybridoma cells and ascites fluid that contained high concentration of IgG was collected after next several weeks[12].
Mouse-hybridoma isotyping kit is used to decide the IgG isotype and light chain identity[10]. In the case of mAb6H4, it was determined to be immunoglobin G1 WITH A kappa light chain[1].
Production, purification and formulationProduction, purification and formulation
To produce gram quantities for pharmacokinetics studies, the hybridoma cell line was grown in a Cell-Pharm System 2500 hollow-fiber bioreactor [1] and the similar method has been previously described[13]or to be produced in a Biostat B 10 liter bioreactor[10]. Antibody purification was performed with an Index-100 cation exchange column[1]. Similar method has been previously described [14],or they were purified by affinity chromatography using protein G-Sepharose[10], or both[10].
After purification, all antibodies were concentrated, and buffer was exchanged into 15mM sodium phosphate containing 150mM sodium chloride[10] as previously described[15].
Pharmacokinetics, pharmacodynamics and metabolism of anti-METH antibody mAb6H4Pharmacokinetics, pharmacodynamics and metabolism of anti-METH antibody mAb6H4
For a mAb therapeutic to have the broadest medical applicability, it should have high affinity and also specificity for members of this drug class. [(+)-METH,(+)-AMP, and (+)-MDMA].First, these stimulant drugs have similar or overlapping effects. In particular, (+)-AMP is both a pharmacologically active metabolite of (+)-METH and a frequently used drug of abuse that could be substituted for (+)-METH. In addition, the minus isomers of these drugs could potentially be purposely taken by drug abusers to neutralize mAb medications with high affinity binding for both plus and minus stereoisomers. In a related way, there are many structurally similar compounds like ephedrine and pseudoephedrine that could be used to lessen the efficacy of mAb therapies if the mAb is not highly specific for (+)-METH-like structures.[10]
Radioimmunoassay determination of mAb6H4 cross-reactivity and dissociation constantsRadioimmunoassay determination of mAb6H4 cross-reactivity and dissociation constants
Screening for anti(+)METH IgG response was conducted by radioimmunoassay (RIA)[10] in which radio labelled [(+)-3H] METH is competing with unlabelled (+)-METH and (+)-AMP for the binding site of Abs in a manner similar to what has been previously described[16]. An IC50 value was determined for each compound after fitting a sigmoidal curve to the data points[1][10].
Anti-(+)METH mAb6H4 was reported with Kd = 11 NM [1], having <.01% cross-reactivity with almost all compounds tested except for (+)-methylenedioxymethamphetamine (MDMA) which has just slightly higher relative affinity than METH (9 nM to 11nM)[1]. Besides (+)METH and (+)MDMA, the tested chemicals also include: (-)-METH, (+/-)-AMP,(-)-MDMA,4-OH METH, Pseudoephedrine, Ephedrine, Dopamine, Norepinephrine, Serotonin, Epinephrine.[1]It was also reported stereospecific[1], having an approximately 100 times higher relative affinity for the plus form than the minus forms of these substances[1]. No significant cross activity has been observed in the test for other compounds[1] including methylenedioxyamphetamine, (+)-norpseudoephedrine, L-phenylephrine, (+)-phenylpropanolamine, beta-phenylethylamine, and tyramine[1].
Effect of mAb6H4 on METH and AMP pharmacokineticsEffect of mAb6H4 on METH and AMP pharmacokinetics
the t1/2lamdaZ of mAb6H4 is reported to be around 8 days[1], and the METH induced behavior effects on locomotor effects is within 400 min after METH administration for the maximum dosage (3.0mg/kg) reported[1]. Thus to compare the disposition of drugs with or without mAb6H4, AUC 4.5h 38min <[1]>was used because it was not possible to conduct complete pharmacokinetic profile for 8 days.
For METH doses of 0.3mg/kg [1]and 1 mg/kg [1]in rats, administration of mAb6H4 has led to significantly higher serum concentrations corresponding to significantly lower brain concentration of METH[1] during the time frame with the drug dose was less than the mAb6H4 binding capacity[1]. It was obvious that mAb6H4 administration initially caused a rapid efflux of METH from the brain due to its high affinity[1], however, compared to METH concentration in the brain without mAb6H4, after 4.5 hours, there seems to be a very slight rebound[1] of the concentration in the brain. The reason is unknown, and could be explained as slower redistribution of the drug from other tissues[1]. Yet, however, at this point, even with the rebound, both the METH concentration in control and in the animals administered with mAb6H4 are well below the threshold associated with increased locomotor activity[1].
mAb6H4 also appeared to have more mild effect to AMP[1] because it had little cross activity with AMP in vitro. It was explained possibility because increased the amounts of METH in the serum available for metabolism[1].
However, for METH dose of 3.0mg/kg[1], which was greater than the mAb6H4 binding capacity, METH induced locomotor effects appeared to be increased[1]. Although there are possible explanations, the reason remained to be understood<>. The idea that mAb may slow the input of METH while prolonging exposure is proposed[1].
Clinical developmentClinical development
Pharmacological effects of anti-methamphetamine monoclonal antibodies mAb4G9Pharmacological effects of anti-methamphetamine monoclonal antibodies mAb4G9
After screening for more than 25000[10] potential hybridoma cell lines for mAb production, mAb with the most favorable immunochemical characteristics were extensively studied. Also the sequence features in each mAb variable regions were analyzed. A high degree of diversity in both compostition and length of CDRs are revealed[10]. Although comparisons of CDR sequences are important, differences can be attributed to differences in germ-line sequences of particular V-region genes and to somatic mutation within the CDRs of these V-region genes[10]. After analyzing sequence genes, each antibody was found unique and not clonal[10]. That is, rather than coming from one germline gene arrangement early in B cell development, they resulted from unique V(D)J recombination events. Thus, no clear pattern of response was found[10].
A common feature is a conserved proline[10] at position 95 or 95a of all CDR L3 regions[10] because of their ability to form 'hinges", except one case replaced with serine[10]. And this residue was immediately followed by a hydrophobic amino acid or an aromatic residue which could be interacting with phenyl ring of (+)METH[10].
mAb4G9, attributing to the design of hapten (+)METH MO10[10], was the only[10] mAb to significantly cross-react with (+)AMP. In order to better understand its affinity for AMP, RIA analysis using [(+)-3H]AMP was conducted in addition to RIA analysis using [(+)-3H]METH[10]. It was reported, the affinity for mAb4G9 for AMP is 51nM[10], demonstrating this molecule actually has the same KD value for (+)AMP and (+)METH[10].
Human-mouse chimeric monoclonal antibody(mAb) Ch-mAb7F9Human-mouse chimeric monoclonal antibody(mAb) Ch-mAb7F9
IgG mAbs are typically chimeric, humanized, or fully human proteins. The longest t1/2lamdaz values are usually achieved when the antibody does not bind to tissue sites and is not prematurely cleared due to antigenicity[2] . Ch-mAb7F9, a chimeric mAb is produced as a treatment medication for METH abuse based on the murine anti-METH mAb7F9[17]. It is created by preserving mAb7f9 variable region with human IgG2 constant domains[2] to minimize the risk of effector function. In vitro, it is shown only binds to (+)METH (KD=6.9nM)[2] , (+)AMP(KI = 350 nM)[2] , (+)MDMA(kI=6.7nM)[2] .
Preclinical characterization of Ch-mAb7F9 for human usePreclinical characterization of Ch-mAb7F9 for human use
Cross reactions in vitro ligand binding studiesCross reactions in vitro ligand binding studies
It did not bind endogenous neurotransmitters or other medications and was not bound by protein C1q, result of the test that was conducted to determine the potential for complement activation, which is an undesired effector function, thus it is unlikely to stimulate in vivo complement-dependent cytotoxicity. [2]
Just as mAb7F9, ch-mAb counterpart does not bind any of these compounds except (+)AMP, (+)METH and (+)MDMA well enough to raise clinical effect and in vitro with only one interesting exception: (-)MDMA, which was capable of inhibiting [3H]METH binding at greater than 50% and it was the only ligand with a KI (ocncentration of inhibitor that prevents 50% of the [H3] from binding) less than 1 microMolar among other tested compounds. However, due to Ecstasy is a racemic mixture contains both (+) and (-) MDMA and the ability that mAb7F9 to bind both forms of MDMA may actually improve its utility as a potential treatment for MDMA abuse.
Table 2 [2]Ligands tested in Ch-mAb7F9 cross-reactivity study. | |
Related stimulates |
(+)-Methamphetamine, (+)-Amphetamine, (+)-MDMA, (-)-MDMA, (+)-MDA |
neurotransmitters |
Dopamine, (-)-Norepinephrine, (-)-Epinephrine, Serotonin, γ-aminobutyric acid, L-Glutamate |
Medications |
(+)-Pseudoephedrine, (+)-Norpseudoephedrine, (-)-Phenylephrine, (±)-Ephedrine, 2-Phenylethylamine, Tyramine, |
Drugs of abuse |
Cocaine, Morphine, Phencyclidine |
Isothermal titration calorimetry potency studiesIsothermal titration calorimetry potency studies
Isothermal titration calorimetry analysis of ch-mAb7F9 binding to METH provided thermodynamic and stoichiometry measurements for its potency.[2]
Antibody thermodynamic values and stoichiometry for target binding[2]. | |
Antibody | Ch-mAb7F9 |
ΔG (kJ/mol) | -43 |
ΔH (kJ/mol) | -45 |
-TΔS (kJ/mol) | 1 |
Stoichiometry | 1.89 |
The gibbs free energy change is very similar to other antibodies reported in study. Like others, binding is mostly driven by a favorable enthalpy change that compensates for an almost nonexistent entropy penalty. The stoichiometry is expected to be 2 binding sites per antibody molecule and it is almost completely active.
Pharmacokinetics studies in ratsPharmacokinetics studies in rats
METH had little effect on ch-mAb7F9 disposition, ch-mAb7F9 substantially altered METH disposition. Both in vitro and in vivo demonstrated ch-mAb7F9 is pharmacologically similar to its murine counter part[2]. Ch-mAb decreased the METH Vd by 5 and 25 fold at the 15 and 150mg/kg doses. Although METH t1/2lamdaz is increased 2 to 5 folds due to the decreased ClT to a greater degree, METH elimination was still rapid compared to ch-mAb elimination. (2-7 hrs v.s 10-13 d). In the current studies, a t1/2lamdaZ of 10-13 days for ch-mAb7F9 in rats was observed. The half life is predicted to be 3 weeks in human roughly 3 folds of that of rat due to the volume of distribution at steady state (Vdss) of IgG in both species is similar yet the clearance time (Clt) in humans is one third of that in rats.
Human StudyHuman Study
Phase 1 Study: First Human study of a chimeric anti-methamphetamine monoclonal antibody in healthy volunteersPhase 1 Study: First Human study of a chimeric anti-methamphetamine monoclonal antibody in healthy volunteers
The first study is conducted in healthy 42 volunteers, 10s of which received saline placebo as control group.[18]. Single, escalating doses of ch-mAb7F9 over the range of 0.2 to 20mg/kg (5 dose groups) were administered and followed for 147 d for pharmacokinetic and immugenicity studies[18]. No serious adverse reactions or discontinuations form the study due to adverse events[18]. No trends emerged of adverse events[18]. Half life of 17-19 d in the 3 highest does groups an volume of distribution of 5-6L suggesting antibody is confined primarily to the vascular compartment[18]. Serum ch-mAb7F9 concentration is plotted and reported for 147 d[18]. Most common AE include: increased blood creatine phosphokinase, upper respiratory tract infection, decreased hemoglobin, headache, increased aspartate aminotransferase and alanine aminotransferase, proteinuria, decreased white blood cell count, and nasal congestion[18]. AEs considered by the investigator to be related to study medication were limited to single events in the 2mg/kg group [18]and included infusion reaction, bronchospasm, and proteinuria[18]. The 3 Grade 4 AEs(life-threatening) were all elevations in blood creatine phosphokinases levels and were considered unrelated to the ch-mAb and resolved without treatment. 6 events as Grade 3 and 47 grade 2 and 160 events as grade 1 (mild)[18]. Four(12.5%) of the 32 subjects receiving ch-mAb7F9 were confirmed to have developed a human anti-chimeric antibody response by the end of the study (147 d)[18];however, this response did not appear to be dose related.[18]
Development and preclinical testing of a high-affinity single-chain antibody against (+)-methamphetamineDevelopment and preclinical testing of a high-affinity single-chain antibody against (+)-methamphetamine
when a short duration of action and greater extravascular penetration are needed, a significantly smaller fragment like Fab or scFv is used.() The genetic re-engineering of mAb6H4 IgG into scFv6H4 changed the protein from an ~150kDa protein with two anti-METH binding sites to an ~27.4kDa protein with one METH binding site achieved by a 15 amino acid linker to join the two separate gene product: VL and VH. A His6 affinity tag was encoded at the carboxyl terminus to aid in protein purification.
Materials and MethodsMaterials and Methods
cDNA Cloning and sequencing of mAbcDNA Cloning and sequencing of mAb
The heavy and light chains cDNA of the mAb were cloned by reverse transcription-polymerase chain reaction[19], and then they were amplified and ligated into the cloning vector[19]. The resulting plasmids of all mAb cloning was transformed into E. coli strain and then sequenced and submitted to Genbank[19].
Sub-cloning and Large-Scale ExpressionSub-cloning and Large-Scale Expression
After the sequence confirmation, the plasmid was linearized with SacI [19] and the coding sequence of scFv6H4 was transformed to P.pastoris strain(yeast) by electroporation[19]. It is expressed in yeast colony which exhibited high zeocin resistance[19].
Production and purificationProduction and purification
Large scale fermentation was done in batches in a 10 liter working volume biostatB bioreactor to express scFv6H4 in large scale for in vivo pharmacokinetic studies[19]. ScFv was purified from the supernantant by metal affinity chromatography and metal affinity column[19] in a naturally occurring mixture of monomer (~75%) amd dimer (~25%)[19] analyzed with SEC.
Determination of Kd Values using bead-based radioimmunoassayDetermination of Kd Values using bead-based radioimmunoassay
Changes have been made to RIA using TALON beads which binds to the His6 tag at the carboxyl terminus of the VL region of scFv6H4[19], which will orient the binding site of METH distal to the beads, allowing unhindered access to METH[19]. Standard curve was constructed with [H3]METH competing with unlabeled METH. The KD was reported to be 10nM nearly identical to parent IgG mAb6H4 11nM[19]. It was reported just like parent mAb6H4, there is no cross reactivity with related compounds[19] including pseudo-ephedrine, norepinephrine, dopamine, and serotonin, even at 100 microMolar[19].
Pharmacokinetic Studies of METH and scFv6H4 in RatsPharmacokinetic Studies of METH and scFv6H4 in Rats
After METH infusion 3.2mg/kg/day[19] by osmotic minipumps which resulted average steady state serum concentration of 25ng/ml after 24h[19], mice were administered scFv6H4 which has led to drastic increase of serum concentration of METH[19], determined by liquid chromatography-tandem mass spectrometry as described previously[20]. The compare of the first 480 min METH concentration with control group was reported[19]. scFv6H4 was reported stable in serum in vitro yet unstable in Urine in vitro[19]. The concentration of scFv in serum in vivo was determined by SEC[19], the method has been previously described [21]. Monomer of scFv was reportd to have been completely eliminated in the serum within the first 30 minutes[19], yet the multivalent larger proteins persisted for >240 mins corresponding to the reported t1/2lamdaz to be 228 +/- 38 min[19]. It is interesting that it was reported the divalent form did not decrease for the first 10 minutes, as if while it is been eliminated, it is also been formed from the mono scFvs. Pharmacokinetic parameters were reported for mono and multi scFvs respectively[19].
The very small molecular size(27k Da) of scFv monomers leads to rapid clearance (40) mins. scFv6H4 was engineered from mAb6H4. Monomer was reported to be quickly cleared or converted to multivalent forms with an apparent t1/2lamdaz of 5.8 min while multivalent forms showed a much longer t1/2lamdaz (228min). Multimers instead of monomers were considered the cause for the prolonged redistribution of METH into the serum.
Structural highlights from crystal structure of scFv6H4Structural highlights from crystal structure of scFv6H4
| |||||||
scFv6H4 free form | |||||||
---|---|---|---|---|---|---|---|
4laq | |||||||
Ligands: | , , | ||||||
Gene: | IgG (LK3 transgenic mice) | ||||||
Activity: | |||||||
Related: | 4lar, 3gkz, 4las
| ||||||
Resources: | FirstGlance, OCA, PDBe, RCSB, PDBsum, ProSAT | ||||||
Coordinates: | save as pdb, mmCIF, xml |
| |||||||
scFv6H4:(+)METH complex | |||||||
---|---|---|---|---|---|---|---|
3gkz | |||||||
Ligands: | |||||||
Gene: | IgG (LK3 transgenic mice) | ||||||
Activity: | |||||||
Related: | 4lar, 4laq, 4las
| ||||||
Resources: | FirstGlance, OCA, PDBe, RCSB, PDBsum, ProSAT | ||||||
Coordinates: | save as pdb, mmCIF, xml |
Aromatic-Aromatic Interaction: A Mechanism of Protein Structure StabilizationAromatic-Aromatic Interaction: A Mechanism of Protein Structure Stabilization
The entrance of the binding pocket is lined with seven amino residues, one residue from each of them H1, H2, and H3 loops, 3 from the L3 loop, one from the beta-strand-3c of the heavy chain. These aromatic residues form a hydrophobic barrel around the aromatic portion of METH[22].
Hydrophilic interactions of METHHydrophilic interactions of METH
The protonated secondary amine of METH anchors the ligand deep in the pocket. There is a salt bridge between the cationic nitrogen of METH and the carboxyl oxygen of Glutamate. In addition, the cationic nitrogen forms a hydrogen bond to Hisdine of the light chain[22].
Water molecules in the binding cavityWater molecules in the binding cavity
two water molecules are in the pocket stablized by hydrogen bonding bewteen and with the side chain residues[22].
| |||||||
scFv6H4:(+)AMP complex | |||||||
---|---|---|---|---|---|---|---|
4lar | |||||||
Ligands: | , | ||||||
Gene: | IgG (LK3 transgenic mice) | ||||||
Activity: | , | ||||||
Related: | 3gkz, 4laq, 4las
| ||||||
Resources: | FirstGlance, OCA, PDBe, RCSB, PDBsum, ProSAT | ||||||
Coordinates: | save as pdb, mmCIF, xml |
ReferencesReferences
[23] [11] [16] [12] [1] [7] [13] [14] [15] [21] [20]
[3] [8] [24] [25] [10] [19] [26] [22] [27] [5] [6] [28] [29] [17] [2] [9] [18] [4] [30] [31]
- ↑ 1.00 1.01 1.02 1.03 1.04 1.05 1.06 1.07 1.08 1.09 1.10 1.11 1.12 1.13 1.14 1.15 1.16 1.17 1.18 1.19 1.20 1.21 1.22 1.23 1.24 1.25 1.26 1.27 1.28 1.29 1.30 1.31 DOI:/10.1016/S0014-2999(03)01313-X
- ↑ 2.00 2.01 2.02 2.03 2.04 2.05 2.06 2.07 2.08 2.09 2.10 2.11 Stevens MW, Tawney RL, West CM, Kight AD, Henry RL, Owens SM, Gentry WB. Preclinical characterization of an anti-methamphetamine monoclonal antibody for human use. MAbs. 2014 Mar-Apr;6(2):547-55. doi: 10.4161/mabs.27620. Epub 2013 Dec 23. PMID:24492290 doi:http://dx.doi.org/10.4161/mabs.27620
- ↑ 3.0 3.1 3.2 3.3 3.4 3.5 3.6 3.7 Peterson EC, Owens SM. Designing immunotherapies to thwart drug abuse. Mol Interv. 2009 Jun;9(3):119-24. doi: 10.1124/mi.9.3.5. PMID:19592672 doi:http://dx.doi.org/10.1124/mi.9.3.5
- ↑ 4.0 4.1 Miller ML, Aarde SM, Moreno AY, Creehan KM, Janda KD, Taffe MA. Effects of active anti-methamphetamine vaccination on intravenous self-administration in rats. Drug Alcohol Depend. 2015 Aug 1;153:29-36. doi: 10.1016/j.drugalcdep.2015.06.014., Epub 2015 Jun 19. PMID:26118833 doi:http://dx.doi.org/10.1016/j.drugalcdep.2015.06.014
- ↑ 5.0 5.1 Miller ML, Moreno AY, Aarde SM, Creehan KM, Vandewater SA, Vaillancourt BD, Wright MJ Jr, Janda KD, Taffe MA. A methamphetamine vaccine attenuates methamphetamine-induced disruptions in thermoregulation and activity in rats. Biol Psychiatry. 2013 Apr 15;73(8):721-8. doi: 10.1016/j.biopsych.2012.09.010., Epub 2012 Oct 23. PMID:23098894 doi:http://dx.doi.org/10.1016/j.biopsych.2012.09.010
- ↑ 6.0 6.1 Ruedi-Bettschen D, Wood SL, Gunnell MG, West CM, Pidaparthi RR, Carroll FI, Blough BE, Owens SM. Vaccination protects rats from methamphetamine-induced impairment of behavioral responding for food. Vaccine. 2013 Sep 23;31(41):4596-602. doi: 10.1016/j.vaccine.2013.07.038. Epub, 2013 Jul 29. PMID:23906885 doi:http://dx.doi.org/10.1016/j.vaccine.2013.07.038
- ↑ 7.0 7.1 7.2 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 Feb;1(2):329-38. PMID:11360933
- ↑ 8.0 8.1 Lobo ED, Hansen RJ, Balthasar JP. Antibody pharmacokinetics and pharmacodynamics. J Pharm Sci. 2004 Nov;93(11):2645-68. doi: 10.1002/jps.20178. PMID:15389672 doi:http://dx.doi.org/10.1002/jps.20178
- ↑ 9.0 9.1 Hambuchen MD, Carroll FI, Ruedi-Bettschen D, Hendrickson HP, Hennings LJ, Blough BE, Brieaddy LE, Pidaparthi RR, Owens SM. Combining Active Immunization with Monoclonal Antibody Therapy To Facilitate Early Initiation of a Long-Acting Anti-Methamphetamine Antibody Response. J Med Chem. 2015 Jun 11;58(11):4665-77. doi: 10.1021/acs.jmedchem.5b00220. Epub, 2015 May 22. PMID:25973614 doi:http://dx.doi.org/10.1021/acs.jmedchem.5b00220
- ↑ 10.00 10.01 10.02 10.03 10.04 10.05 10.06 10.07 10.08 10.09 10.10 10.11 10.12 10.13 10.14 10.15 10.16 10.17 10.18 10.19 10.20 10.21 10.22 10.23 10.24 10.25 10.26 10.27 10.28 10.29 10.30 Peterson EC, Gunnell M, Che Y, Goforth RL, Carroll FI, Henry R, Liu H, Owens SM. Using hapten design to discover therapeutic monoclonal antibodies for treating methamphetamine abuse. J Pharmacol Exp Ther. 2007 Jul;322(1):30-9. doi: 10.1124/jpet.106.117150. Epub, 2007 Apr 23. PMID:17452421 doi:http://dx.doi.org/10.1124/jpet.106.117150
- ↑ 11.0 11.1 Davis MT, Preston JF. A simple modified carbodiimide method for conjugation of small-molecular-weight compounds to immunoglobulin G with minimal protein crosslinking. Anal Biochem. 1981 Sep 15;116(2):402-7. PMID:7316174
- ↑ 12.0 12.1 12.2 Valentine JL, Arnold LW, Owens SM. Anti-phencyclidine monoclonal Fab fragments markedly alter phencyclidine pharmacokinetics in rats. J Pharmacol Exp Ther. 1994 Jun;269(3):1079-85. PMID:8014850
- ↑ 13.0 13.1 Valentine JL, Mayersohn M, Wessinger WD, Arnold LW, Owens SM. Antiphencyclidine monoclonal Fab fragments reverse phencyclidine-induced behavioral effects and ataxia in rats. J Pharmacol Exp Ther. 1996 Aug;278(2):709-16. PMID:8768722
- ↑ 14.0 14.1 Hardin JS, Wessinger WD, Proksch JW, Owens SM. Pharmacodynamics of a monoclonal antiphencyclidine Fab with broad selectivity for phencyclidine-like drugs. J Pharmacol Exp Ther. 1998 Jun;285(3):1113-22. PMID:9618414
- ↑ 15.0 15.1 McMillan DE, Hardwick WC, Li M, Owens SM. Pharmacokinetic antagonism of (+)-methamphetamine discrimination by a low-affinity monoclonal anti-methamphetamine antibody. Behav Pharmacol. 2002 Sep;13(5-6):465-73. PMID:12394422
- ↑ 16.0 16.1 Owens SM, Zorbas M, Lattin DL, Gunnell M, Polk M. Antibodies against arylcyclohexylamines and their similarities in binding specificity with the phencyclidine receptor. J Pharmacol Exp Ther. 1988 Aug;246(2):472-8. PMID:2457075
- ↑ 17.0 17.1 Laurenzana EM, Stevens MW, Frank JC, Hambuchen MD, Hendrickson HP, White SJ, Williams DK, Owens SM, Gentry WB. Pharmacological effects of two anti-methamphetamine monoclonal antibodies. Supporting data for lead candidate selection for clinical development. Hum Vaccin Immunother. 2014;10(9):2638-47. doi: 10.4161/hv.29707. Epub 2014 Nov, 1. PMID:25483484 doi:http://dx.doi.org/10.4161/hv.29707
- ↑ 18.00 18.01 18.02 18.03 18.04 18.05 18.06 18.07 18.08 18.09 18.10 18.11 18.12 Stevens MW, Henry RL, Owens SM, Schutz R, Gentry WB. First human study of a chimeric anti-methamphetamine monoclonal antibody in healthy volunteers. MAbs. 2014;6(6):1649-56. doi: 10.4161/19420862.2014.976431. PMID:25484042 doi:http://dx.doi.org/10.4161/19420862.2014.976431
- ↑ 19.00 19.01 19.02 19.03 19.04 19.05 19.06 19.07 19.08 19.09 19.10 19.11 19.12 19.13 19.14 19.15 19.16 19.17 19.18 19.19 19.20 19.21 19.22 19.23 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 Apr;325(1):124-33. doi: 10.1124/jpet.107.134395. Epub , 2008 Jan 11. PMID:18192498 doi:http://dx.doi.org/10.1124/jpet.107.134395
- ↑ 20.0 20.1 Hendrickson HP, Milesi-Halle A, Laurenzana EM, Owens SM. Development of a liquid chromatography-tandem mass spectrometric method for the determination of methamphetamine and amphetamine using small volumes of rat serum. J Chromatogr B Analyt Technol Biomed Life Sci. 2004 Jul 5;806(2):81-7. doi:, 10.1016/j.jchromb.2004.03.038. PMID:15171915 doi:http://dx.doi.org/10.1016/j.jchromb.2004.03.038
- ↑ 21.0 21.1 Proksch JW, Gentry WB, Owens SM. Pharmacokinetic mechanisms for obtaining high renal coelimination of phencyclidine and a monoclonal antiphencyclidine antigen-binding fragment of immunoglobulin G in the rat. J Pharmacol Exp Ther. 1998 Nov;287(2):616-24. PMID:9808688
- ↑ 22.0 22.1 22.2 22.3 Celikel R, Peterson EC, Owens SM, Varughese KI. Crystal structures of a therapeutic single chain antibody in complex with two drugs of abuse - methamphetamine and 3,4 methylenedioxy-N-methylamphetamine. Protein Sci. 2009 Sep 16. PMID:19760665 doi:10.1002/pro.244
- ↑ Burley SK, Petsko GA. Aromatic-aromatic interaction: a mechanism of protein structure stabilization. Science. 1985 Jul 5;229(4708):23-8. PMID:3892686
- ↑ Kosten T, Owens SM. Immunotherapy for the treatment of drug abuse. Pharmacol Ther. 2005 Oct;108(1):76-85. doi: 10.1016/j.pharmthera.2005.06.009. PMID:16023218 doi:http://dx.doi.org/10.1016/j.pharmthera.2005.06.009
- ↑ 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 Nov 22;8(4):E709-17. doi: 10.1208/aapsj080480. PMID:17233534 doi:http://dx.doi.org/10.1208/aapsj080480
- ↑ Gentry WB, Ruedi-Bettschen D, Owens SM. Development of active and passive human vaccines to treat methamphetamine addiction. Hum Vaccin. 2009 Apr;5(4):206-13. Epub 2009 Apr 20. PMID:19276653
- ↑ Nanaware-Kharade N, Gonzalez GA 3rd, Lay JO Jr, Hendrickson HP, Peterson EC. Therapeutic anti-methamphetamine antibody fragment-nanoparticle conjugates: synthesis and in vitro characterization. Bioconjug Chem. 2012 Sep 19;23(9):1864-72. doi: 10.1021/bc300204n. Epub 2012 Aug , 28. PMID:22873701 doi:http://dx.doi.org/10.1021/bc300204n
- ↑ Peterson EC, Celikel R, Gokulan K, Varughese KI. Structural characterization of a therapeutic anti-methamphetamine antibody fragment: oligomerization and binding of active metabolites. PLoS One. 2013 Dec 5;8(12):e82690. doi: 10.1371/journal.pone.0082690. eCollection, 2013. PMID:24349338 doi:http://dx.doi.org/10.1371/journal.pone.0082690
- ↑ Thakkar S, Nanaware-Kharade N, Celikel R, Peterson EC, Varughese KI. Affinity improvement of a therapeutic antibody to methamphetamine and amphetamine through structure-based antibody engineering. Sci Rep. 2014 Jan 14;4:3673. doi: 10.1038/srep03673. PMID:24419156 doi:http://dx.doi.org/10.1038/srep03673
- ↑ Hay CE, Gonzalez GA 3rd, Ewing LE, Reichard EE, Hambuchen MD, Nanaware-Kharade N, Alam S, Bolden CT, Owens SM, Margaritis P, Peterson EC. Development and testing of AAV-delivered single-chain variable fragments for the treatment of methamphetamine abuse. PLoS One. 2018 Jun 29;13(6):e0200060. doi: 10.1371/journal.pone.0200060., eCollection 2018. PMID:29958300 doi:http://dx.doi.org/10.1371/journal.pone.0200060
- ↑ Vidarsson G, Dekkers G, Rispens T. IgG subclasses and allotypes: from structure to effector functions. Front Immunol. 2014 Oct 20;5:520. doi: 10.3389/fimmu.2014.00520. eCollection, 2014. PMID:25368619 doi:http://dx.doi.org/10.3389/fimmu.2014.00520